1
|
Hao MJ, Cheng ZY, Gao Y, Xin L, Yu CT, Wang TL, Li ZS, Wang LW. Liquid biopsy of oesophageal squamous cell carcinoma: implications in diagnosis, prognosis, and treatment monitoring. Scand J Gastroenterol 2024; 59:698-709. [PMID: 38466190 DOI: 10.1080/00365521.2024.2310167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/20/2024] [Indexed: 03/12/2024]
Abstract
Oesophageal squamous cell carcinoma (ESCC) is a common malignant tumour of the gastrointestinal tract. Early detection and access to appropriate treatment are crucial for the long-term survival of patients. However, limited diagnostic and monitoring methods are available for identifying early stage ESCC. Endoscopic screening and surgical resection are commonly used to diagnose and treat early ESCC. However, these methods have disadvantages, such as high recurrence, lethality, and mortality rates. Therefore, methods to improve early diagnosis of ESCC and reduce its mortality rate are urgently required. In 1961, Gary et al. proposed a novel liquid biopsy approach for clinical diagnosis. This involved examining exosomes, circulating tumour cells, circulating free DNA, and circulating free RNA in body fluids. The ability of liquid biopsy to obtain samples repeatedly, wide detection range, and fast detection speed make it a feasible option for non-invasive tumour detection. In clinical practice, liquid biopsy technology has gained popularity for early screening, diagnosis, treatment efficacy monitoring, and prognosis assessment. Thus, this is a highly promising examination method. However, there have been no comprehensive reviews on the four factors of liquid biopsy in the context of ESCC. This review aimed to analyse the progress of liquid biopsy research for ESCC, including its classification, components, and potential future applications.
Collapse
Affiliation(s)
- Mei-Juan Hao
- University of Shanghai for Science and Technology, Shanghai, China
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Anaesthesia and Surgery, Guiyang Fourth People's Hospital, Guiyang, China
| | - Zhi-Yuan Cheng
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Gao
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Lei Xin
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chu-Ting Yu
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ting-Lu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Luo-Wei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
2
|
Zhang C, Guo Z, Jing Z. Prediction of Response to Chemoradiotherapy by Dynamic Changes of Circulating Exosome Levels in Patients with Esophageal Squamous Cell Carcinoma. Int J Nanomedicine 2024; 19:1351-1362. [PMID: 38352821 PMCID: PMC10863473 DOI: 10.2147/ijn.s440684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/01/2024] [Indexed: 02/16/2024] Open
Abstract
Background The exosomes-based liquid biopsy represents a prospective biomarker for tumor screening, prognosis prediction, and tumor regression. This study aimed to isolate circulating exosomes (CEs) from plasma of the esophageal squamous cell carcinoma (ESCC) patients who received chemoradiotherapy through exosome detection method via the ultrafast-isolation system (EXODUS) and investigated the association between the dynamic changes of CE levels and therapeutic effect. Methods We isolated and quantitatively analyzed CEs from plasma of locally advanced ESCC patients received chemoradiotherapy at 2 time points: baseline (pre-chemoradiotherapy) and 2 months after the chemoradiotherapy (post-chemoradiotherapy). We isolated exosomes from plasma by EXODUS platform and confirmed them through nanoparticle tracking analysis (NTA), transmission electron microscope (TEM), and Western blot. The associations of CE level with clinicopathological characteristics, tumor regression, and progression-free survival (PFS) were analyzed. Results The average diameter of CEs was 107.4±14.3 nm at baseline and 101.7±17.1 nm at post-chemoradiotherapy. The mean exosome concentration significantly decreased after chemoradiotherapy (7.3×1011 particles/mL vs 5.4×1011 particles/mL, P < 0.001). The patients with stage III-IVA and tumor length ≥5cm had obviously higher baseline CE levels. Dynamic changes in CE levels were successfully applied for evaluation of chemoradiotherapy response and PFS. Furthermore, through multivariate Cox regression analysis, it was revealed that dynamic changes of CE levels were an independent predictor of PFS in locally advanced ESCC patients who received chemoradiotherapy. Conclusion Here, we demonstrated EXODUS platform isolated and enriched CEs from plasma of ESCC patients with high-purity and high-yield. The EXODUS platform can facilitate liquid biopsy based on exosomes translation to the clinic. Baseline CE levels can reflect ESCC tumor burden. The dynamic changes of CE levels during chemoradiotherapy allow the prediction of treatment effect and PFS of ESCC patients, requiring further investigations in larger patient cohorts.
Collapse
Affiliation(s)
- Chuanfeng Zhang
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, People’s Republic of China
| | - Zhen Guo
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, People’s Republic of China
| | - Zhao Jing
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, People’s Republic of China
| |
Collapse
|
3
|
Zhang WT, Wang YJ, Zhang GX, Zhang YH, Gao SS. Diagnostic value of circulating microRNAs for esophageal cancer: a meta-analysis based on Asian data. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2023; 115:504-514. [PMID: 35040334 DOI: 10.17235/reed.2022.8348/2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND OBJECTIVE esophageal cancer (EC) is one of the most common gastrointestinal malignant diseases. We conducted a comprehensive meta-analysis to explore the clinical applicability of circulating microRNA for the diagnosis of EC. METHODS as of September 10, 2021, a comprehensive literature search was conducted on PubMed, Embase, Web of Science, Cochrane Library, Wanfang Database, and China National Knowledge Infrastructure (CNKI) to identify eligible studies. The sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and area under the curve (AUC) were pooled to evaluate the test performance. The potential sources of heterogeneity were analyzed by subgroup analysis. Deeks' funnel plot was used to assess publication bias. RESULTS 85 studies from 50 articles were included in the current meta-analysis. The overall pooled sensitivity was 0.82 (95 % CI, 0.79-0.84), specificity was 0.84 (95 % CI, 0.81-0.86), PLR was 4.9 (95 % CI, 4.2-5.9), NLR was 0.22 (95 % CI, 0.19-0.25), DOR was 22 (95 % CI, 17-29) and AUC was 0.89 (95 % CI, 0.86-0.92), respectively. Subgroup analysis suggested that miRNA clusters with a large sample size showed better diagnostic accuracy. Publication bias was not found. CONCLUSIONS circulating miRNAs can be used as a potential non-invasive biomarker for the diagnosis of EC in Asian populations.
Collapse
|
4
|
Vosough P, Khatami SH, Hashemloo A, Tajbakhsh A, Karimi-Fard F, Taghvimi S, Taheri-Anganeh M, Soltani Fard E, Savardashtaki A, Movahedpour A. Exosomal lncRNAs in gastrointestinal cancer. Clin Chim Acta 2023; 540:117216. [PMID: 36592922 DOI: 10.1016/j.cca.2022.117216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
Gastrointestinal cancer (GIC) remains a leading cause of morbidity and mortality worldwide. Unfortunately, these cancers are diagnosed in advanced metastatic stages due to lack of reliable biomarkers that are sufficiently specific and sensitive in early disease. There has been growing evidence that circulating exosomes can be used to diagnose cancer non-invasively with limited risks and side effects. Furthermore, exosomal long non-coding RNAs (lncRNAs) are emerging as a new class of promising biomarkers in cancer. This review provides an overview of the extraction and detection of exosomal lncRNAs with a focus on their potential role in GIC.
Collapse
Affiliation(s)
- Parisa Vosough
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Sina Taghvimi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Elahe Soltani Fard
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | |
Collapse
|
5
|
Extracellular Vesicles Are Important Mediators That Regulate Tumor Lymph Node Metastasis via the Immune System. Int J Mol Sci 2023; 24:ijms24021362. [PMID: 36674900 PMCID: PMC9865533 DOI: 10.3390/ijms24021362] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/27/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Extracellular vesicles (EVs) are particles with a lipid bilayer structure, and they are secreted by various cells in the body. EVs interact with and modulate the biological functions of recipient cells by transporting their cargoes, such as nucleic acids and proteins. EVs influence various biological phenomena, including disease progression. They also participate in tumor progression by stimulating a variety of signaling pathways and regulating immune system activation. EVs induce immune tolerance by suppressing CD8+ T-cell activation or polarizing macrophages toward the M2 phenotype, which results in tumor cell proliferation, migration, invasion, and metastasis. Moreover, immune checkpoint molecules are also expressed on the surface of EVs that are secreted by tumors that express these molecules, allowing tumor cells to not only evade immune cell attack but also acquire resistance to immune checkpoint inhibitors. During tumor metastasis, EVs contribute to microenvironmental changes in distant organs before metastatic lesions appear; thus, EVs establish a premetastatic niche. In particular, lymph nodes are adjacent organs that are connected to tumor lesions via lymph vessels, so that tumor cells metastasize to draining lymph nodes at first, such as sentinel lymph nodes. When EVs influence the microenvironment of lymph nodes, which are secondary lymphoid tissues, the immune response against tumor cells is weakened; subsequently, tumor cells spread throughout the body. In this review, we will discuss the association between EVs and tumor progression via the immune system as well as the clinical application of EVs as biomarkers and therapeutic agents.
Collapse
|
6
|
Jiawei S, Zhi C, Kewei T, Xiaoping L. Magnetic bead-based adsorption strategy for exosome isolation. Front Bioeng Biotechnol 2022; 10:942077. [PMID: 36051582 PMCID: PMC9424818 DOI: 10.3389/fbioe.2022.942077] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/30/2022] [Indexed: 12/04/2022] Open
Abstract
Exosomes, one type of extracellular vesicle (EV) secreted by cells, participate in intercellular communication and other biological processes as carriers of lipids, functional proteins, mRNAs, miRNAs, lncRNAs, and DNA fragments. Their presence in biofluids makes them attractive candidates as innovative clinical diagnostic tools. However, the conventional isolation and analysis of high-purity exosomes in clinical application is challenging, with traditional methods facing a number of shortcomings, including low yield or purity, long periods of processing, high cost, and difficulties in standardization. In this study, we provide an overview of commonly used exosome isolation approaches with a focus on magnetic bead-based capture, an ideal methodology with high purity and integrality of exosomes. The current challenges on exosome isolation methods are also described to highlight areas for future research and development.
Collapse
Affiliation(s)
- Sun Jiawei
- Shulan International Medical College, Zhejiang Shuren College, Hangzhou, China
| | - Chen Zhi
- Zhejiang University School of Medicine, Hangzhou, China
| | - Tian Kewei
- Shulan International Medical College, Zhejiang Shuren College, Hangzhou, China
| | - Li Xiaoping
- Shulan International Medical College, Zhejiang Shuren College, Hangzhou, China,*Correspondence: Li Xiaoping,
| |
Collapse
|
7
|
Meng L, Song K, Li S, Kang Y. Exosomes: Small Vesicles with Important Roles in the Development, Metastasis and Treatment of Breast Cancer. MEMBRANES 2022; 12:membranes12080775. [PMID: 36005690 PMCID: PMC9414313 DOI: 10.3390/membranes12080775] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 05/12/2023]
Abstract
Breast cancer (BC) has now overtaken lung cancer as the most common cancer, while no biopredictive marker isolated from biological fluids has yet emerged clinically. After traditional chemotherapy, with the huge side effects brought by drugs, patients also suffer from the double affliction of drugs to the body while fighting cancer, and they often quickly develop drug resistance after the drug, leading to a poor prognosis. And the treatment of some breast cancer subtypes, such as triple negative breast cancer (TNBC), is even more difficult. Exosomes (Exos), which are naturally occurring extracellular vesicles (EVs) with nanoscale acellular structures ranging in diameter from 40 to 160 nm, can be isolated from various biological fluids and have been widely studied because they are derived from the cell membrane, have extremely small diameter, and are widely involved in various biological activities of the body. It can be used directly or modified to make derivatives or to make some analogs for the treatment of breast cancer. This review will focus on the involvement of exosomes in breast cancer initiation, progression, invasion as well as metastasis and the therapeutic role of exosomes in breast cancer.
Collapse
Affiliation(s)
- Ling’ao Meng
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
- Correspondence: (S.L.); (Y.K.)
| | - Yue Kang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
- Correspondence: (S.L.); (Y.K.)
| |
Collapse
|
8
|
Yang L, Patel KD, Rathnam C, Thangam R, Hou Y, Kang H, Lee KB. Harnessing the Therapeutic Potential of Extracellular Vesicles for Biomedical Applications Using Multifunctional Magnetic Nanomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104783. [PMID: 35132796 PMCID: PMC9344859 DOI: 10.1002/smll.202104783] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/12/2022] [Indexed: 04/14/2023]
Abstract
Extracellular vesicles (e.g., exosomes) carrying various biomolecules (e.g., proteins, lipids, and nucleic acids) have rapidly emerged as promising platforms for many biomedical applications. Despite their enormous potential, their heterogeneity in surfaces and sizes, the high complexity of cargo biomolecules, and the inefficient uptake by recipient cells remain critical barriers for their theranostic applications. To address these critical issues, multifunctional nanomaterials, such as magnetic nanomaterials, with their tunable physical, chemical, and biological properties, may play crucial roles in next-generation extracellular vesicles (EV)-based disease diagnosis, drug delivery, tissue engineering, and regenerative medicine. As such, one aims to provide cutting-edge knowledge pertaining to magnetic nanomaterials-facilitated isolation, detection, and delivery of extracellular vesicles and their associated biomolecules. By engaging the fields of extracellular vesicles and magnetic nanomaterials, it is envisioned that their properties can be effectively combined for optimal outcomes in biomedical applications.
Collapse
Affiliation(s)
- Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123 Bevier Road, Piscataway, NJ 08854, USA
| | - Kapil D. Patel
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Christopher Rathnam
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123 Bevier Road, Piscataway, NJ 08854, USA
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123 Bevier Road, Piscataway, NJ 08854, USA
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers-the State University of New Jersey, 123 Bevier Road, Pis cataway, NJ 08854, USA
| |
Collapse
|
9
|
Translating cancer exosomes detection into the color change of phenol red based on target-responsive DNA microcapsules. Anal Chim Acta 2022; 1192:339357. [PMID: 35057959 DOI: 10.1016/j.aca.2021.339357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/29/2021] [Accepted: 12/05/2021] [Indexed: 11/15/2022]
Abstract
Emerging evidence indicates that exosomes can be used as a potential biomarker for monitoring diseases, including cancer. However, enhancing the sensing performance in terms of convenience and sensitivity remains an urgent demand for exosomes detection. In this study, a pH-sensitive colorimetric biosensing strategy was developed for exosomes detection by integrating stimuli-responsive DNA microcapsules and acetylcholinesterase to produce acetic acid. The constructed DNA microcapsules consisted of DNA shells crosslinked by anti-CD63 aptamers and loaded with acetylcholinesterase. With exosomes addition, an energetically stabilized aptamer-CD63 compound was produced and microcapsules dissociated due to the reaction of surface protein CD63 of exosomes and aptamer of CD63, resulting in the release of encapsulated AChE. Through a simple centrifugation separation, unreacted DNA microcapsules were removed and the supernatant containing released acetylcholinesterase collected, which was then used for colorimetric exosomes detection through the ability of acetylcholinesterase to hydrolyze acetylcholine to release acetic acid. The resulting decreased solution pH was detected with phenol red indicator, with the sharp color transition conveniently by naked eye. Exosomes quantification was also achieved using the solution's absorption intensity ratio of 558 vs. 432 nm. The linear range was from 2.0 × 103 to 5.0 × 105 particles/μL, and the limit of detection and limit of quantification were 1.2 × 103 particles/μL and 2.2 × 103 particles/μL, respectively. In addition, this proposed strategy for exosomes detection showed a relative standard deviation of 3.1% and high recovery efficiency (>94%), exhibiting a bright application future in exsomes analysis.
Collapse
|
10
|
Zhu Q, Huang L, Yang Q, Ao Z, Yang R, Krzesniak J, Lou D, Hu L, Dai X, Guo F, Liu F. Metabolomic analysis of exosomal-markers in esophageal squamous cell carcinoma. NANOSCALE 2021; 13:16457-16464. [PMID: 34648610 DOI: 10.1039/d1nr04015d] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a worldwide malignancy with high mortality rates and poor prognosis due to the lack of effective biomarkers for early detection. Exosomes have been extensively explored as attractive biomarkers for cancer diagnosis and treatment. However, little is known about exosome metabolomics and their roles in ESCC. Here, we performed a targeted metabolomic analysis of plasma exosomes and identified 196 metabolites, mainly including lipid fatty acids, benzene, amino acids, organic acids, carbohydrates and fatty acyls. We systematically compared metabolome patterns of exosomes via machine learning from patients with recrudescence and patients without recrudescence and demonstrated a marker set consisting of 3'-UMP, palmitoleic acid, palmitaldehyde, and isobutyl decanoate for predicting ESCC recurrence with an AUC of 98%. These metabolome signatures of exosomes retained a high absolute fold change value at all ESCC stages and were very likely associated with cancer metabolism, which could be potentially applied as novel biomarkers for diagnosis and prognosis of ESCC.
Collapse
Affiliation(s)
- Qingfu Zhu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Liu Huang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| | - Zheng Ao
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Rui Yang
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jonathan Krzesniak
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Doudou Lou
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Liang Hu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xiaodan Dai
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN 47405, USA.
| | - Fei Liu
- Eye Hospital, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| |
Collapse
|
11
|
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
|
12
|
Jing Z, Chen K, Gong L. The Significance of Exosomes in Pathogenesis, Diagnosis, and Treatment of Esophageal Cancer. Int J Nanomedicine 2021; 16:6115-6127. [PMID: 34511909 PMCID: PMC8423492 DOI: 10.2147/ijn.s321555] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/16/2021] [Indexed: 12/21/2022] Open
Abstract
Esophageal cancer is one of the most common malignancy in China with high mortality. Understanding pathogenesis and identifying early diagnosis biomarkers can significantly improve the prognosis of patients with esophageal cancer. Exosomes are small vesicular structures containing a variety of components (including DNA, RNA, and proteins) mediating cell-to-cell material exchange and signal communication. Growing evidences have shown that exosomes and its components are involved in growth, metastasis and angiogenesis in cancer, and could also be used as diagnostic and prognostic markers. In this review, we summarized recent progress to elucidate the significance of exosomes in the esophageal cancer progression, microenvironment remodeling, therapeutic resistance, and immunosuppression. We also discuss the utility of exosomes as diagnostic and prognostic biomarkers and therapeutic tool in esophageal cancer.
Collapse
Affiliation(s)
- Zhao Jing
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Kai Chen
- Department of Cardiovascular and Thoracic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Ling Gong
- Department of Infectious Disease (Liver Diseases), The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
13
|
Extracellular Vesicles as a Novel Liquid Biopsy-Based Diagnosis for the Central Nervous System, Head and Neck, Lung, and Gastrointestinal Cancers: Current and Future Perspectives. Cancers (Basel) 2021; 13:cancers13112792. [PMID: 34205183 PMCID: PMC8200014 DOI: 10.3390/cancers13112792] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary To improve clinical outcomes, early diagnosis is mandatory in cancer patients. Several diagnostic approaches have been proposed, however, the main drawback relies on the invasive procedures required. Extracellular vesicles (EVs) are bilayer lipid membrane structures released by almost all cells and transferred to remote sites via the bloodstream. The observation that their cargo reflects the cell of origin has opened a new frontier for non-invasive biomarker discovery in oncology. Moreover, since EVs can be recovered from different body fluids, their impact as a Correctdiagnostic tool has gained particular interest. Hence, in the last decade, several studies using different biological fluids have been performed, showing the valuable contributions of EVs as tumour biomarkers, and their improved diagnostic power when combined with currently available tumour markers. In this review, the most relevant data on the diagnostic relevance of EVs, alone or in combination with the well-established tumour markers, are discussed. Abstract Early diagnosis, along with innovative treatment options, are crucial to increase the overall survival of cancer patients. In the last decade, extracellular vesicles (EVs) have gained great interest in biomarker discovery. EVs are bilayer lipid membrane limited structures, released by almost all cell types, including cancer cells. The EV cargo, which consists of RNAs, proteins, DNA, and lipids, directly mirrors the cells of origin. EVs can be recovered from several body fluids, including blood, cerebral spinal fluid (CSF), saliva, and Broncho-Alveolar Lavage Fluid (BALF), by non-invasive or minimally invasive approaches, and are therefore proposed as feasible cancer diagnostic tools. In this review, methodologies for EV isolation and characterization and their impact as diagnostics for the central nervous system, head and neck, lung, and gastrointestinal cancers are outlined. For each of these tumours, recent data on the potential clinical applications of the EV’s unique cargo, alone or in combination with currently available tumour biomarkers, have been deeply discussed.
Collapse
|
14
|
Liu T, Du LT, Wang YS, Gao SY, Li J, Li PL, Sun ZW, Binang H, Wang CX. Development of a Novel Serum Exosomal MicroRNA Nomogram for the Preoperative Prediction of Lymph Node Metastasis in Esophageal Squamous Cell Carcinoma. Front Oncol 2020; 10:573501. [PMID: 33123480 PMCID: PMC7573187 DOI: 10.3389/fonc.2020.573501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Preoperative prediction of lymph node (LN) metastasis is accepted as a crucial independent risk factor for treatment decision-making for esophageal squamous cell carcinoma (ESCC) patients. Our study aimed to establish a non-invasive nomogram to identify LN metastasis preoperatively in ESCC patients. Construction of the nomogram involved three sequential phases with independent patient cohorts. In the discovery phase (N = 20), LN metastasis-associated microRNAs (miRNAs) were selected from next-generation sequencing (NGS) assay of human ESCC serum exosome samples. In the training phase (N = 178), a nomogram that incorporated exosomal miRNA model and clinicopathologic was developed by multivariate logistic regression analysis to preoperatively predict LN status. In the validation phase (n = 188), we validated the predicted nomogram's calibration, discrimination, and clinical usefulness. Four differently expressed miRNAs (chr 8-23234-3p, chr 1-17695-5p, chr 8-2743-5p, and miR-432-5p) were tested and selected in the serum exosome samples from ESCC patients who have or do not have LN metastasis. Subsequently, an optimized four-exosomal miRNA model was constructed and validated in the clinical samples, which could effectively identify ESCC patients with LN metastasis, and was significantly superior to preoperative computed tomography (CT) report. In addition, a clinical nomogram consisting of the four-exosomal miRNA model and CT report was established in training cohort, which showed high predictive value in both training and validation cohorts [area under the receiver operating characteristic curve (AUC): 0.880 and 0.869, respectively]. The Hosmer–Lemeshow test and decision curve analysis implied the nomogram's clinical applicability. Our novel non-invasive nomogram is a robust prediction tool with promising clinical potential for preoperative LN metastasis prediction of ESCC patients, especially in T1 stage.
Collapse
Affiliation(s)
- Tong Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lu-Tao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Yun-Shan Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Shan-Yu Gao
- Department of Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Pei-Long Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Zhao-Wei Sun
- Department of Surgery, The Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Helen Binang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuan-Xin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| |
Collapse
|
15
|
Słomka A, Mocan T, Wang B, Nenu I, Urban SK, Gonzalez-Carmona MA, Schmidt-Wolf IGH, Lukacs-Kornek V, Strassburg CP, Spârchez Z, Kornek M. EVs as Potential New Therapeutic Tool/Target in Gastrointestinal Cancer and HCC. Cancers (Basel) 2020; 12:E3019. [PMID: 33080904 PMCID: PMC7603109 DOI: 10.3390/cancers12103019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/04/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
For more than a decade, extracellular vesicles (EVs) have been in focus of science. Once thought to be an efficient way to eliminate undesirable cell content, EVs are now well-accepted as being an important alternative to cytokines and chemokines in cell-to-cell communication route. With their cargos, mainly consisting of functional proteins, lipids and nucleic acids, they can activate signalling cascades and thus change the phenotype of recipient cells at local and systemic levels. Their substantial role as modulators of various physiological and pathological processes is acknowledged. Importantly, more and more evidence arises that EVs play a pivotal role in many stages of carcinogenesis. Via EV-mediated communication, tumour cells can manipulate cells from host immune system or from the tumour microenvironment, and, ultimately, they promote tumour progression and modulate host immunity towards tumour's favour. Additionally, the role of EVs in modulating resistance to pharmacological and radiological therapy of many cancer types has become evident lately. Our understanding of EV biology and their role in cancer promotion and drug resistance has evolved considerably in recent years. In this review, we specifically discuss the current knowledge on the association between EVs and gastrointestinal (GI) and liver cancers, including their potential for diagnosis and treatment.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85-067 Bydgoszcz, Poland;
| | - Tudor Mocan
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (T.M.); (I.N.); (Z.S.)
| | - Bingduo Wang
- Department of Internal Medicine I, University Hospital of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany; (B.W.); (S.K.U.); (M.G.-C.); (C.P.S.)
| | - Iuliana Nenu
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (T.M.); (I.N.); (Z.S.)
| | - Sabine K. Urban
- Department of Internal Medicine I, University Hospital of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany; (B.W.); (S.K.U.); (M.G.-C.); (C.P.S.)
| | - Maria A. Gonzalez-Carmona
- Department of Internal Medicine I, University Hospital of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany; (B.W.); (S.K.U.); (M.G.-C.); (C.P.S.)
| | - Ingo G. H. Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany;
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, University Hospital of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany;
| | - Christian P. Strassburg
- Department of Internal Medicine I, University Hospital of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany; (B.W.); (S.K.U.); (M.G.-C.); (C.P.S.)
| | - Zeno Spârchez
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania; (T.M.); (I.N.); (Z.S.)
| | - Miroslaw Kornek
- Department of Internal Medicine I, University Hospital of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany; (B.W.); (S.K.U.); (M.G.-C.); (C.P.S.)
| |
Collapse
|
16
|
Liskova A, Samec M, Koklesova L, Giordano FA, Kubatka P, Golubnitschaja O. Liquid Biopsy is Instrumental for 3PM Dimensional Solutions in Cancer Management. J Clin Med 2020; 9:E2749. [PMID: 32854390 PMCID: PMC7563444 DOI: 10.3390/jcm9092749] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
One in every four deaths is due to cancer in Europe. In view of its increasing incidence, cancer became the leading cause of death and disease burden in Denmark, France, the Netherlands, and the UK. Without essential improvements in cancer prevention, an additional 775,000 cases of annual incidence have been prognosed until 2040. Between 1995 and 2018, the direct costs of cancer doubled from EUR 52 billion to EUR 103 billion in Europe, and per capita health spending on cancer increased by 86% from EUR 105 to EUR 195 in general, whereby Austria, Germany, Switzerland, Benelux, and France spend the most on cancer care compared to other European countries. In view of the consequent severe socio-economic burden on society, the paradigm change from a reactive to a predictive, preventive, and personalized medical approach in the overall cancer management is essential. Concepts of predictive, preventive, and personalized medicine (3PM) demonstrate a great potential to revise the above presented trends and to implement cost-effective healthcare that benefits the patient and society as a whole. At any stage, application of early and predictive diagnostics, targeted prevention, and personalization of medical services are basic pillars making 3PM particularly attractive for the patients as well as ethical and cost-effective healthcare. Optimal 3PM approach requires novel instruments such as well-designed liquid biopsy application. This review article highlights current achievements and details liquid biopsy approaches specifically in cancer management. 3PM-relevant expert recommendations are provided.
Collapse
Affiliation(s)
- Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (A.L.); (M.S.); (L.K.)
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (A.L.); (M.S.); (L.K.)
| | - Lenka Koklesova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia; (A.L.); (M.S.); (L.K.)
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| |
Collapse
|
17
|
Guo D, Yuan J, Xie A, Lin Z, Li X, Chen J. Diagnostic performance of circulating exosomes in human cancer: A meta-analysis. J Clin Lab Anal 2020; 34:e23341. [PMID: 32309888 PMCID: PMC7439344 DOI: 10.1002/jcla.23341] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/29/2020] [Accepted: 03/28/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cancer has become a public health problem with high morbidity and mortality. Recent publications have shown that exosomes can be used as potential diagnostic biomarkers of cancer. However, the diagnostic accuracy and reliability of circulating exosomes remain unclear. The present meta-analysis was conducted to comprehensively summarize the overall diagnostic performance of circulating exosomes for cancer. METHODS Eligible studies published up to June 27, 2019, on PubMed, Embase, and Cochrane Library were selected for the meta-analysis. All statistical analyses were performed by STATA 15.1 statistical software and Meta-DiSc 1.4. Quality Assessment for Studies of Diagnostic Accuracy 2 tool was used to access the quality of included studies. A bivariate mixed-effects model was applied to calculate the diagnostic indexes from included studies. RESULTS A total of 5924 participants comprising 3161 cases and 2763 controls from 42 eligible studies were analyzed. The pooled sensitivity, specificity, positive likelihood ratio, negative likelihood ratio, diagnostic odds ratio, and the area under the curve with 95% confidence intervals (95% CI) were as follows: 0.79 (0.75-0.82), 0.81 (0.78-0.84), 4.1 (3.5-4.8), 0.26 (0.22-0.31), 16 (12-21), and 0.87 (0.84-0.89), respectively. Sensitivity analysis suggested no study exclusively contributed to the heterogeneity, and Deeks' funnel plot asymmetry test indicated no potential publication bias (P = .09). CONCLUSIONS The meta-analysis indicated that circulating exosomes could serve as effective and minimally invasive biomarkers for diagnosis of cancer, especially in patients with hepatocellular carcinoma or ovarian cancer, serum-based samples and exosomal proteins.
Collapse
Affiliation(s)
- Dongming Guo
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
- Shantou University Medical CollegeShantouChina
| | - Jinpeng Yuan
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Aosi Xie
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Zeyin Lin
- Department of UltrasoundThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Xinxin Li
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Juntian Chen
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Shantou University Medical CollegeShantouChina
| |
Collapse
|
18
|
Five-lncRNA signature in plasma exosomes serves as diagnostic biomarker for esophageal squamous cell carcinoma. Aging (Albany NY) 2020; 12:15002-15010. [PMID: 32597791 PMCID: PMC7425435 DOI: 10.18632/aging.103559] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
Changes in expression of long non-coding RNAs (lncRNAs) in plasma exosomes can be useful for diagnosis of cancer patients. Here, we conducted a four-stage study to identify plasma exosome lncRNAs with diagnostic potential in esophageal squamous cell carcinoma (ESCC). First, plasma exosome lncRNA expression profiles were examined in ESCC patients, esophagitis patients, and healthy controls using RNA sequencing. Differentially expressed plasma exosome lncRNAs from the lncRNA expression profile were then evaluated by qRT-PCR in a large cohort of ESCC patients, esophagitis patients, and healthy controls. Expression levels of the lncRNAs NR_039819, NR_036133, NR_003353, ENST00000442416.1, and ENST00000416100.1 were significantly higher in exosomes from ESCC patients than non-cancer controls. We also confirmed that levels of these five plasma exosome lncRNAs decreased markedly in ESCC patients after surgery. Our results suggest that these five exosome lncRNAs may serve as highly effective, noninvasive biomarkers for ESCC diagnosis.
Collapse
|
19
|
Makler A, Asghar W. Exosomal biomarkers for cancer diagnosis and patient monitoring. Expert Rev Mol Diagn 2020; 20:387-400. [PMID: 32067543 PMCID: PMC7071954 DOI: 10.1080/14737159.2020.1731308] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/14/2020] [Indexed: 02/06/2023]
Abstract
Introduction: In recent years, extensive research has been conducted on using exosomes as biomarkers for cancer detection. Exosomes are 40-150 nm-sized extracellular vesicles released by all cell types, including tumor cells. Exosomes are stable in body fluids due to their lipid bilayer member and often contain DNA, RNA, and proteins. These exosomes can be harvested from blood, plasma, serum, urine, or saliva and analyzed for tumor-relevant mutations. Thus, exosomes provide an alternative to current methods of tumor detection.Areas covered: This review discusses the use of exosomal diagnostics in various tumor types as well as their examination in various clinical trials. The authors also discuss the limitations of exosome-based diagnostics in the clinical setting and provide examples of several studies in which the development and usage of microfluidic chips and nano-sensing devices have been utilized to address these obstacles.Expert commentary: In recent years, exosomes and their contents have exhibited potential as novel tumor detection markers despite the labor involved in their harvest and isolation. Despite this, much work is being done to optimize exosome capture and analysis. Thus, their roles as biomarkers in the clinical setting appear promising.
Collapse
Affiliation(s)
- Amy Makler
- Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431
| | - Waseem Asghar
- Asghar-Lab, Micro and Nanotechnology in Medicine, College of Engineering and Computer Science, Boca Raton, FL 33431
- Department of Computer & Electrical Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431
- Department of Biological Sciences (courtesy appointment), Florida Atlantic University, Boca Raton, FL 33431
| |
Collapse
|
20
|
Bernardi S, Foroni C, Zanaglio C, Re F, Polverelli N, Turra A, Morello E, Farina M, Cattina F, Gandolfi L, Zollner T, Buttini EA, Malagola M, Russo D. Feasibility of tumor‑derived exosome enrichment in the onco‑hematology leukemic model of chronic myeloid leukemia. Int J Mol Med 2019; 44:2133-2144. [PMID: 31638195 PMCID: PMC6844640 DOI: 10.3892/ijmm.2019.4372] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/12/2019] [Indexed: 01/18/2023] Open
Abstract
Due to the discovery of their role in intra-cellular communications, exosomes, which carry information specific to the cell of origin, have garnered considerable attention in cancer research. Moreover, there is evidence to suggest the possibility of isolating different exosome sub-populations based on target antigens at the cell surface. Philadelphia chromosome-positive (Ph+) chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasia characterized by the break-point cluster region-proto-oncogene 1 tyrosine-protein kinase (BCR-ABL1) fusion-gene, derived from the t (9;22) translocation. Tyrosine kinase inhibitors (TKIs) target BCR-ABL1 protein and induce major or deep molecular responses in the majority of patients. Despite the fact that several studies have demonstrated the persistence of leukemic cells in the bone marrow niche, even following treatment, TKIs prolong patient survival time and facilitate treatment-free remission. These characteristics render CML a plausible model for investigating the feasibility of tumor-derived exosome fraction enrichment. In the present study, patients in the chronic phase (CP) of CML were treated with TKIs, and the quantification of the BCR-ABL1 exosomal transcript was performed using digital PCR (dPCR). The possibility of tumor-derived exosomes enrichment was confirmed, and for the first time, to the best of our knowledge, the detection of the BCR-ABL1 transcript highlighted the presence of active leukemic cells in patients with CP-CML. According to these findings, tumor-derived exosomes may be considered a novel tool for the identification of active leukemic cells, and for the assessment of innovative monitoring focused on the biological functions of exosomes in CML.
Collapse
Affiliation(s)
- Simona Bernardi
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Chiara Foroni
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Camilla Zanaglio
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Federica Re
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Nicola Polverelli
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Alessandro Turra
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Enrico Morello
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Mirko Farina
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Federica Cattina
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Lisa Gandolfi
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Tatiana Zollner
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Eugenia Accorsi Buttini
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Michele Malagola
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Domenico Russo
- Chair of Hematology, Unit of Blood Diseases and Stem Cell Transplantation, Department of Clinical and Experimental Sciences, University of Brescia, Italy
| |
Collapse
|
21
|
Zhao A, Guo L, Xu J, Zheng L, Guo Z, Ling Z, Wang L, Mao W. Identification and validation of circulating exosomes-based liquid biopsy for esophageal cancer. Cancer Med 2019; 8:3566-3574. [PMID: 31099496 PMCID: PMC6601589 DOI: 10.1002/cam4.2224] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/12/2019] [Accepted: 04/12/2019] [Indexed: 01/14/2023] Open
Abstract
Background Early detection of esophageal squamous cell carcinoma (ESCC) recurrence is a key element for follow‐up care and surveillance. The aim of this study is to detect the level of circulating exosomes (CEs) in ESCC patient and clarify its clinical significance. Methods In this study, 200 serum samples of ESCC patients were obtained from the Zhejiang Cancer Hospital Biospecimen Repository. Total CEs were purified by selectively capturing epithelial cell adhesion molecule positive exosomes, using magnetic‐bead technique. enzyme‐linked immunosorbent assay (ELISA) was performed to measure the concentration level of CEs. The oncogenic potential of CEs was analyzed in vitro. Results Serum concentration of CEs was significantly higher in ESCC patients than in healthy controls (P < 0.01). Receiver‐operating characteristic curve analysis demonstrated that CEs concentration could distinguish patients with ESCC from healthy individuals with a sensitivity of 75% and a specificity of 85%. Kaplan‐Meier analysis demonstrated that the increased CEs concentration was associated with poor overall survival (P = 0.01) and progression free survival (P = 0.03) in ESCC patients. Multivariate cox regression analysis revealed that CEs concentration was an independent prognostic marker for overall survival in ESCC patients (P < 0.01). Results from transwell and wound scratching experiments showed that the CEs could promote cell migration and invasion. Conclusions This study clearly demonstrates that CEs from ESCC patients are stable enough to be measured and their levels in ESCC patients are significantly upregulated. Circulating exosomes could serve as a novel noninvasive biomarker for detection of ESCC. Their involvement in carcinogenesis must be further established.
Collapse
Affiliation(s)
- An Zhao
- Institute of Cancer Research, Zhejiang Cancer Hospital, Hangzhou, China
| | - Liwei Guo
- Institute of Cancer Research, Zhejiang Cancer Hospital, Hangzhou, China.,Zhejiang University School of Medicine, Hangzhou, China
| | - Ji Xu
- Department of Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Lei Zheng
- Institute of Cancer Research, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhenying Guo
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhiqiang Ling
- Institute of Cancer Research, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lidong Wang
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weimin Mao
- Institute of Cancer Research, Zhejiang Cancer Hospital, Hangzhou, China.,Department of Thoracic Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|