1
|
Liu Y, Zhang B, Wu X, Wang F, Yang Z, Li M, Sheng K, Yan Y, Zhu L, Jing H, Wu Y, Hu L, Yu Y, Li C. A facile liquid biopsy assay for highly efficient CTCs capture and reagent-less monitoring of immune checkpoint PD-L1 expression on CTCs with non-small cell lung cancer patients. Biosens Bioelectron 2025; 275:117236. [PMID: 39929085 DOI: 10.1016/j.bios.2025.117236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/23/2025]
Abstract
Programmed cell death 1 ligand 1 (PD-L1) immunotherapy holds a pivotal role in lung cancer treatment. However, current methods for monitoring PD-L1 expression exhibit several limitations, including hysteresis and the invasive nature of tissue sampling. Circulating tumor cells (CTCs), the important biomarkers in liquid biopsy, are minimally invasive and facilitate continuous monitoring. Consequently, integrating CTC counting with PD-L1 expression analysis offers more comprehensive insights for the development of personalized treatment strategies development and efficacy evaluations. In this study, we presented a facile liquid biopsy assay designed for the dynamic monitoring of PD-L1 expression on CTCs captured from non-small cell lung cancer (NSCLC) patients. This assay was achieved by fabricating two high-performance probes: EpCAM and Vimentin dual-aptamer modified nitrogen-doped carbon quantum dots probe (E/V-apt-N-CQDs) and hairpin PD-L1 aptamer coupled with gold nanoparticles (PD-L1-apt-AuNPs). The E/V-apt-N-CQDs probe effectively captured two types of CTC models (H1299 and A549) exhibiting differential PD-L1 expression. Additionally, reagent-less detection of PD-L1 levels on CTCs was achieved using a portable magnetic electrochemical sensor with excellent specificity and sensitivity, which was capable of measuring PD-L1 concentrations as low as 2 ng/mL. Finally, this assay was applied in 41 NSCLC patients to investigate the correlation between CTC numbers or PD-L1 expression and disease progression and immunotherapy efficacy. The results indicated a significant association between elevated CTC counts or reduced PD-L1 levels and clinical progression. Moreover, this liquid assay successfully monitored dynamic changes in CTCs and PD-L1 expression in NSCLC patients receiving immunotherapy, indicating its potential for clinical application.
Collapse
Affiliation(s)
- Yuping Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China
| | - Beibei Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China
| | - Xueyuan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China; Department of Pharmacy, Xuzhou Tongshan District Traditional Chinese Medicine Hospital, Xuzhou, 221116, Jiangsu, PR China
| | - Fan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China
| | - Zhiyi Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China
| | - Mengyi Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China
| | - Kaixuan Sheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China
| | - Yue Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China
| | - Liang Zhu
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu, PR China
| | - Hui Jing
- Department of Respiratory and Critical Care Medicine, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou Central Hospital, Xuzhou, 221009, Jiangsu, PR China
| | - Yanmin Wu
- Department of Respiratory and Critical Care Medicine, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou Central Hospital, Xuzhou, 221009, Jiangsu, PR China
| | - Lili Hu
- Department of Pharmacy, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, PR China.
| |
Collapse
|
2
|
Liu P, Zhang Q, Liu F. Biological roles and clinical applications of EpCAM in HCC. Discov Oncol 2025; 16:319. [PMID: 40087210 PMCID: PMC11909382 DOI: 10.1007/s12672-025-02095-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/07/2025] [Indexed: 03/17/2025] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is an important biomarker in tumors. In hepatocellular carcinoma (HCC), EpCAM + cells exhibit high invasiveness, tumorigenic ability, therapeutic resistance, and self-renewal ability, often identified as liver cancer stem cells (CSCs). Detecting EpCAM + cells in tumor lesions and circulation is valuable for predicting patient prognosis and monitoring therapeutic outcomes, emphasizing its clinical significance. Given its broad expression in HCC, especially in CSCs and circulating tumor cells (CTCs), EpCAM-targeting agents have garnered substantial research interest. However, the role of EpCAM in HCC progression and its regulatory mechanisms remains poorly understood. Furthermore, clinical applications of EpCAM, such as liquid biopsy and targeted therapies, are still controversial. This review summarizes the biological properties of EpCAM + HCC cells, explores the regulatory mechanisms governing EpCAM expression, and discusses its clinical significance of using EpCAM as a prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Peng Liu
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qun Zhang
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fengchao Liu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
3
|
Shahhosseini R, Pakmehr S, Elhami A, Shakir MN, Alzahrani AA, Al-Hamdani MM, Abosoda M, Alsalamy A, Mohammadi-Dehcheshmeh M, Maleki TE, Saffarfar H, Ali-Khiavi P. Current biological implications and clinical relevance of metastatic circulating tumor cells. Clin Exp Med 2024; 25:7. [PMID: 39546080 PMCID: PMC11567993 DOI: 10.1007/s10238-024-01518-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Metastatic disease and cancer recurrence are the primary causes of cancer-related deaths. Circulating tumor cells (CTCs) and disseminated tumor cells (DTCs) are the driving forces behind the spread of cancer cells. The emergence and development of liquid biopsy using rare CTCs as a minimally invasive strategy for early-stage tumor detection and improved tumor management is a promising advancement in recent years. However, before blood sample analysis and clinical translation, precise isolation of CTCs from patients' blood based on their biophysical properties, followed by molecular identification of CTCs using single-cell multi-omics technologies is necessary to understand tumor heterogeneity and provide effective diagnosis and monitoring of cancer progression. Additionally, understanding the origin, morphological variation, and interaction between CTCs and the primary and metastatic tumor niche, as well as and regulatory immune cells, will offer new insights into the development of CTC-based advanced tumor targeting in the future clinical trials.
Collapse
Affiliation(s)
| | - SeyedAbbas Pakmehr
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ahvaz Jundishapur University of Medical Sciences Ahvaz, Ahvaz, Iran
| | - Anis Elhami
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | | | - Munther Abosoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Pharmacy, Imam Ja'afar Al-Sadiq University, Al-Samawa, Al-Muthanna, 66002, Iraq
| | | | | | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Payam Ali-Khiavi
- Medical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
5
|
Xu H, Zuo Y, Gao S, Liu Y, Liu T, He S, Wang M, Hu L, Li C, Yu Y. Circulating Tumor Cell Phenotype Detection and Epithelial-Mesenchymal Transition Tracking Based on Dual Biomarker Co-Recognition in an Integrated PDMS Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310360. [PMID: 38698606 DOI: 10.1002/smll.202310360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/13/2024] [Indexed: 05/05/2024]
Abstract
Circulating tumor cells (CTCs) are widely considered as a reliable and promising class of markers in the field of liquid biopsy. As CTCs undergo epithelial-mesenchymal transition (EMT), phenotype detection of heterogeneous CTCs based on EMT markers is of great significance. In this report, an integrated analytical strategy that can simultaneously capture and differentially detect epithelial- and mesenchymal-expressed CTCs in bloods of non-small cell lung cancer (NSCLS) patients is proposed. First, a commercial biomimetic polycarbonate (PCTE) microfiltration membrane is employed as the capture interface for heterogenous CTCs. Meanwhile, differential detection of the captured CTCs is realized by preparing two distinct CdTe quantum dots (QDs) with red and green emissions, attached with EpCAM and Vimentin aptamers, respectively. For combined analysis, a polydimethylsiloxane (PDMS) chip with simple structure is designed, which integrates the membrane capture and QDs-based phenotype detection of CTCs. This chip not only implements the analysis of the number of CTCs down to 2 cells mL-1, but enables EMT process tracking according to the specific signals of the two QDs. Finally, this method is successfully applied to inspect the correlations of numbers or proportions of heterogenous CTCs in 94 NSCLS patients with disease stage and whether there is distant metastasis.
Collapse
Affiliation(s)
- Hao Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Yingchun Zuo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Shuai Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Yuping Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Tingting Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Shiyu He
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Mengjiao Wang
- Department of Pharmacy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Lili Hu
- Department of Pharmacy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| |
Collapse
|
6
|
Wang X, Du Y, Jing W, Cao C, Wu X, Yang K, Zhu L. Fluorescent identification of immunomagnetically captured CTCs using triplex-aptamer-targeted dendritic SiO 2@Fe 3O 4 nanocomposite. Mikrochim Acta 2024; 191:424. [PMID: 38922365 DOI: 10.1007/s00604-024-06504-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
The enumeration of circulating tumor cells (CTCs) in peripheral blood plays a crucial role in the early diagnosis, recurrence monitoring, and prognosis assessment of cancer patients. There is a compelling need to develop an efficient technique for the capture and identification of these rare CTCs. However, the exclusive reliance on a single criterion, such as the epithelial cell adhesion molecule (EpCAM) antibody or aptamer, for the specific recognition of epithelial CTCs is not universally suitable for clinical applications, as it usually falls short in identifying EpCAM-negative CTCs. To address this limitation, we propose a straightforward and cost-effective method involving triplex fluorescently labelled aptamers (FAM-EpCAM, Cy5-PTK7, and Texas Red-CSV) to modify Fe3O4-loaded dendritic SiO2 nanocomposite (dmSiO2@Fe3O4/Apt). This multi-recognition-based strategy not only enhanced the efficiency in capturing heterogeneous CTCs, but also facilitated the rapid and accurate identification of CTCs. The capture efficiency of heterogenous CTCs reached up to 93.33%, with a detection limit as low as 5 cells/mL. Notably, the developed dmSiO2@Fe3O4/Apt nanoprobe enabled the swift identification of captured cells in just 30 min, relying solely on the fluorescently modified aptamers, which reduced the identification time by approximately 90% compared with the conventional immunocytochemistry (ICC) technique. Finally, these nanoprobe characteristics were validated using blood samples from patients with various types of cancers.
Collapse
Affiliation(s)
- Xinwen Wang
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Yu Du
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Weijun Jing
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, P. R. China
| | - Changchun Cao
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Xiaoli Wu
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Kangqun Yang
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China
| | - Liang Zhu
- Department of Pharmacy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, 6 Beijing West Road, Huaian, 223300, Jiangsu, P. R. China.
| |
Collapse
|
7
|
Li T, Li Y, Chen H, Li J, Liu Y, Tan W. Engineering a Dual-Receptor Targeted Multivalent Probe for Enhanced Magnetic Resonance Imaging of Metastatic Cancer. Anal Chem 2024; 96:4394-4401. [PMID: 38451935 DOI: 10.1021/acs.analchem.3c04036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Noninvasive monitoring of cancer metastasis is essential to improving clinical outcomes. Molecular MRI (mMRI) is a special implementation of noninvasive molecular imaging that promises to offer a powerful means for early detection and analysis of pathological states of cancer by tracking molecular markers. However, this is often hindered by the challenging issue of obtaining transformable mMRI contrast agents with high sensitivity, specificity, and broad applicability, given the high tumor heterogeneity and complex metastatic features. Herein, we present a dual-receptor targeted, multivalent recognition strategy and report a new class of mMRI probes for enhanced imaging of metastatic cancer. This probe is designed by covalently conjugating Gd-chelate with phenylboronic acid and an aptamer via an affordable polymerization chemistry to concurrently target two different cell-membrane receptors that are commonly overexpressed and highly implicated in both tumorigenesis and metastasis. Moreover, the polymerization chemistry allows the probe to contain a bunch of targeting ligands and signal reporters in a single chain, which not only leads to more than 2-fold enhancement in T1 relaxivity at 1.5 T compared to the commercial contrast agent but also enables it to actively target tumor cells in a multivalent recognition manner, contributing to a much higher imaging contrast than single-receptor targeted probes and the commercial agent in mouse models with lung metastases, yet without inducing systemic side effects. We expect this study to offer a useful molecular tool to promote transformable applications of mMRI and a better understanding of molecular mechanisms involved in cancer development.
Collapse
Affiliation(s)
- Ting Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yazhou Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Hong Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Jili Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
8
|
Cheng C, Wang H, Zhao J, Wang Y, Zhao G, Zhang Y, Liu X, Wang Y. Advances in the application of metal oxide nanozymes in tumor detection and treatment. Colloids Surf B Biointerfaces 2024; 235:113767. [PMID: 38295464 DOI: 10.1016/j.colsurfb.2024.113767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/02/2024]
Abstract
Natural enzymes play an important role to support the regular life activities of the human body. However, the application conditions of natural enzymes are harsh and there are limitations in their use. As artificial enzymes, nanozymes possess the substrate specificity of natural enzymes. Due to the advantages of low cost, good stability and strong catalytic properties, nanozymes hold a wide range of applications in the fields of sensing, chemical, food and medicine. Some of the more common ones are noble metal nanozymes, metal oxide nanozymes and carbon-based nanozymes. Among them, metal oxide nanozymes have attracted much attention because of their decent fixity, exceedingly good physicochemical properties and other advantages. Today, malignant tumors pose a great danger to the human body and are a serious threat to human health. However, traditional treatments have more side effects, and finding new treatment modalities is particularly important for tumor treatment. For example, enzyme therapy can be used to catalyze reactions in the body to achieve tumor treatment. Nanozymes can exert enzymatic activity and effectively treat malignant tumors through catalysis and synergy, and have made certain progress. This paper reviews the detection and application of metal oxide nanozymes in tumor detection and treatment in recent years and provides an outlook on their future application and development.
Collapse
Affiliation(s)
- Chunfang Cheng
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
| | - Huixin Wang
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
| | - Jingyu Zhao
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
| | - Yingying Wang
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
| | - Guanhui Zhao
- College of Chemistry and Chemical Engineering, Qilu Normal University, Jinan 250200, People's Republic of China.
| | - Yong Zhang
- Provincial Key Laboratory of Rural Energy Engineering in Yunnan, School of Energy and Environment Science, Yunnan Normal University, Kunming 650500, People's Republic of China
| | - Xin Liu
- Clinical Laboratory, Jinan Shizhong District People's Hospital, Jinan 250000, People's Republic of China
| | - Yaoguang Wang
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China.
| |
Collapse
|
9
|
Zhang Q, Gao X, Ho YP, Liu M, Han Y, Li DL, Yuan HM, Zhang CY. Controllable Assembly of a Quantum Dot-Based Aptasensor Guided by CRISPR/Cas12a for Direct Measurement of Circulating Tumor Cells in Human Blood. NANO LETTERS 2024; 24:2360-2368. [PMID: 38347661 DOI: 10.1021/acs.nanolett.3c04828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Accurate and sensitive analysis of circulating tumor cells (CTCs) in human blood provides a non-invasive approach for the evaluation of cancer metastasis and early cancer diagnosis. Herein, we demonstrate the controllable assembly of a quantum dot (QD)-based aptasensor guided by CRISPR/Cas12a for direct measurement of CTCs in human blood. We introduce a magnetic bead@activator/recognizer duplex core-shell structure to construct a multifunctional platform for the capture and direct detection of CTCs in human blood, without the need for additional CTC release and re-identification steps. Notably, the introduction of magnetic separation ensures that only a target-induced free activator can initiate the downstream catalysis, efficiently avoiding the undesired catalysis triggered by inappropriate recognition of the activator/recognizer duplex structure by crRNAs. This aptasensor achieves high CTC-capture efficiency (82.72%) and sensitive detection of CTCs with a limit of detection of 2 cells mL-1 in human blood, holding great promise for the liquid biopsy of cancers.
Collapse
Affiliation(s)
- Qian Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Xin Gao
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR 999077, China
| | - Meng Liu
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Yun Han
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Dong-Ling Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Hui-Min Yuan
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Chun-Yang Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
10
|
Gong J, Hu J, Yan X, Xiang L, Chen S, Yang H, Chen Z, Hou Q, Song Y, Xu Y, Liu D, Ji C, Qin Q, Sun H, Peng J, Cao B, Lu Y. Injectable Hydrogels Including Magnetic Nanosheets for Multidisciplinary Treatment of Hepatocellular Carcinoma via Magnetic Hyperthermia. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300733. [PMID: 37452437 DOI: 10.1002/smll.202300733] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/03/2023] [Indexed: 07/18/2023]
Abstract
Relapse and unresectability have become the main obstacle for further improving hepatocellular carcinoma (HCC) treatment effect. Currently, single therapy for HCC in clinical practice is limited by postoperative recurrence, intraoperative blood loss and poor patient outcomes. Multidisciplinary therapy has been recognized as the key to improving the long-term survival rate for HCC. However, the clinical application of HCC synthetic therapy is restricted by single functional biomaterials. In this study, a magnetic nanocomposite hydrogel (CG-IM) with iron oxide nanoparticle-loaded mica nanosheets (Iron oxide nanoparticles@Mica, IM) is reported. This biocompatible magnetic hydrogel integrated high injectability, magnetocaloric property, mechanical robustness, wet adhesion, and hemostasis, leading to efficient HCC multidisciplinary therapies including postoperative tumor margin treatment and percutaneous locoregional ablation. After minimally invasive hepatectomy of HCC, the CG-IM hydrogel can facilely seal the bleeding hepatic margin, followed by magnetic hyperthermia ablation to effectively prevent recurrence. In addition, CG-IM hydrogel can inhibit unresectable HCC by magnetic hyperthermia through the percutaneous intervention under ultrasound guidance.
Collapse
Affiliation(s)
- Jinyu Gong
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Jinlong Hu
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, 230041, P. R. China
| | - Xu Yan
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Luyao Xiang
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Sheng Chen
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Huai Yang
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Zichao Chen
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Qingbing Hou
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, 230041, P. R. China
| | - Yonghong Song
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| | - Yunjun Xu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230036, P. R. China
| | - Dongquan Liu
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, 230041, P. R. China
| | - Chaofei Ji
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, 230041, P. R. China
| | - Qin Qin
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, 230041, P. R. China
| | - Haiyi Sun
- The First Clinical College, Anhui Medical University, Hefei, 230022, P. R. China
| | - Junbin Peng
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, 230041, P. R. China
| | - Baoqiang Cao
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Hefei, 230041, P. R. China
| | - Yang Lu
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, 230009, P. R. China
| |
Collapse
|
11
|
Nie C, Shaw I, Chen C. Application of microfluidic technology based on surface-enhanced Raman scattering in cancer biomarker detection: A review. J Pharm Anal 2023; 13:1429-1451. [PMID: 38223444 PMCID: PMC10785256 DOI: 10.1016/j.jpha.2023.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 01/16/2024] Open
Abstract
With the continuous discovery and research of predictive cancer-related biomarkers, liquid biopsy shows great potential in cancer diagnosis. Surface-enhanced Raman scattering (SERS) and microfluidic technology have received much attention among the various cancer biomarker detection methods. The former has ultrahigh detection sensitivity and can provide a unique fingerprint. In contrast, the latter has the characteristics of miniaturization and integration, which can realize accurate control of the detection samples and high-throughput detection through design. Both have the potential for point-of-care testing (POCT), and their combination (lab-on-a-chip SERS (LoC-SERS)) shows good compatibility. In this paper, the basic situation of circulating proteins, circulating tumor cells, exosomes, circulating tumor DNA (ctDNA), and microRNA (miRNA) in the diagnosis of various cancers is reviewed, and the detection research of these biomarkers by the LoC-SERS platform in recent years is described in detail. At the same time, the challenges and future development of the platform are discussed at the end of the review. Summarizing the current technology is expected to provide a reference for scholars engaged in related work and interested in this field.
Collapse
Affiliation(s)
- Changhong Nie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Ibrahim Shaw
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Chuanpin Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| |
Collapse
|
12
|
Ren J, Chen Z, Ma E, Wang W, Zheng S, Wang H. Dual-source powered nanomotors coupled with dual-targeting ligands for efficient capture and detection of CTCs in whole blood and in vivo tumor imaging. Colloids Surf B Biointerfaces 2023; 231:113568. [PMID: 37826963 DOI: 10.1016/j.colsurfb.2023.113568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/02/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023]
Abstract
Circulating tumor cells (CTCs) are important biomarkers in cancer diagnosis. However, the specific labeling of CTCs with high capture efficiency in whole blood remains a problem. Herein, a dual-source-driven nanomotor coupled with dual-targeting ligands (CD@NM) was designed for efficient capture, specific imaging and quantitative detection of CTCs. In both water and biological fluid, CD@NMs moved autonomously under the propulsion of a magnetic field and H2O2 solution, which improved the capture efficiency of CTCs to 97.50 ± 2.38%. More importantly, specific labeling of CTCs was achieved by fluorescence quenching and recovery of fluorescent carbon dots modified on the CD@NMs. As a result, the CD@NMs exhibited efficient CTC capture, specific CTC imaging and recognition in whole blood. CD@NMs were also successfully deployed in the specific imaging of tumor tissues in vivo. On this basis, CD@NMs are expected to provide a new platform for tumor diagnosis both in vitro and in vivo.
Collapse
Affiliation(s)
- Jiaoyu Ren
- School of Chemical Engineering & Technology, China University of Mining and Technology, Xuzhou, Jiangsu 221116, PR China
| | - Zekun Chen
- School of Chemical Engineering & Technology, China University of Mining and Technology, Xuzhou, Jiangsu 221116, PR China
| | - Enhui Ma
- School of Chemical Engineering & Technology, China University of Mining and Technology, Xuzhou, Jiangsu 221116, PR China
| | - Wenjun Wang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221116, PR China
| | - Shaohui Zheng
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu 221116, PR China.
| | - Hong Wang
- School of Chemical Engineering & Technology, China University of Mining and Technology, Xuzhou, Jiangsu 221116, PR China.
| |
Collapse
|
13
|
Jiang L, Lin X, Chen F, Qin X, Yan Y, Ren L, Yu H, Chang L, Wang Y. Current research status of tumor cell biomarker detection. MICROSYSTEMS & NANOENGINEERING 2023; 9:123. [PMID: 37811123 PMCID: PMC10556054 DOI: 10.1038/s41378-023-00581-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/26/2023] [Accepted: 07/23/2023] [Indexed: 10/10/2023]
Abstract
With the annual increases in the morbidity and mortality rates of tumors, the use of biomarkers for early diagnosis and real-time monitoring of tumor cells is of great importance. Biomarkers used for tumor cell detection in body fluids include circulating tumor cells, nucleic acids, protein markers, and extracellular vesicles. Among them, circulating tumor cells, circulating tumor DNA, and exosomes have high potential for the prediction, diagnosis, and prognosis of tumor diseases due to the large amount of valuable information on tumor characteristics and evolution; in addition, in situ monitoring of telomerase and miRNA in living cells has been the topic of extensive research to understand tumor development in real time. Various techniques, such as enzyme-linked immunosorbent assays, immunoblotting, and mass spectrometry, have been widely used for the detection of these markers. Among them, the detection of tumor cell markers in body fluids based on electrochemical biosensors and fluorescence signal analysis is highly preferred because of its high sensitivity, rapid detection and portable operation. Herein, we summarize recent research progress in the detection of tumor cell biomarkers in body fluids using electrochemical and fluorescence biosensors, outline the current research status of in situ fluorescence monitoring and the analysis of tumor markers in living cells, and discuss the technical challenges for their practical clinical application to provide a reference for the development of new tumor marker detection methods.
Collapse
Affiliation(s)
- Liying Jiang
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
- Academy for Quantum Science and Technology, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Xinyi Lin
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Fenghua Chen
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Xiaoyun Qin
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Yanxia Yan
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Linjiao Ren
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002 China
| | - Hongyu Yu
- Department of Mechanical and Aerospace Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Lingqian Chang
- key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083 China
| | - Yang Wang
- key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083 China
- School of Engineering Medicine, Beihang University, Beijing, 100083 China
| |
Collapse
|
14
|
Guo W, Ying P, Ma R, Jing Z, Ma G, Long J, Li G, Liu Z. Liquid biopsy analysis of lipometabolic exosomes in pancreatic cancer. Cytokine Growth Factor Rev 2023; 73:69-77. [PMID: 37684117 DOI: 10.1016/j.cytogfr.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 09/10/2023]
Abstract
Pancreatic cancer is characterized by its high malignancy, insidious onset and poor prognosis. Most patients with pancreatic cancer are usually diagnosed at advanced stage or with the distant metastasis due to the lack of an effective early screening method. Liquid biopsy technology is promising in studying the occurrence, progression, and early metastasis of pancreatic cancer. In particular, exosomes are pivotal biomarkers in lipid metabolism and liquid biopsy of blood exosomes is valuable for the evaluation of pancreatic cancer. Lipid metabolism is crucial for the formation and activity of exosomes in the extracellular environment. Exosomes and lipids have a complex relationship of mutual influence. Furthermore, spatial metabolomics can quantify the levels and spatial locations of individual metabolites in cancer tissue, cancer stroma, and para-cancerous tissue in pancreatic cancer. However, the relationship among exosomes, lipid metabolism, and pancreatic cancer is also worth considering. This study mainly updates the research progress of metabolomics in pancreatic cancer, their relationship with exosomes, an important part of liquid biopsy, and their lipometabolic roles in pancreatic cancer. We also discuss the mechanisms by which possible metabolites, especially lipid metabolites through exosome transport and other processes, contribute to the recurrence and metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Peiyao Ying
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Ruiyang Ma
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
| | - Zuoqian Jing
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Gang Ma
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jin Long
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Guichen Li
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, China.
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
15
|
Tao Y, Yan C, Wu Y, Li D, Li J, Xie Y, Cheng Y, Xu Y, Yang K, Zhu W, Guo Z. Uniting Dual‐Modal MRI/Chemiluminescence Nanotheranostics: Spatially and Sensitively Self‐Reporting Photodynamic Therapy in Oral Cancer. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202303240] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Indexed: 01/22/2025]
Abstract
AbstractUnpredictable in vivo therapeutic feedback of reactive oxygen species (ROS) efficiency is the major bottleneck of photodynamic therapy (PDT). Herein, novel PDT‐based nanotheranostics Pa–Mn&CH‐A@P are elaborately constructed for in vivo tracking biodistribution and in situ self‐reporting PDT, which innovatively unites magnetic resonance imaging (MRI) and chemiluminescence (CL) signals. Taking advantages of the versatility of lanthanide coordination chemistry and flash nanoprecipitation (FNP) technology, photosensitizers, MRI, and CL agents are unprecedently integrated within a stable and uniform nanotheranostic. Specifically, MRI signal offers detailed dose distribution of nanotheranostics with high‐spatial resolution, and CL signal timely performs in situ evaluation of ROS generation with high sensitivity. This dual‐modal MRI/CL nanotheranostic makes a breakthrough in high fidelity feedback for oral tumor, conquering the inherent unpredictable obstacles on spatially and sensitively reporting PDT.
Collapse
Affiliation(s)
- Yining Tao
- Department of Interventional Radiology Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai 200233 China
- State Key Laboratory of Bioreactor Engineering Shanghai Key Laboratory of Functional Materials Chemistry Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering Frontiers Science Center for Materiobiology and Dynamic Chemistry Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism Institute of Fine Chemicals School of Chemistry and Molecular Engineering East China University of Science and Technology
| | - Chenxu Yan
- State Key Laboratory of Bioreactor Engineering Shanghai Key Laboratory of Functional Materials Chemistry Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering Frontiers Science Center for Materiobiology and Dynamic Chemistry Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism Institute of Fine Chemicals School of Chemistry and Molecular Engineering East China University of Science and Technology
| | - Yue Wu
- State Key Laboratory of Chemical Engineering East China University of Science and Technology Shanghai 200237 China
| | - Dan Li
- State Key Laboratory of Bioreactor Engineering Shanghai Key Laboratory of Functional Materials Chemistry Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering Frontiers Science Center for Materiobiology and Dynamic Chemistry Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism Institute of Fine Chemicals School of Chemistry and Molecular Engineering East China University of Science and Technology
| | - Juan Li
- State Key Laboratory of Bioreactor Engineering Shanghai Key Laboratory of Functional Materials Chemistry Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering Frontiers Science Center for Materiobiology and Dynamic Chemistry Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism Institute of Fine Chemicals School of Chemistry and Molecular Engineering East China University of Science and Technology
| | - Yuchen Xie
- Human Oncology and Pathogenesis Program Memorial Sloan Kettering Cancer Center New York NY 10065 USA
| | - Yingsheng Cheng
- Department of Interventional Radiology Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai 200233 China
- Department of Imaging Medicine and Nuclear Medicine Tongji Hospital Shanghai 200065 China
| | - Yisheng Xu
- State Key Laboratory of Chemical Engineering East China University of Science and Technology Shanghai 200237 China
| | - Kai Yang
- Department of Interventional Radiology Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai 200233 China
- Department of Imaging Medicine and Nuclear Medicine Tongji Hospital Shanghai 200065 China
| | - Wei‐Hong Zhu
- State Key Laboratory of Bioreactor Engineering Shanghai Key Laboratory of Functional Materials Chemistry Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering Frontiers Science Center for Materiobiology and Dynamic Chemistry Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism Institute of Fine Chemicals School of Chemistry and Molecular Engineering East China University of Science and Technology
| | - Zhiqian Guo
- State Key Laboratory of Bioreactor Engineering Shanghai Key Laboratory of Functional Materials Chemistry Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering Frontiers Science Center for Materiobiology and Dynamic Chemistry Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism Institute of Fine Chemicals School of Chemistry and Molecular Engineering East China University of Science and Technology
| |
Collapse
|
16
|
Hu H, Krishaa L, Fong ELS. Magnetic force-based cell manipulation for in vitro tissue engineering. APL Bioeng 2023; 7:031504. [PMID: 37736016 PMCID: PMC10511261 DOI: 10.1063/5.0138732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 08/22/2023] [Indexed: 09/23/2023] Open
Abstract
Cell manipulation techniques such as those based on three-dimensional (3D) bioprinting and microfluidic systems have recently been developed to reconstruct complex 3D tissue structures in vitro. Compared to these technologies, magnetic force-based cell manipulation is a simpler, scaffold- and label-free method that minimally affects cell viability and can rapidly manipulate cells into 3D tissue constructs. As such, there is increasing interest in leveraging this technology for cell assembly in tissue engineering. Cell manipulation using magnetic forces primarily involves two key approaches. The first method, positive magnetophoresis, uses magnetic nanoparticles (MNPs) which are either attached to the cell surface or integrated within the cell. These MNPs enable the deliberate positioning of cells into designated configurations when an external magnetic field is applied. The second method, known as negative magnetophoresis, manipulates diamagnetic entities, such as cells, in a paramagnetic environment using an external magnetic field. Unlike the first method, this technique does not require the use of MNPs for cell manipulation. Instead, it leverages the magnetic field and the motion of paramagnetic agents like paramagnetic salts (Gadobutrol, MnCl2, etc.) to propel cells toward the field minimum, resulting in the assembly of cells into the desired geometrical arrangement. In this Review, we will first describe the major approaches used to assemble cells in vitro-3D bioprinting and microfluidics-based platforms-and then discuss the use of magnetic forces for cell manipulation. Finally, we will highlight recent research in which these magnetic force-based approaches have been applied and outline challenges to mature this technology for in vitro tissue engineering.
Collapse
Affiliation(s)
- Huiqian Hu
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - L. Krishaa
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Eliza Li Shan Fong
- Present address: Translational Tumor Engineering Laboratory, 15 Kent Ridge Cres, E7, 06-01G, Singapore 119276, Singapore. Author to whom correspondence should be addressed:
| |
Collapse
|
17
|
Wang Z, Qin H, Liu S, Sheng J, Zhang X. Precision diagnosis of hepatocellular carcinoma. Chin Med J (Engl) 2023; 136:1155-1165. [PMID: 36939276 PMCID: PMC10278703 DOI: 10.1097/cm9.0000000000002641] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Indexed: 03/21/2023] Open
Abstract
ABSTRACT Hepatocellular carcinoma (HCC) is the most common type of primary hepatocellular carcinoma (PHC). Early diagnosis of HCC remains the key to improve the prognosis. In recent years, with the promotion of the concept of precision medicine and more in-depth analysis of the biological mechanism underlying HCC, new diagnostic methods, including emerging serum markers, liquid biopsies, molecular diagnosis, and advances in imaging (novel contrast agents and radiomics), have emerged one after another. Herein, we reviewed and analyzed scientific advances in the early diagnosis of HCC and discussed their application and shortcomings. This review aimed to provide a reference for scientific research and clinical practice of HCC.
Collapse
Affiliation(s)
- Zhenxiao Wang
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Hanjiao Qin
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Shui Liu
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| |
Collapse
|
18
|
Xia W, Singh N, Goel S, Shi S. Molecular Imaging of Innate Immunity and Immunotherapy. Adv Drug Deliv Rev 2023; 198:114865. [PMID: 37182699 DOI: 10.1016/j.addr.2023.114865] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/17/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
The innate immune system plays a key role as the first line of defense in various human diseases including cancer, cardiovascular and inflammatory diseases. In contrast to tissue biopsies and blood biopsies, in vivo imaging of the innate immune system can provide whole body measurements of immune cell location and function and changes in response to disease progression and therapy. Rationally developed molecular imaging strategies can be used in evaluating the status and spatio-temporal distributions of the innate immune cells in near real-time, mapping the biodistribution of novel innate immunotherapies, monitoring their efficacy and potential toxicities, and eventually for stratifying patients that are likely to benefit from these immunotherapies. In this review, we will highlight the current state-of-the-art in noninvasive imaging techniques for preclinical imaging of the innate immune system particularly focusing on cell trafficking, biodistribution, as well as pharmacokinetics and dynamics of promising immunotherapies in cancer and other diseases; discuss the unmet needs and current challenges in integrating imaging modalities and immunology and suggest potential solutions to overcome these barriers.
Collapse
Affiliation(s)
- Wenxi Xia
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Neetu Singh
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States
| | - Shreya Goel
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States
| | - Sixiang Shi
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, United States; Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
19
|
Guo L, Liu C, Qi M, Cheng L, Wang L, Li C, Dong B. Recent progress of nanostructure-based enrichment of circulating tumor cells and downstream analysis. LAB ON A CHIP 2023; 23:1493-1523. [PMID: 36776104 DOI: 10.1039/d2lc00890d] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The isolation and detection of circulating tumor cells (CTCs) play an important role in early cancer diagnosis and prognosis, providing easy access to identify metastatic cells before clinically detectable metastases. In the past 20 years, according to the heterogeneous expression of CTCs on the surface and their special physical properties (size, morphology, electricity, etc.), a series of in vitro enrichment methods of CTCs have been developed based on microfluidic chip technology, nanomaterials and various nanostructures. In recent years, the in vivo detection of CTCs has attracted considerable attention. Photoacoustic flow cytometry and fluorescence flow cytometry were used to detect CTCs in a noninvasive manner. In addition, flexible magnetic wire and indwelling intravascular non-circulating CTCs isolation system were developed for in vivo CTCs study. In the aspect of downstream analysis, gene analysis and drug sensitivity tests of enriched CTCs were developed based on various existing molecular analysis techniques. All of these studies constitute a complete study of CTCs. Although the existing reviews mainly focus on one aspect of capturing CTCs study, a review that includes the in vivo and in vitro capture and downstream analysis study of CTCs is highly needed. This review focuses on not only the classic work and latest research progress in in vitro capture but also includes the in vivo capture and downstream analysis, discussing the advantages and significance of the different research methods and providing new ideas for solving the heterogeneity and rarity of CTCs.
Collapse
Affiliation(s)
- Lihua Guo
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, P. R. China.
| | - Chang Liu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, P. R. China.
| | - Manlin Qi
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School of Stomatology, Jilin University, Changchun, 130021, P. R. China.
| | - Liang Cheng
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School of Stomatology, Jilin University, Changchun, 130021, P. R. China.
| | - Lin Wang
- Department of Oral Implantology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School of Stomatology, Jilin University, Changchun, 130021, P. R. China.
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China.
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, P. R. China.
| |
Collapse
|
20
|
Chen J, Niu C, Yang N, Liu C, Zou SS, Zhu S. Biomarker discovery and application-An opportunity to resolve the challenge of liver cancer diagnosis and treatment. Pharmacol Res 2023; 189:106674. [PMID: 36702425 DOI: 10.1016/j.phrs.2023.106674] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/10/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
Liver cancer is one of the most common malignancies, with severe morbidity and mortality. While considerable progress has been made in liver cancer treatment, the 5-year overall survival (OS) of patients has not improved significantly. Reasons include the inadequate capability of early screening and diagnosis, a high incidence of recurrence and metastasis, a high degree of tumor heterogeneity, and an immunosuppressive tumor microenvironment. Therefore, the identification and validation of specific and robust liver cancer biomarkers are of major importance for early screening, timely diagnosis, accurate prognosis, and the prevention of tumor progression. In this review, we highlight some of the latest research progress and potential applications of liver cancer biomarkers, describing hotspots and prospective directions in biomarker discovery.
Collapse
Affiliation(s)
- Jingtao Chen
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China; Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China
| | - Chao Niu
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Ning Yang
- Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China
| | - Chunyan Liu
- Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shan-Shan Zou
- Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shan Zhu
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China; Laboratory for Tumor Immunology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
21
|
Wang X, Gao T, Zhu J, Long S, Zhao S, Yuan L, Wang Z. Fabrication of Channeled and Three-Dimensional Electrodes for the Integrated Capture and Detection of Invasive Circulating Tumor Cells during Hematogenous Metastasis. Anal Chem 2023; 95:2496-2503. [PMID: 36639744 DOI: 10.1021/acs.analchem.2c04809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Hematogenous metastasis is the main route of cancer spreading, causing majority death of cancer patients. During this process, platelets in the blood are found increasingly essential to promote hematogenous metastasis by forming platelet-interacted circulating tumor cells (CTCs). Hence, we aim to fabricate an integrated method for the availability of capture and detection of such invasive CTCs. Specifically, a new form of channeled and conductive three-dimensional (3D) electrode is constructed by modifying a conductive layer and capture antibody on the templated and channeled poly(dimethylsiloxane) scaffold. The modified antibody enables the capture of the platelet-interacted CTC hybrid, while the conductive layer significantly facilitates electron transfer from electro-active signal molecules that are targeting platelets. Therefore, sensitive electrochemical detection of platelet-interacted CTCs has been realized. Efficient capture and sensitive detection have been demonstrated by this work. Additionally, dynamic analysis of patients' CTCs has also been conducted to provide accurate information about disease assessment and efficacy evaluation. The cut-off line was set as 5.15 nA based on the sample signals from healthy volunteers. Thus, stage III cancer patients with high risk of hematogenous metastasis have been identified. Together, this work shows the development of a new strategy for simultaneous capture and detection of the invasive CTC subtype form patient blood, which favors precise monitoring of hematogenous metastasis.
Collapse
Affiliation(s)
- Xiaoying Wang
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210000, P. R. China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 211100, P. R. China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, P. R. China
| | - Tao Gao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, P. R. China
| | - Jin Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, P. R. China
| | - Shipeng Long
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, P. R. China
| | - Songyan Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, P. R. China
| | - Li Yuan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing 211100, P. R. China
| | - Zhaoxia Wang
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210000, P. R. China
| |
Collapse
|
22
|
Jiang B, Xie D, Wang S, Li X, Wu G. Advances in early detection methods for solid tumors. Front Genet 2023; 14:1091223. [PMID: 36911396 PMCID: PMC9998680 DOI: 10.3389/fgene.2023.1091223] [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: 11/06/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
During the last decade, non-invasive methods such as liquid biopsy have slowly replaced traditional imaging and invasive pathological methods used to diagnose and monitor cancer. Improvements in the available detection methods have enabled the early screening and diagnosis of solid tumors. In addition, advances in early detection methods have made the continuous monitoring of tumor progression using repeat sampling possible. Previously, the focus of liquid biopsy techniques included the following: 1) the isolation of circulating tumor cells, circulating tumor DNA, and extracellular tumor vesicles from solid tumor cells in the patient's blood; in addition to 2) analyzing genomic and proteomic data contained within the isolates. Recently, there has been a rapid devolvement in the techniques used to isolate and analyze molecular markers. This rapid evolvement in detection techniques improves their accuracy, especially when few samples are available. In addition, there is a tremendous expansion in the acquisition of samples and targets for testing; solid tumors can be detected from blood and other body fluids. Test objects have also expanded from samples taken directly from cancer to include indirect objects affected in cancer development. Liquid biopsy technology has limitations. Even so, this detection technique is the key to a new phase of oncogenetics. This review aims to provide an overview of the current advances in liquid biopsy marker selection, isolation, and detection methods for solid tumors. The advantages and disadvantages of liquid biopsy technology will also be explored.
Collapse
Affiliation(s)
- Bowen Jiang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deqian Xie
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiunan Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
23
|
Jia L, Zhen X, Chen L, Feng Q, Yuan W, Bu Y, Wang S, Xie X. Bioinspired nano-plate-coral platform enabled efficient detection of circulating tumor cells via the synergistic capture of multivalent aptamer and tumor cell membrane. J Colloid Interface Sci 2022; 631:55-65. [DOI: 10.1016/j.jcis.2022.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/31/2022] [Accepted: 11/06/2022] [Indexed: 11/10/2022]
|
24
|
Jia W, Han Y, Mao X, Xu W, Zhang Y. Nanotechnology strategies for hepatocellular carcinoma diagnosis and treatment. RSC Adv 2022; 12:31068-31082. [PMID: 36349046 PMCID: PMC9621307 DOI: 10.1039/d2ra05127c] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/20/2022] [Indexed: 10/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy threatening human health, and existing diagnostic and therapeutic techniques are facing great challenges. In the last decade or so, nanotechnology has been developed and improved for tumor diagnosis and treatment. For example, nano-intravenous injections have been approved for malignant perivascular epithelioid cell tumors. This article provides a comprehensive review of the applications of nanotechnology in HCC in recent years: (I) in radiological imaging, magnetic resonance imaging (MRI), fluorescence imaging (FMI) and multimodality imaging. (II) For diagnostic applications in HCC serum markers. (III) As embolic agents in transarterial chemoembolization (TACE) or directly as therapeutic drugs. (IV) For application in photothermal therapy and photodynamic therapy. (V) As carriers of chemotherapeutic drugs, targeted drugs, and natural plant drugs. (VI) For application in gene and immunotherapy. Compared with the traditional methods for diagnosis and treatment of HCC, nanoparticles have high sensitivity, reduce drug toxicity and have a long duration of action, and can also be combined with photothermal and photodynamic multimodal combination therapy. These summaries provide insights for the further development of nanotechnology applications in HCC.
Collapse
Affiliation(s)
- WeiLu Jia
- Medical School, Southeast University Nanjing 210009 China
| | - YingHui Han
- Outpatient Department, The Second Affiliated Hospital of Nanjing Medical University Nanjing 210009 China
| | - XinYu Mao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University Nanjing 210009 China
| | - WenJing Xu
- Medical School, Southeast University Nanjing 210009 China
| | - YeWei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University Nanjing 210009 China
| |
Collapse
|
25
|
Jiang X, Zhang X, Guo C, Yu Y, Ma B, Liu Z, Chai Y, Wang L, Du Y, Wang B, Li N, Dong D, Li Y, Huang X, Ou L. Protein corona-coated immunomagnetic nanoparticles with enhanced isolation of circulating tumor cells. NANOSCALE 2022; 14:8474-8483. [PMID: 35661186 DOI: 10.1039/d2nr01568d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Immunomagnetic nanoparticles (IMNs) have been widely developed as a detection tool to isolate rare circulating tumor cells (CTCs) from whole blood as a potential method for early cancer diagnosis, metastasis examination, and treatment guidance. However, a spontaneous interaction between nanoparticles and proteins results in the formation of a protein corona that reduces the performance of IMNs when they enter body fluids. To address this issue, the protein corona was precoated onto magnetic nanoparticles (C-MNs), and then their surfaces were conjugated with an immuno-antibody. The adsorption of proteins on C-MNs was decreased 6-fold and non-specific cell binding was reduced 5-fold, compared with magnetic nanoparticles (MNs). Furthermore, the immuno-antibody functionalized C-MNs (IC-MNs) maintained highly specific CTC capture performance when exposed to blood plasma. By using artificial spiked blood samples, IC-MNs exhibited 90.2% CTC isolation efficiency, compared with 60.3% by using IMNs. IC-MNs also successfully captured CTCs with high purity in 24 out of 26 female breast cancer patient blood samples. This work demonstrated that a novel preformed protein corona strategy can provide a useful clinically applicable diagnostic tool.
Collapse
Affiliation(s)
- Xinbang Jiang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Xiangyun Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Chen Guo
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Yameng Yu
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Boya Ma
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Zhuang Liu
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Yamin Chai
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Lichun Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Yunzheng Du
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Biao Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Nan Li
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Dong Dong
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yueguo Li
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| | - Lailiang Ou
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China.
| |
Collapse
|
26
|
Yan X, Sun T, Song Y, Peng W, Xu Y, Luo G, Li M, Chen S, Fang WW, Dong L, Xuan S, He T, Cao B, Lu Y. In situ Thermal-Responsive Magnetic Hydrogel for Multidisciplinary Therapy of Hepatocellular Carcinoma. NANO LETTERS 2022; 22:2251-2260. [PMID: 35254836 DOI: 10.1021/acs.nanolett.1c04413] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Current surgical single modality treatments for hepatocellular carcinoma (HCC) were restricted by recurrence, blood loss, significant trauma, and poor prognostic. Although multidisciplinary strategies for HCC treatment have been highly recommended by the clinical guidelines, there was limited choice of materials and treatments. Herein, we reported an in situ formed magnetic hydrogel with promising bioapplicable thermal-responsiveness, strong adhesion in wet conditions, high magnetic hyperthermia, and biocompatibility, leading to efficient HCC multidisciplinary treatment including postoperative treatment and transarterial embolization therapy. In vivo results indicated that this hydrogel could reduce the postoperative recurrence rate. The hemostatic ability of the thermal-responsive hydrogel was further demonstrated in both the liver scratch model and liver tumor resection. Computed tomography imaging suggested that the hydrogel could completely embolize the arterial vessels of rabbit liver tumor by vascular intervention operation, which could serve as multidisciplinary responsive materials to external magnetic field and body temperature for HCC treatment.
Collapse
Affiliation(s)
- Xu Yan
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Tianci Sun
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Yonghong Song
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Wei Peng
- Department of General Surgery, Department of Interventional Radiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, China
| | - Yunjun Xu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Guangyi Luo
- Department of General Surgery, Department of Interventional Radiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, China
| | - Min Li
- Department of General Surgery, Department of Interventional Radiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, China
| | - Sheng Chen
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Wei-Wei Fang
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Liang Dong
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Shouhu Xuan
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230036, China
| | - Tao He
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| | - Baoqiang Cao
- Department of General Surgery, Department of Interventional Radiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, China
| | - Yang Lu
- School of Chemistry and Chemical Engineering, Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, Anhui 230009, People's Republic of China
| |
Collapse
|
27
|
Liu X, Ma L, Yan W, Aazmi A, Fang M, Xu X, Kang H, Xu X. A review of recent progress toward the efficient separation of circulating tumor cells via micro‐/nanostructured microfluidic chips. VIEW 2022. [DOI: 10.1002/viw.20210013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Xiaoshi Liu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Liang Ma
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou P. R. China
| | - Wenyuan Yan
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou P. R. China
| | - Minghe Fang
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Xiuzhen Xu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Hanyue Kang
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Xiaobin Xu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| |
Collapse
|
28
|
He L, He F, Feng Y, Wang X, Li Y, Tian Y, Gao A, Zhang P, Qi X, Luo Z, Duan Y. Hybridized nanolayer modified Ω-shaped fiber-optic synergistically enhances localized surface plasma resonance for ultrasensitive cytosensor and efficient photothermal therapy. Biosens Bioelectron 2021; 194:113599. [PMID: 34521011 DOI: 10.1016/j.bios.2021.113599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 02/07/2023]
Abstract
Inadequate sensitivity and side-effect are the main challenges to develop cytosensors combining with therapeutic potential simultaneously for cancer diagnosis and treatment. Herein, localized surface plasma resonance (LSPR) based on hybridized nanolayer modified Ω-shaped fiber-optic (HN/Ω-FO) was developed to integrate cytosensor and plasmonic photothermal treatment (PPT). On one hand, hybridized nanolayers improve the coverage of nanoparticles and refractive index sensitivity (RIS). Moreover, the hybridized nanoploymers of gold nanorods/gold nanoparticles (AuNRs/AuNPs) also result in intense enhancement in electronic field intensity (I). On the other hand, Ω-shaped fiber-optic (Ω-FO) led to strong bending loss in its bending part. To be specific, a majority of light escaped from fiber will interact with HN. Thus, HN/Ω-FO synergistically enhances the plasmonic, which achieved the goal of ultrasensitive cytosensor and highly-efficient plasmonic photothermal treatment (PPT). The proposed cytosensor exhibits ultrasensitivity for detection of cancer cells with a low limit of detection down to 2.6 cells/mL was realized just in 30 min. HN/Ω-FO-based LSPR exhibits unique characteristics of highly efficient, localized, and geometry-dependent heat distribution, which makes it suitable for PPT to only kill the cancer cells specifically on the surface or surrounding fiber-optic (FO) surface. Thus, HN/Ω-FO provides a new approach to couple cytosensor with PPT, indicating its great potential in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Lu He
- Research Center of Analytical Instrumentation, Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, 710069, Shaanxi, PR China
| | - Fan He
- School of Physics, Northwest University, Xi'an, 710069, Shaanxi, PR China
| | - Yanting Feng
- Research Center of Analytical Instrumentation, Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, 710069, Shaanxi, PR China
| | - Xu Wang
- Research Center of Analytical Instrumentation, School of Mechanical Engineering, Sichuan University, Chengdu, 610065, PR China
| | - Yongxin Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Yonghui Tian
- Research Center of Analytical Instrumentation, Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, 710069, Shaanxi, PR China
| | - Aihua Gao
- School of Physics, Northwest University, Xi'an, 710069, Shaanxi, PR China
| | - Pei Zhang
- Research Center of Analytical Instrumentation, Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, 710069, Shaanxi, PR China
| | - Xinyuan Qi
- School of Physics, Northwest University, Xi'an, 710069, Shaanxi, PR China.
| | - Zewei Luo
- Research Center of Analytical Instrumentation, Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, 710069, Shaanxi, PR China.
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an, 710069, Shaanxi, PR China.
| |
Collapse
|
29
|
Lin D, Shen L, Luo M, Zhang K, Li J, Yang Q, Zhu F, Zhou D, Zheng S, Chen Y, Zhou J. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther 2021; 6:404. [PMID: 34803167 PMCID: PMC8606574 DOI: 10.1038/s41392-021-00817-8] [Citation(s) in RCA: 431] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cells (CTCs) are tumor cells that have sloughed off the primary tumor and extravasate into and circulate in the blood. Understanding of the metastatic cascade of CTCs has tremendous potential for the identification of targets against cancer metastasis. Detecting these very rare CTCs among the massive blood cells is challenging. However, emerging technologies for CTCs detection have profoundly contributed to deepening investigation into the biology of CTCs and have facilitated their clinical application. Current technologies for the detection of CTCs are summarized herein, together with their advantages and disadvantages. The detection of CTCs is usually dependent on molecular markers, with the epithelial cell adhesion molecule being the most widely used, although molecular markers vary between different types of cancer. Properties associated with epithelial-to-mesenchymal transition and stemness have been identified in CTCs, indicating their increased metastatic capacity. Only a small proportion of CTCs can survive and eventually initiate metastases, suggesting that an interaction and modulation between CTCs and the hostile blood microenvironment is essential for CTC metastasis. Single-cell sequencing of CTCs has been extensively investigated, and has enabled researchers to reveal the genome and transcriptome of CTCs. Herein, we also review the clinical applications of CTCs, especially for monitoring response to cancer treatment and in evaluating prognosis. Hence, CTCs have and will continue to contribute to providing significant insights into metastatic processes and will open new avenues for useful clinical applications.
Collapse
Affiliation(s)
- Danfeng Lin
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lesang Shen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Luo
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinfan Li
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Yang
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fangfang Zhu
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhou
- Department of Surgery, Traditional Chinese Medical Hospital of Zhuji, Shaoxing, China
| | - Shu Zheng
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiding Chen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiaojiao Zhou
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
30
|
Gao R, Zhan C, Wu C, Lu Y, Cao B, Huang J, Wang F, Yu L. Simultaneous single-cell phenotype analysis of hepatocellular carcinoma CTCs using a SERS-aptamer based microfluidic chip. LAB ON A CHIP 2021; 21:3888-3898. [PMID: 34387639 DOI: 10.1039/d1lc00516b] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Hepatocellular carcinoma (HCC) is a harmful malady that truly debilitates human health, and hence it is of significance to isolate and on-line profile the phenotype of HCC cells for further diagnosis and therapy. We developed a novel strategy for efficient capture and in situ heterogeneous phenotype analysis of circulating tumor cells (CTCs) at the single-cell level based on surface-enhanced Raman scattering (SERS) fingerprint characteristics. Herein, a new microfluidic chip with lantern-like bypass structure was designed to capture CTCs by their large size from whole blood. Furthermore, two types of SERS-aptamer nanotags were fabricated, realizing spectral recognition of single CTCs in accordance with the surface membrane protein expression. Up to 84% of CTCs with a purity of 95% were captured from whole blood samples using the present SERS-aptamer based microfluidic chip at 20 μL min-1. The results showed that the proposed strategy can successfully identify HCC cell subtypes by SERS measurements, which was related to the clinical surface biomarkers. This may open a new avenue for serving as a powerful tool of cancer diagnosis and prognosis evaluation.
Collapse
Affiliation(s)
- Rongke Gao
- State Key Laboratory of Advanced Display Technology, School of Instrument Science and Optoelectronic Engineering, Hefei University of Technology, Hefei 230009, China
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Changbiao Zhan
- State Key Laboratory of Advanced Display Technology, School of Instrument Science and Optoelectronic Engineering, Hefei University of Technology, Hefei 230009, China
| | - Chunyu Wu
- State Key Laboratory of Advanced Display Technology, School of Instrument Science and Optoelectronic Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yang Lu
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| | - Baoqiang Cao
- Department of Hepatobiliary Pancreatic Surgery, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China
| | - Jing Huang
- Hefei University of Technology Hospital, Hefei 230009, China
| | - Feng Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Liandong Yu
- College of Control Science and Engineering, China University of Petroleum (East China), Qingdao 266580, China.
| |
Collapse
|
31
|
Xu Y, Tan Y, Ma X, Jin X, Tian Y, Li M. Photodynamic Therapy with Tumor Cell Discrimination through RNA-Targeting Ability of Photosensitizer. Molecules 2021; 26:5990. [PMID: 34641533 PMCID: PMC8512109 DOI: 10.3390/molecules26195990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/19/2022] Open
Abstract
Photodynamic therapy (PDT) represents an effective treatment to cure cancer. The targeting ability of the photosensitizer is of utmost importance. Photosensitizers that discriminate cancer cells can avoid the killing of normal cells and improve PDT efficacy. However, the design and synthesis of photosensitizers conjugated with a recognition unit of cancer cell markers is complex and may not effectively target cancer. Considering that the total RNA content in cancer cells is commonly higher than in normal cells, this study has developed the photosensitizer QICY with RNA-targeting abilities for the discrimination of cancer cells. QICY was specifically located in cancer cells rather than normal cells due to their stronger electrostatic interactions with RNA, thereby further improving the PDT effects on the cancer cells. After intravenous injection into mice bearing a xenograft tumor, QICY accumulated into the tumor location through the enhanced permeability and retention effect, automatically targeted cancer cells under the control of RNA, and inhibited tumor growth under 630 nm laser irradiation without obvious side effects. This intelligent photosensitizer with RNA-targeting ability not only simplifies the design and synthesis of cancer-cell-targeting photosensitizers but also paves the way for the further development of highly efficient PDTs.
Collapse
Affiliation(s)
- Yuan Xu
- College of Marine Technology and Environment, Dalian Ocean University, Dalian 116023, China;
- Key Laboratory of Environment Controlled Aquaculture, Ministry of Education, Dalian 116023, China
| | - Yang Tan
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116023, China; (Y.T.); (X.M.); (X.J.)
| | - Xiuqin Ma
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116023, China; (Y.T.); (X.M.); (X.J.)
| | - Xiaoyi Jin
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116023, China; (Y.T.); (X.M.); (X.J.)
| | - Ye Tian
- College of Marine Technology and Environment, Dalian Ocean University, Dalian 116023, China;
- Key Laboratory of Environment Controlled Aquaculture, Ministry of Education, Dalian 116023, China
| | - Miao Li
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116023, China; (Y.T.); (X.M.); (X.J.)
| |
Collapse
|
32
|
Moldogazieva NT, Zavadskiy SP, Terentiev AA. Genomic Landscape of Liquid Biopsy for Hepatocellular Carcinoma Personalized Medicine. Cancer Genomics Proteomics 2021; 18:369-383. [PMID: 33994362 DOI: 10.21873/cgp.20266] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most frequently diagnosed cancer and the third leading cause of cancer-related deaths worldwide. Advanced-stage HCC patients have poor survival rates and this requires the discovery of novel clear biomarkers for HCC early diagnosis and prognosis, identifying risk factors, distinguishing HCC from non-HCC liver diseases, and assessment of treatment response. Liquid biopsy has emerged as a novel minimally invasive approach to enable monitoring tumor progression, metastasis, and recurrence. Since the liquid biopsy analysis has relatively high specificity and low sensitivity in cancer early detection, there is a risk of bias. Next-generation sequencing (NGS) technologies provide accurate and comprehensive gene expression and mutational profiling of liquid biopsies including cell-free circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and genomic components of extracellular vesicles (EVs) including micro-RNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs). Since HCC is a highly heterogeneous cancer, HCC patients can display various genomic, epigenomic, and transcriptomic patterns and exhibit varying sensitivity to treatment options. Identification of individual variabilities in genomic signatures in liquid biopsy has the potential to greatly enhance precision oncology capabilities. In this review, we highlight and critically discuss the latest progress in characterizing the genomic landscape of liquid biopsy, which can advance HCC personalized medicine.
Collapse
Affiliation(s)
- Nurbubu T Moldogazieva
- Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia;
| | - Sergey P Zavadskiy
- A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Alexander A Terentiev
- Department of Biochemistry and Molecular Biology, N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
33
|
Hou M, Yin X, Jiang J, He J. DNAzyme-Triggered Sol-Gel-Sol Transition of a Hydrogel Allows Target Cell Enrichment. ACS APPLIED MATERIALS & INTERFACES 2021; 13:15031-15039. [PMID: 33764744 DOI: 10.1021/acsami.1c02262] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Enrichment of rare cancer cells from various cell mixtures for subsequent analysis or culture is essential for understanding cancer formation and progression. In particular, maintaining the viability of captured cancer cells and gently releasing them for relevant applications remain challenging for many reported methods. Here, a physically cross-linked deoxyribozyme (DNAzyme)-based hydrogel strategy was developed for the specific envelopment and release of targeted cancer cells, allowing the aptamer-guided capture, 3D envelopment, and Zn2+-dependent release of viable cancer cells. The DNAzyme hydrogel is constructed through the intertwinement and hybridization of two complementary DNAzyme strands located on two rolling circle amplification-synthesized ultralong DNA chains. The enveloping and separation of target cells were achieved during the formation of the DNAzyme hydrogel (sol-gel transition). Triggered by Zn2+, the encapsulated cells can be gently released from the dissociated DNAzyme hydrogel with high viability (gel-sol transition). Successful isolations of target cells from cancer cell mixtures and peripheral blood mononuclear cells (PBMC) were demonstrated. This method offers an attractive approach for the separation of target cancer cells for various downstream applications that require viable cells.
Collapse
Affiliation(s)
- Min Hou
- College of Biology, Hunan University, Changsha 410082, China
| | - Xiang Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jianhui Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jianjun He
- College of Biology, Hunan University, Changsha 410082, China
| |
Collapse
|