1
|
Zhang Z, Liu S, Yao W, Yang X, Que Z, Luo B, Wang Y, Sun M, Yang Y, Tian J. Maimendong decoction inhibits lung cancer metastasis by increasing the proportion and killing activity of NK cells. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119127. [PMID: 39566862 DOI: 10.1016/j.jep.2024.119127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/05/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In China, Maimendong Decoction (MMDD) is a classic Chinese medicine prescription for treating lung diseases, such as cough, upper respiratory tract infection, bronchitis, asthma, pulmonary fibrosis and so on. However, the mechanism by which MMDD inhibits lung cancer metastasis remains unclear. AIM OF THE STUDY This study seeks to examine the effect of MMDD on suppressing lung cancer metastasis and to uncover its mechanism through the regulation of tumor immunity. MATERIALS AND METHODS The impact of MMDD on lung cancer cell growth and metastasis was assessed via cell apoptosis, proliferation, and migration assays, alongside lung metastasis models in mice. Hematoxylin-eosin (H&E) staining, immunohistochemistry, and immunofluorescence were used to examine the tissue morphology of metastatic tumors, Ki-67 protein expression, and NK cell infiltration. The potential anti-lung cancer mechanism of MMDD was explored through network pharmacology. Immune cell levels and NK cell function in mouse peripheral blood were analyzed using flow cytometry. Additionally, co-culture and lactate dehydrogenase (LDH) assays were performed to assess NK cell cytotoxicity. RESULTS MMDD significantly reduced the number of metastatic tumors and the neoplastic burden in the lungs of mice. While MMDD did not affect the proliferation, apoptosis, or migration of lung cancer cells at 1 mg/ml concentration in vitro, it substantially increased NK cell proportion in peripheral blood and metastatic tumor tissues. Moreover, MMDD significantly boosted NK cell cytotoxicity and enhanced their killing effect on lung cancer cells. Importantly, depletion of NK cells abolished the survival-prolonging effect of MMDD in mice. CONCLUSION This study demonstrates that MMDD restrains lung cancer cell metastasis primarily by increasing NK cell levels and enhancing their cytotoxic activity. These results offer experimental support for the use of MMDD in treating lung cancer metastasis.
Collapse
Affiliation(s)
- Zhipeng Zhang
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Shihui Liu
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China; Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Wang Yao
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Xiaoyu Yang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Zujun Que
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Bin Luo
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Yanhong Wang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Mingyu Sun
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yun Yang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Jianhui Tian
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China; Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
2
|
Zhou Z, Cai S, Zhou X, Zhao W, Sun J, Zhou Z, Yang Z, Li W, Wang Z, Zou H, Fu H, Wang X, Khoo BL, Yang M. Circulating Tumor Cells Culture: Methods, Challenges, and Clinical Applications. SMALL METHODS 2024:e2401026. [PMID: 39726345 DOI: 10.1002/smtd.202401026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/10/2024] [Indexed: 12/28/2024]
Abstract
Circulating tumor cells (CTCs) play a pivotal role in cancer metastasis and hold considerable potential for clinical diagnosis, therapeutic monitoring, and prognostic evaluation. Nevertheless, the limited quantity of CTCs in liquid biopsy samples poses challenges for comprehensive downstream analysis. In vitro culture of CTCs can effectively address the issue of insufficient CTC numbers. Furthermore, research based on CTC cell lines serves as a valuable complement to traditional cancer cell line-based research. While numerous reports exist on CTC in vitro culture and even the establishment of CTC cell lines, the methods used vary, leading to disparate culture outcomes. This review presents the developmental history and current status of CTC in vitro culture research. Additionally, the culture strategies applied in different methods and analyzed the impact of various steps on culture outcomes are compared. Overall, the review indicates that while the short-term culture of CTCs is relatively straightforward, long-term culture success has been achieved for various specific cancer types but still faces challenges. Further optimization of efficient and widely applicable culture strategies is needed. Additionally, ongoing applications of CTC in vitro culture are summarized, highlighting the potential of expanded CTCs for drug susceptibility testing and as therapeutic tools in personalized treatment.
Collapse
Affiliation(s)
- Zhengdong Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Songhua Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Wei Zhao
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Jiayu Sun
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zhihang Zhou
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zihan Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Wenxiu Li
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zhe Wang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Heng Zou
- Cellomics (Shenzhen) Limited, Shenzhen, 518118, China
| | - Huayang Fu
- Cellomics (Shenzhen) Limited, Shenzhen, 518118, China
| | - Xicheng Wang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Bee Luan Khoo
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| |
Collapse
|
3
|
Smit DJ, Schneegans S, Pantel K. Clinical applications of circulating tumor cells in patients with solid tumors. Clin Exp Metastasis 2024; 41:403-411. [PMID: 38281256 PMCID: PMC11374849 DOI: 10.1007/s10585-024-10267-5] [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: 08/22/2023] [Accepted: 01/11/2024] [Indexed: 01/30/2024]
Abstract
The concept of liquid biopsy analysis has been established more than a decade ago. Since the establishment of the term, tremendous advances have been achieved and plenty of methods as well as analytes have been investigated in basic research as well in clinical trials. Liquid biopsy refers to a body fluid-based biopsy that is minimal-invasive, and most importantly, allows dense monitoring of tumor responses by sequential blood sampling. Blood is the most important analyte for liquid biopsy analyses, providing an easily accessible source for a plethora of cells, cell-derived products, free nucleic acids, proteins as well as vesicles. More than 12,000 publications are listed in PubMed as of today including the term liquid biopsy. In this manuscript, we critically review the current implications of liquid biopsy, with special focus on circulating tumor cells, and describe the hurdles that need to be addressed before liquid biopsy can be implemented in clinical standard of care guidelines.
Collapse
Affiliation(s)
- Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Fleur Hiege Center for Skin Cancer Research, Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Svenja Schneegans
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
- Fleur Hiege Center for Skin Cancer Research, Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
4
|
Smit DJ, Pantel K. Circulating tumor cells as liquid biopsy markers in cancer patients. Mol Aspects Med 2024; 96:101258. [PMID: 38387225 DOI: 10.1016/j.mam.2024.101258] [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: 11/14/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Over the past decade, novel methods for enrichment and identification of cancer cells circulating in the blood have been established. Blood-based detection of cancer cells and other tumor-associated products can be summarized under the term of Liquid Biopsy. Circulating tumor cells (CTCs) have been used for diagnosis, risk stratification and treatment selection as well as treatment monitoring in several studies over the past years, thus representing a valuable biomarker for cancer patients. A plethora of methods to enrich, detect and analyze CTCs has been established. In contrast to other liquid biopsy analytes (e.g. ctDNA), CTCs represent a viable analyte that provides a unique opportunity to understand the underlaying biology of cancer and the metastatic cascade on the molecular level. In this review, we provide an overview on the current methods used for enrichment, detection, molecular and functional characterization of CTCs.
Collapse
Affiliation(s)
- Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
5
|
Zhou J, Luo J, Gan R, Zhi L, Zhou H, Lv M, Huang Y, Liang G. SSPH I, A Novel Anti-cancer Saponin, Inhibits EMT and Invasion and Migration of NSCLC by Suppressing MAPK/ERK1/2 and PI3K/AKT/ mTOR Signaling Pathways. Recent Pat Anticancer Drug Discov 2024; 19:543-555. [PMID: 38305308 DOI: 10.2174/0115748928283132240103073039] [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: 10/21/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND Saponin of Schizocapsa plantaginea Hance I (SSPH I).a bioactive saponin found in Schizocapsa plantaginea, exhibits significant anti-proliferation and antimetastasis in lung cancer. OBJECTIVE To explore the anti-metastatic effects of SSPH I on non-small cell lung cancer (NSCLC) with emphasis on epithelial-mesenchymal transition (EMT) both in vitro and in vivo. METHODS The effects of SSPH I at the concentrations of 0, 0.875,1.75, and 3.5 μM on A549 and PC9 lung cancer cells were evaluated using colony formation assay, CCK-8 assay, transwell assay and wound-healing assay. The actin cytoskeleton reorganization of PC9 and A549 cells was detected using the FITC-phalloidin fluorescence staining assay. The proteins related to EMT (N-cadherin, E-cadherin and vimentin), p- PI3K, p- AKT, p- mTOR and p- ERK1/2 were detected by Western blotting. A mouse model of lung cancer metastasis was established by utilizing 95-D cells, and the mice were treated with SSPH I by gavage. RESULTS The results suggested that SSPH I significantly inhibited the migration and invasion of NSCLC cells under a non-cytotoxic concentration. Furthermore, SSPH I at a non-toxic concentration of 0.875 μM inhibited F-actin cytoskeleton organization. Importantly, attenuation of EMT was observed in A549 cells with upregulation in the expression of epithelial cell marker E-cadherin and downregulation of the mesenchymal cell markers vimentin as well as Ncadherin. Mechanistic studies revealed that SSPH I inhibited MAPK/ERK1/2 and PI3K/AKT/mTOR signaling pathways. CONCLUSION SSPH I inhibited EMT, migration, and invasion of NSCLC cells by suppressing MAPK/ERK1/2 and PI3K/AKT/mTOR signaling pathways, suggesting that the natural compound SSPH I could be used for inhibiting metastasis of NSCLC.
Collapse
Affiliation(s)
- Jinling Zhou
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Jian Luo
- Department of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Rizhi Gan
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Limin Zhi
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Huan Zhou
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Meixian Lv
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Yinmei Huang
- College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Gang Liang
- College of Pharmacy, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
6
|
Yang Y, Zhu L, Liu J, Yu P, Que Z, Li Y, Li H, Tian J. Jinfukang inhibits clustering and invasion of circulating lung tumor cells by regulating the EGFR signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1851-1854. [PMID: 37723875 PMCID: PMC10679872 DOI: 10.3724/abbs.2023148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/29/2023] [Indexed: 09/20/2023] Open
Affiliation(s)
- Yun Yang
- Clinical Oncology CenterShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071China
| | - Lihua Zhu
- Department of OncologyLonghua HospitalShanghai University of Traditional Chinese MedicineShanghai200032China
| | - Jiajun Liu
- Institute of OncologyShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071China
| | - Pan Yu
- Clinical Oncology CenterShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071China
| | - Zujun Que
- Institute of OncologyShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071China
| | - Yan Li
- Clinical Oncology CenterShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071China
| | - Hegen Li
- Department of OncologyLonghua HospitalShanghai University of Traditional Chinese MedicineShanghai200032China
| | - Jianhui Tian
- Clinical Oncology CenterShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071China
- Institute of OncologyShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071China
| |
Collapse
|
7
|
Luo B, Que Z, Lu X, Qi D, Qiao Z, Yang Y, Qian F, Jiang Y, Li Y, Ke R, Shen X, Xiao H, Li H, Wu E, Tian J. Identification of exosome protein panels as predictive biomarkers for non-small cell lung cancer. Biol Proced Online 2023; 25:29. [PMID: 37953280 PMCID: PMC10641949 DOI: 10.1186/s12575-023-00223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related deaths worldwide, primarily due to its propensity for metastasis. Patients diagnosed with localized primary cancer have higher survival rates than those with metastasis. Thus, it is imperative to discover biomarkers for the early detection of NSCLC and the timely prediction of tumor metastasis to improve patient outcomes. METHODS Here, we utilized an integrated approach to isolate and characterize plasma exosomes from NSCLC patients as well as healthy individuals. We then conducted proteomics analysis and parallel reaction monitoring to identify and validate the top-ranked proteins of plasma exosomes. RESULTS Our study revealed that the proteome in exosomes from NSCLC patients with metastasis was distinctly different from that from healthy individuals. The former had larger diameters and lower concentrations of exosomes than the latter. Furthermore, among the 1220 identified exosomal proteins, we identified two distinct panels of biomarkers. The first panel of biomarkers (FGB, FGG, and VWF) showed potential for early NSCLC diagnosis and demonstrated a direct correlation with the survival duration of NSCLC patients. The second panel of biomarkers (CFHR5, C9, and MBL2) emerged as potential biomarkers for assessing NSCLC metastasis, of which CFHR5 alone was significantly associated with the overall survival of NSCLC patients. CONCLUSIONS These findings underscore the potential of plasma exosomal biomarkers for early NSCLC diagnosis and metastasis prediction. Notably, CFHR5 stands out as a promising prognostic indicator for NSCLC patients. The clinical utility of exosomal biomarkers offers the potential to enhance the management of NSCLC.
Collapse
Affiliation(s)
- Bin Luo
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zujun Que
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xinyi Lu
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Dan Qi
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76502, USA
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX, 76508, USA
| | - Zhi Qiao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yun Yang
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Fangfang Qian
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yi Jiang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yan Li
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Ronghu Ke
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76502, USA
| | - Xiaoyun Shen
- Prism Genomic Medicine, Sugar Land, TX, 77478, USA
| | - Hua Xiao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Hegen Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX, 76502, USA.
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX, 76508, USA.
- School of Medicine, Texas A&M University, College Station, TX, 77843, USA.
- Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, 77843, USA.
- LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Jianhui Tian
- Clinical Oncology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
8
|
Que Z, Luo B, Yu P, Qi D, Shangguan W, Wang P, Liu J, Li Y, Li H, Ke R, Wu E, Tian J. Polyphyllin VII induces CTC anoikis to inhibit lung cancer metastasis through EGFR pathway regulation. Int J Biol Sci 2023; 19:5204-5217. [PMID: 37928267 PMCID: PMC10620814 DOI: 10.7150/ijbs.83682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/27/2023] [Indexed: 11/07/2023] Open
Abstract
Circulating tumor cells (CTCs) are cells that detach from the primary tumor and enter the bloodstream, playing a crucial role in the metastasis of lung cancer. Unfortunately, there is currently a lack of drugs specifically designed to target CTCs and prevent tumor metastasis. In this study, we present evidence that polyphyllin VII, a potent anticancer compound, effectively inhibits the metastasis of lung cancer by inducing a process called anoikis in CTCs. We observed that polyphyllin VII had significant cytotoxicity and inhibited colony formation, migration, and invasion in both our newly established cell line CTC-TJH-01 and a commercial lung cancer cell line H1975. Furthermore, we found that polyphyllin VII induced anoikis and downregulated the TrkB and EGFR-MEK/ERK signaling pathways. Moreover, activation of TrkB protein did not reverse the inhibitory effect of polyphyllin VII on CTCs, while upregulation of EGFR protein effectively reversed it. Furthermore, our immunodeficient mouse models recapitulated that polyphyllin VII inhibited lung metastasis, which was associated with downregulation of the EGFR protein, and reduced the number of CTCs disseminated into the lungs by inducing anoikis. Together, these results suggest that polyphyllin VII may be a promising compound for the treatment of lung cancer metastasis by targeting CTCs.
Collapse
Affiliation(s)
- Zujun Que
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, 200071, China
- Institute of TCM Oncology, Longhua Hospital, Shanghai University of TCM, Shanghai, 200032, China
| | - Bin Luo
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of TCM, Shanghai, 200071, China
| | - Pan Yu
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of TCM, Shanghai, 200071, China
| | - Dan Qi
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76508, USA
- Department of Neurosurgery, Baylor College of Medicine, Temple, Texas 76508, USA
| | - Wenji Shangguan
- Institute of TCM Oncology, Longhua Hospital, Shanghai University of TCM, Shanghai, 200032, China
- Department of TCM, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Panpan Wang
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of TCM, Shanghai, 200071, China
| | - Jiajun Liu
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, 200071, China
| | - Yan Li
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of TCM, Shanghai, 200071, China
| | - He Li
- Department of TCM, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Ronghu Ke
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76508, USA
| | - Erxi Wu
- Department of Neurosurgery and Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76508, USA
- Department of Neurosurgery, Baylor College of Medicine, Temple, Texas 76508, USA
- School of Medicine, Texas A&M University, College Station, TX 77843, USA
- Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX 77843, USA
- LIVESTRONG Cancer Institutes and Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jianhui Tian
- Institute of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, 200071, China
- Institute of TCM Oncology, Longhua Hospital, Shanghai University of TCM, Shanghai, 200032, China
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of TCM, Shanghai, 200071, China
| |
Collapse
|
9
|
Kahounová Z, Pícková M, Drápela S, Bouchal J, Szczyrbová E, Navrátil J, Souček K. Circulating tumor cell-derived preclinical models: current status and future perspectives. Cell Death Dis 2023; 14:530. [PMID: 37591867 PMCID: PMC10435501 DOI: 10.1038/s41419-023-06059-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023]
Abstract
Despite the advancements made in the diagnosis and treatment of cancer, the stages associated with metastasis remain largely incurable and represent the primary cause of cancer-related deaths. The dissemination of cancer is facilitated by circulating tumor cells (CTCs), which originate from the primary tumor or metastatic sites and enter the bloodstream, subsequently spreading to distant parts of the body. CTCs have garnered significant attention in research due to their accessibility in peripheral blood, despite their low abundance. They are being extensively studied to gain a deeper understanding of the mechanisms underlying cancer dissemination and to identify effective therapeutic strategies for advanced stages of the disease. Therefore, substantial efforts have been directed towards establishing and characterizing relevant experimental models derived from CTCs, aiming to provide relevant tools for research. In this review, we provide an overview of recent progress in the establishment of preclinical CTC-derived models, such as CTC-derived xenografts (CDX) and cell cultures, which show promise for the study of CTCs. We discuss the advantages and limitations of these models and conclude by summarizing the potential future use of CTCs and CTC-derived models in cancer treatment decisions and their utility as precision medicine tools.
Collapse
Affiliation(s)
- Zuzana Kahounová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 00, Brno, Czech Republic
| | - Markéta Pícková
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 00, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 602 00, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00, Brno, Czech Republic
| | - Stanislav Drápela
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 00, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 602 00, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00, Brno, Czech Republic
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital, 779 00, Olomouc, Czech Republic
| | - Eva Szczyrbová
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital, 779 00, Olomouc, Czech Republic
| | - Jiří Navrátil
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 656 53, Brno, Czech Republic
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 00, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, 602 00, Brno, Czech Republic.
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|
10
|
Fernández-Santiago C, López-López R, Piñeiro R. Models to study CTCs and CTC culture methods. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 381:57-98. [PMID: 37739484 DOI: 10.1016/bs.ircmb.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
The vast majority of cancer-related deaths are due to the presence of disseminated disease. Understanding the metastatic process is key to achieving a reduction in cancer mortality. Particularly, there is a need to understand the molecular mechanisms that drive cancer metastasis, which will allow the identification of curative treatments for metastatic cancers. Liquid biopsies have arisen as a minimally invasive approach to gain insights into the biology of metastasis. Circulating tumour cells (CTCs), shed to the circulation from the primary tumour or metastatic lesions, are a key component of liquid biopsy. As metastatic precursors, CTCs hold the potential to unravel the mechanisms involved in metastasis formation as well as new therapeutic strategies for treating metastatic disease. However, the complex biology of CTCs together with their low frequency in circulation are factors hampering an in-depth mechanistic investigation of the metastatic process. To overcome these problems, CTC-derived models, including CTC-derived xenograft (CDX) and CTC-derived ex vivo cultures, in combination with more traditional in vivo models of metastasis, have emerged as powerful tools to investigate the biological features of CTCs facilitating cancer metastasis and uncover new therapeutic opportunities. In this chapter, we provide an up to date view of the diverse models used in different cancers to study the biology of CTCs, and of the methods developed for CTC culture and expansion, in vivo and ex vivo. We also report some of the main challenges and limitations that these models are facing.
Collapse
Affiliation(s)
- Cristóbal Fernández-Santiago
- Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Rafael López-López
- Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; University Clinical Hospital of Santiago de Compostela (CHUS/SERGAS), Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Roberto Piñeiro
- Roche-Chus Joint Unit, Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
11
|
Zhu Z, Hu E, Shen H, Tan J, Zeng S. The functional and clinical roles of liquid biopsy in patient-derived models. J Hematol Oncol 2023; 16:36. [PMID: 37031172 PMCID: PMC10082989 DOI: 10.1186/s13045-023-01433-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/28/2023] [Indexed: 04/10/2023] Open
Abstract
The liquid biopsy includes the detection of circulating tumor cells (CTCs) and CTC clusters in blood, as well as the detection of, cell-free DNA (cfDNA)/circulating tumor DNA (ctDNA) and extracellular vesicles (EVs) in the patient's body fluid. Liquid biopsy has important roles in translational research. But its clinical utility is still under investigation. Newly emerged patient-derived xenograft (PDX) and CTC-derived xenograft (CDX) faithfully recapitulate the genetic and morphological features of the donor patients' tumor and patient-derived organoid (PDO) can mostly mimic tumor growth, tumor microenvironment and its response to drugs. In this review, we describe how the development of these patient-derived models has assisted the studies of CTCs and CTC clusters in terms of tumor biological behavior exploration, genomic analysis, and drug testing, with the help of the latest technology. We then summarize the studies of EVs and cfDNA/ctDNA in PDX and PDO models in early cancer diagnosis, tumor burden monitoring, drug test and response monitoring, and molecular profiling. The challenges faced and future perspectives of research related to liquid biopsy using patient-derived models are also discussed.
Collapse
Affiliation(s)
- Ziqing Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Erya Hu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Jun Tan
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
12
|
Suvilesh KN, Manjunath Y, Pantel K, Kaifi JT. Preclinical models to study patient-derived circulating tumor cells and metastasis. Trends Cancer 2023; 9:355-371. [PMID: 36759267 DOI: 10.1016/j.trecan.2023.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 02/10/2023]
Abstract
Circulating tumor cells (CTCs) that are detached from the tumor can be precursors of metastasis. The majority of studies focus on enumeration of CTCs from patient blood to predict recurrence and therapy outcomes. Very few studies have managed to expand CTCs to investigate their functional dynamics with respect to genetic changes, tumorigenic potential, and response to drug treatment. A growing amount of evidence based on successful CTC expansion has revealed novel therapeutic targets that are associated with the process of metastasis. In this review, we summarize the successes, challenges, and limitations that collectively contribute to the better understanding of metastasis using patient-derived CTCs as blood-borne seeds of metastasis. The roadblocks and future avenues to move CTC-based scientific discoveries forward are also discussed.
Collapse
Affiliation(s)
- Kanve N Suvilesh
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA.
| | - Yariswamy Manjunath
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Klaus Pantel
- Institute for Tumor Biology, University of Hamburg, Hamburg, Germany
| | - Jussuf T Kaifi
- Hugh E. Stephenson Jr., MD, Department of Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO, USA; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA; Siteman Cancer Center, St. Louis, MO, USA.
| |
Collapse
|
13
|
Tretyakova MS, Menyailo ME, Schegoleva AA, Bokova UA, Larionova IV, Denisov EV. Technologies for Viable Circulating Tumor Cell Isolation. Int J Mol Sci 2022; 23:ijms232415979. [PMID: 36555625 PMCID: PMC9788311 DOI: 10.3390/ijms232415979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
The spread of tumor cells throughout the body by traveling through the bloodstream is a critical step in metastasis, which continues to be the main cause of cancer-related death. The detection and analysis of circulating tumor cells (CTCs) is important for understanding the biology of metastasis and the development of antimetastatic therapy. However, the isolation of CTCs is challenging due to their high heterogeneity and low representation in the bloodstream. Different isolation methods have been suggested, but most of them lead to CTC damage. However, viable CTCs are an effective source for developing preclinical models to perform drug screening and model the metastatic cascade. In this review, we summarize the available literature on methods for isolating viable CTCs based on different properties of cells. Particular attention is paid to the importance of in vitro and in vivo models obtained from CTCs. Finally, we emphasize the current limitations in CTC isolation and suggest potential solutions to overcome them.
Collapse
Affiliation(s)
- Maria S. Tretyakova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Maxim E. Menyailo
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Single Cell Biology Laboratory, Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Anastasia A. Schegoleva
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Single Cell Biology Laboratory, Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ustinia A. Bokova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Irina V. Larionova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Evgeny V. Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Single Cell Biology Laboratory, Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
- Correspondence: ; Tel./Fax: +7-3822-282676 (ext. 3375)
| |
Collapse
|
14
|
Wang L, Wang X, Guo E, Mao X, Miao S. Emerging roles of platelets in cancer biology and their potential as therapeutic targets. Front Oncol 2022; 12:939089. [PMID: 35936717 PMCID: PMC9355257 DOI: 10.3389/fonc.2022.939089] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022] Open
Abstract
The main role of platelets is to control bleeding and repair vascular damage via thrombosis. They have also been implicated to promote tumor metastasis through platelet-tumor cell interactions. Platelet-tumor cell interactions promote tumor cell survival and dissemination in blood circulation. Tumor cells are known to induce platelet activation and alter platelet RNA profiles. Liquid biopsies based on tumor-educated platelet biomarkers can detect tumors and correlate with prognosis, personalized therapy, treatment monitoring, and recurrence prediction. Platelet-based strategies for cancer prevention and tumor-targeted therapy include developing drugs that target platelet receptors, interfere with the release of platelet particles, inhibit platelet-specific enzymes, and utilize platelet-derived “nano-platelets” as a targeted drug delivery platform for tumor therapy. This review elaborates on platelet-tumor cell interactions and the molecular mechanisms and discusses future research directions for platelet-based liquid biopsy techniques and platelet-targeted anti-tumor strategies.
Collapse
Affiliation(s)
- Lei Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Erliang Guo
- Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xionghui Mao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Xionghui Mao, ; Susheng Miao,
| | - Susheng Miao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Xionghui Mao, ; Susheng Miao,
| |
Collapse
|
15
|
Functional analysis of circulating tumour cells: the KEY to understand the biology of the metastatic cascade. Br J Cancer 2022; 127:800-810. [PMID: 35484215 PMCID: PMC9427839 DOI: 10.1038/s41416-022-01819-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 02/06/2023] Open
Abstract
Metastasis formation is the main cause of cancer-related death in patients with solid tumours. At the beginning of this process, cancer cells escape from the primary tumour to the blood circulation where they become circulating tumour cells (CTCs). Only a small subgroup of CTCs will survive during the harsh journey in the blood and colonise distant sites. The in-depth analysis of these metastasis-competent CTCs is very challenging because of their extremely low concentration in peripheral blood. So far, only few groups managed to expand in vitro and in vivo CTCs to be used as models for large-scale descriptive and functional analyses of CTCs. These models have shown already the high variability and complexity of the metastatic cascade in patients with cancer, and open a new avenue for the development of new diagnostic and therapeutic approaches.
Collapse
|
16
|
Cell Lines of Circulating Tumor Cells: What Is Known and What Needs to Be Resolved. J Pers Med 2022; 12:jpm12050666. [PMID: 35629089 PMCID: PMC9148030 DOI: 10.3390/jpm12050666] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 12/03/2022] Open
Abstract
The importance of circulating tumor cells (CTC) is well recognized. However, the biological characteristics of CTC in the bloodstream have not yet been examined in detail, due to the limited number of CTC cell lines currently available. Thirty-nine CTC cell lines were reported by 2021. For successful cell culturing, these CTC cell lines were reviewed. Previous studies on short-term cultures of CTC also analyzed approaches for establishing the long-term culture of CTC. Negative selection, hypoxic conditions, three-dimensional conditions, and careful management are preferable for the long-term culture of CTC. However, the establishment of CTC cell lines is dependent on the specific characteristics of each cell type. Therefore, a method to establish CTC cell lines has not yet been developed. Further efforts are needed to resolve this issue.
Collapse
|
17
|
Kapeleris J, Ebrahimi Warkiani M, Kulasinghe A, Vela I, Kenny L, Ladwa R, O’Byrne K, Punyadeera C. Clinical Applications of Circulating Tumour Cells and Circulating Tumour DNA in Non-Small Cell Lung Cancer-An Update. Front Oncol 2022; 12:859152. [PMID: 35372000 PMCID: PMC8965052 DOI: 10.3389/fonc.2022.859152] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/14/2022] [Indexed: 12/14/2022] Open
Abstract
Despite efforts to improve earlier diagnosis of non-small cell lung cancer (NSCLC), most patients present with advanced stage disease, which is often associated with poor survival outcomes with only 15% surviving for 5 years from their diagnosis. Tumour tissue biopsy is the current mainstream for cancer diagnosis and prognosis in many parts of the world. However, due to tumour heterogeneity and accessibility issues, liquid biopsy is emerging as a game changer for both cancer diagnosis and prognosis. Liquid biopsy is the analysis of tumour-derived biomarkers in body fluids, which has remarkable advantages over the use of traditional tumour biopsy. Circulating tumour cells (CTCs) and circulating tumour DNA (ctDNA) are two main derivatives of liquid biopsy. CTC enumeration and molecular analysis enable monitoring of cancer progression, recurrence, and treatment response earlier than traditional biopsy through a minimally invasive liquid biopsy approach. CTC-derived ex-vivo cultures are essential to understanding CTC biology and their role in metastasis, provide a means for personalized drug testing, and guide treatment selection. Just like CTCs, ctDNA provides opportunity for screening, monitoring, treatment evaluation, and disease surveillance. We present an updated review highlighting the prognostic and therapeutic significance of CTCs and ctDNA in NSCLC.
Collapse
Affiliation(s)
- Joanna Kapeleris
- Saliva and Liquid Biopsy Translational Laboratory, The Centre for Biomedical Technologies, The School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, QLD, Australia
- Translational Research Institute, Brisbane, QLD, Australia
| | | | - Arutha Kulasinghe
- Translational Research Institute, Brisbane, QLD, Australia
- The School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ian Vela
- The School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- Australian Prostate Cancer Research Centre, Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, QLD, Australia
- Department of Urology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Liz Kenny
- School of Medicine, University of Queensland, Royal Brisbane and Women’s Hospital, Central Integrated Regional Cancer Service, Queensland Health, Brisbane, QLD, Australia
| | - Rahul Ladwa
- Department of Medical Oncology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
- School of Medicine, University of Queensland, Herston, QLD, Australia
| | - Kenneth O’Byrne
- Translational Research Institute, Brisbane, QLD, Australia
- Department of Medical Oncology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Laboratory, The Centre for Biomedical Technologies, The School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Kelvin Grove, QLD, Australia
- Translational Research Institute, Brisbane, QLD, Australia
- Saliva and Liquid Biopsy Translational Laboratory, Griffith Institute for Drug Discovery and Menzies Health Institute Queensland, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
18
|
The potential of liquid biopsy in the management of cancer patients. Semin Cancer Biol 2022; 84:69-79. [PMID: 35331850 DOI: 10.1016/j.semcancer.2022.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 03/06/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
|
19
|
Rupp B, Ball H, Wuchu F, Nagrath D, Nagrath S. Circulating tumor cells in precision medicine: challenges and opportunities. Trends Pharmacol Sci 2022; 43:378-391. [DOI: 10.1016/j.tips.2022.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022]
|
20
|
Espejo-Cruz ML, González-Rubio S, Zamora-Olaya J, Amado-Torres V, Alejandre R, Sánchez-Frías M, Ciria R, De la Mata M, Rodríguez-Perálvarez M, Ferrín G. Circulating Tumor Cells in Hepatocellular Carcinoma: A Comprehensive Review and Critical Appraisal. Int J Mol Sci 2021; 22:ijms222313073. [PMID: 34884878 PMCID: PMC8657934 DOI: 10.3390/ijms222313073] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/27/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common neoplasm and a major cause of cancer-related death worldwide. There is no ideal biomarker allowing early diagnosis of HCC and tumor surveillance in patients receiving therapy. Liquid biopsy, and particularly circulating tumor cells (CTCs), have emerged as a useful tool for diagnosis and monitoring therapeutic responses in different tumors. In the present manuscript, we evaluate the current evidence supporting the quantitative and qualitative assessment of CTCs as potential biomarkers of HCC, as well as technical aspects related to isolation, identification, and classification of CTCs. Although the dynamic assessment of CTCs in patients with HCC may aid the decision-making process, there are still many uncertainties and technical caveats to be solved before this methodology has a true impact on clinical practice guidelines. More studies are needed to identify the optimal combination of surface markers, to increase the efficiency of ex-vivo expansion of CTCs, or even to target CTCs as a potential therapeutic strategy to prevent HCC recurrence after surgery or to hamper tumor progression and extrahepatic spreading.
Collapse
Affiliation(s)
- María Lola Espejo-Cruz
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
| | - Sandra González-Rubio
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
| | - Javier Zamora-Olaya
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
| | - Víctor Amado-Torres
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
| | - Rafael Alejandre
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
| | - Marina Sánchez-Frías
- Department of Pathology, Reina Sofía University Hospital, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain;
| | - Rubén Ciria
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
- Department of Hepatobiliary Surgery and Liver Transplantation, Reina Sofía University Hospital, 14004 Córdoba, Spain
| | - Manuel De la Mata
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Manuel Rodríguez-Perálvarez
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
- Correspondence:
| | - Gustavo Ferrín
- Maimónides Institute of Biomedical Research (IMIBIC), University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain; (M.L.E.-C.); (S.G.-R.); (J.Z.-O.); (V.A.-T.); (R.A.); (R.C.); (M.D.l.M.); (G.F.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| |
Collapse
|
21
|
Maeda Y, Yoshino T, Kogiso A, Negishi R, Takabayashi T, Tago H, Lim TK, Harada M, Matsunaga T, Tanaka T. Lensless imaging-based discrimination between tumour cells and blood cells towards circulating tumour cell cultivation. Analyst 2021; 146:7327-7335. [PMID: 34766603 DOI: 10.1039/d1an01414e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Circulating tumour cells (CTCs) are recognized as important markers for cancer research. Nonetheless, the extreme rarity of CTCs in blood samples limits their availability for multiple characterization. The cultivation of CTCs is still technically challenging due to the lack of information of CTC proliferation, and it is difficult for conventional microscopy to monitor CTC cultivation owing to low throughput. In addition, for precise monitoring, CTCs need to be distinguished from the blood cells which co-exist with CTCs. Lensless imaging is an emerging technique to visualize micro-objects over a wide field of view, and has been applied for various cytometry analyses including blood tests. However, discrimination between tumour cells and blood cells was not well studied. In this study, we evaluated the potential of the lensless imaging system as a tool for monitoring CTC cultivation. Cell division of model tumour cells was examined using the lensless imaging system composed of a simple setup. Subsequently, we confirmed that tumour cells, JM cells (model lymphocytes), and erythrocytes exhibited cell line-specific patterns on the lensless images. After several discriminative parameters were extracted, discrimination between the tumour cells and other blood cells was demonstrated based on linear discriminant analysis. We also combined the highly efficient CTC recovery device, termed microcavity array, with the lensless-imaging to demonstrate recovery, monitoring and discrimination of the tumour cells spiked into whole blood samples. This study indicates that lensless imaging can be a powerful tool to investigate CTC proliferation and cultivation.
Collapse
Affiliation(s)
- Yoshiaki Maeda
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| | - Tomoko Yoshino
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| | - Atsushi Kogiso
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| | - Ryo Negishi
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| | - Tomohiro Takabayashi
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| | - Hikaru Tago
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| | - Tae-Kyu Lim
- Malcom Co., Ltd, 4-15-10, Honmachi, Shibuya-ku, Tokyo, 151-0071, Japan
| | - Manabu Harada
- Malcom Co., Ltd, 4-15-10, Honmachi, Shibuya-ku, Tokyo, 151-0071, Japan
| | - Tadashi Matsunaga
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan. .,Japan Agency for Marine-Earth Science and Technology (JAMSTEC), 2-15, Natsushima-cho, Yokosuka, Kanagawa, 237-0061, Japan
| | - Tsuyoshi Tanaka
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo, 184-8588, Japan.
| |
Collapse
|
22
|
Patel DA, Blay J. Seeding metastases: The role and clinical utility of circulating tumour cells. Tumour Biol 2021; 43:285-306. [PMID: 34690152 DOI: 10.3233/tub-210001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Peripheral human blood is a readily-accessible source of patient material in which circulating tumour cells (CTCs) can be found. Their isolation and characterization holds the potential to provide prognostic value for various solid cancers. Enumeration of CTCs from blood is becoming a common practice in informing prognosis and may guide therapy decisions. It is further recognized that enumeration alone does not capture perspective on the heterogeneity of tumours and varying functional abilities of the CTCs to interact with the secondary microenvironment. Characterizing the isolated CTCs further, in particular assessing their functional abilities, can track molecular changes in the disease progress. As a step towards identifying a suite of functional features of CTCs that could aid in clinical decisions, developing a CTC isolation technique based on extracellular matrix (ECM) interactions may provide a more solid foundation for isolating the cells of interest. Techniques based on size, charge, density, and single biomarkers are not sufficient as they underutilize other characteristics of cancer cells. The ability of cancer cells to interact with ECM proteins presents an opportunity to utilize their full character in capturing, and also allows assessment of the features that reveal how cells might behave at secondary sites during metastasis. This article will review some common techniques and recent advances in CTC capture technologies. It will further explore the heterogeneity of the CTC population, challenges they experience in their metastatic journey, and the advantages of utilizing an ECM-based platform for CTC capture. Lastly, we will discuss how tailored ECM approaches may present an optimal platform to capture an influential heterogeneous population of CTCs.
Collapse
Affiliation(s)
- Deep A Patel
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Jonathan Blay
- School of Pharmacy, University of Waterloo, Waterloo, ON, Canada.,Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
23
|
Yao W, Yao J, Qian F, Que Z, Yu P, Luo T, Zheng D, Zhang Z, Tian J. Paclitaxel-loaded and folic acid-modified PLGA nanomedicine with glutathione response for the treatment of lung cancer. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1027-1036. [PMID: 34109980 DOI: 10.1093/abbs/gmab073] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Indexed: 12/19/2022] Open
Abstract
Targeted delivery and smart response of nanomedicine hold great promise for improving the therapeutic efficacy and alleviating the side effects of chemotherapy agents in cancer treatment. However, availability of only a few studies that discuss organic nanomedicines with these properties limits the development prospects of nanomedicines. In the present study, folic acid (FA)-targeted delivery and glutathione (GSH) smart responsive nanomedicine were rationally designed for paclitaxel (PTX) delivery for the treatment of lung cancer. Compared with other stimuli-responsive nanomedicines, this nanocarrier was not only sensitive to biologically relevant GSH for on-demand drug release but also biodegradable into biocompatible products after fulfilling its delivery task. The nanomedicine first entered tumor cells via FA and its receptor-mediated endocytosis. After the lysosomal escape, poly(lactic-co-glycolic acid) (PLGA) nanomedicine was triggered by a higher level of GSH and released its cargo into the tumor microenvironment. In vitro and in vivo results revealed that the PLGA nanomedicine not only inhibited the proliferation and promoted the apoptosis of lung cancer cells significantly but also possessed less toxic side effects when compared with free PTX. Therefore, the proposed drug delivery system demonstrates the potential of a multifunctional nano-platform to enhance bioavailability and reduce the side effects of chemotherapy agents.
Collapse
Affiliation(s)
- Wang Yao
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jialiang Yao
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Fangfang Qian
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zujun Que
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Pan Yu
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Tianle Luo
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Dan Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhanxia Zhang
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jianhui Tian
- Oncology Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Research Center for Cancer, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| |
Collapse
|
24
|
Que ZJ, Yao JL, Zhou ZY, Yu P, Luo B, Li HG, Liu JX, Xu HX, Tian JH. Jinfukang inhibits lung cancer metastasis by upregulating CX3CL1 to recruit NK cells to kill CTCs. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114175. [PMID: 33933571 DOI: 10.1016/j.jep.2021.114175] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/07/2021] [Accepted: 04/26/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Circulating tumor cells (CTCs) play an important role in tumor metastasis and may be a target for metastasis prevention. The traditional Chinese medicine Jinfukang functions to improve immunity, prevent metastasis, and prolong lung cancer patient survival periods. Yet, whether Jinfukang prevents metastasis by regulating immune cells to clearance CTCs is still unknown. AIM OF THE STUDY To explore the anti-metastasis mechanism of Jinfukang from the perspective of regulating NK cells to clear CTCs. MATERIALS AND METHODS CTC-TJH-01 cell was treated with Jinfukang. Cytokine chip was used to detect cytokines in cell culture supernatant. Lymphocyte recruitment assay was detected by Transwell and flow cytometry. Protein expression was analysis by Western blot. LDH kit was used to detect cytotoxicity. NOD-SCID mice used for tail vein injection to study lung metastasis. RESULTS Jinfukang could promote the expression and secretion of the chemokine CX3CL1 by CTCs. In addition, Jinfukang could promote the recruitment of natural killer (NK) cells by CTCs and significantly increase the cytotoxic effect of NK cells on CTCs. Moreover, Jinfukang could upregulate the expression of FasL and promote the secretion of TNF-α by NK cells and that NK cells could induce the apoptosis of CTCs through the Fas/FasL signaling pathway. Finally, we confirmed that Jinfukang could promote NK cells to kill CTCs and then inhibit lung cancer metastasis in vivo. The above effects of Jinfukang could be partially reversed by an anti-CX3CL1 mAb. CONCLUSIONS These results suggest that Jinfukang may prevent lung cancer metastasis by enhancing the clearance of CTCs in the peripheral blood by NK cells, providing evidence for the anti-metastasis effect of Jinfukang.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Apoptosis/drug effects
- Cell Line, Tumor
- Chemokine CX3CL1/antagonists & inhibitors
- Chemokine CX3CL1/genetics
- Chemokine CX3CL1/metabolism
- Disease Models, Animal
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- GPI-Linked Proteins/metabolism
- Humans
- Intercellular Signaling Peptides and Proteins/metabolism
- Intracellular Signaling Peptides and Proteins/metabolism
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lung Neoplasms/complications
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Male
- Mice, Inbred NOD
- Mice, SCID
- Neoplasm Metastasis/immunology
- Neoplasm Metastasis/prevention & control
- Neoplastic Cells, Circulating/drug effects
- Neoplastic Cells, Circulating/immunology
- Neoplastic Cells, Circulating/pathology
- Receptors, Death Domain/metabolism
- Signal Transduction/drug effects
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Tumor Necrosis Factor-alpha/metabolism
- Up-Regulation/drug effects
- fas Receptor/metabolism
- Mice
Collapse
Affiliation(s)
- Zu-Jun Que
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China.
| | - Jia-Liang Yao
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - Zhi-Yi Zhou
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - Pan Yu
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - Bin Luo
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - He-Gen Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - Jia-Xiang Liu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| | - Hong-Xi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China.
| | - Jian-Hui Tian
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China; Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| |
Collapse
|
25
|
Wang J, Li Y, Wang R, Han C, Xu S, You T, Li Y, Xia J, Xu X, Wang D, Tang H, Yang C, Chen X, Peng Z. A Fully Automated and Integrated Microfluidic System for Efficient CTC Detection and Its Application in Hepatocellular Carcinoma Screening and Prognosis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:30174-30186. [PMID: 34142547 DOI: 10.1021/acsami.1c06337] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Analysis of circulating tumor cells (CTCs) is regarded as a useful diagnostic index to monitor tumor development and guide precision medicine. Although the immunoassay is a common strategy for CTC identification and heterogeneity characterization, it is challenged by poor reaction efficiency and laborious manipulations in microdevices, which hinder the sensitivity, throughput, simplification, and applicability. To meet the need for rapid, sensitive, and simple CTC analysis, we developed an efficient CTC detection system by integrating a 3D printed off-chip multisource reagent platform, a bubble retainer, and a single CTC capture microchip, which can achieve CTC capture and identification within 90 min. Compared with traditional CTC identification methods, this system decreases immunostaining time and antibody consumption by 90% and performs the on-chip immunoassay in a fully automated manner. Using this system, CTCs from the peripheral blood of 19 patients with various cancers were captured, detected, and compared with clinical data. The system shows great potential for early screening, real-time monitoring, and precision medicine for hepatocellular carcinoma (HCC). With the advantages of automation, stability, economy, and user-friendly operation, the proposed system is promising for clinical scenarios.
Collapse
Affiliation(s)
- Jie Wang
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Yang Li
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Rui Wang
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Chao Han
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiquan Xu
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Tingting You
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Yuhuan Li
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Junjie Xia
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Xing Xu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Dongmei Wang
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Huamei Tang
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiang Chen
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhihai Peng
- Department of General Surgery, Department of Pathology, Department of Ultrasound, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361101, China
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, School of Medicine, Xiamen University, Xiamen 361101, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20080, China
| |
Collapse
|
26
|
Functional Implications of the Dynamic Regulation of EpCAM during Epithelial-to-Mesenchymal Transition. Biomolecules 2021; 11:biom11070956. [PMID: 34209658 PMCID: PMC8301972 DOI: 10.3390/biom11070956] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is a transmembrane glycoprotein expressed in epithelial tissues. EpCAM forms intercellular, homophilic adhesions, modulates epithelial junctional protein complex formation, and promotes epithelial tissue homeostasis. EpCAM is a target of molecular therapies and plays a prominent role in tumor biology. In this review, we focus on the dynamic regulation of EpCAM expression during epithelial-to-mesenchymal transition (EMT) and the functional implications of EpCAM expression on the regulation of EMT. EpCAM is frequently and highly expressed in epithelial cancers, while silenced in mesenchymal cancers. During EMT, EpCAM expression is downregulated by extracellular signal-regulated kinases (ERK) and EMT transcription factors, as well as by regulated intramembrane proteolysis (RIP). The functional impact of EpCAM expression on tumor biology is frequently dependent on the cancer type and predominant oncogenic signaling pathways, suggesting that the role of EpCAM in tumor biology and EMT is multifunctional. Membrane EpCAM is cleaved in cancers and its intracellular domain (EpICD) is transported into the nucleus and binds β-catenin, FHL2, and LEF1. This stimulates gene transcription that promotes growth, cancer stem cell properties, and EMT. EpCAM is also regulated by epidermal growth factor receptor (EGFR) signaling and the EpCAM ectoderm (EpEX) is an EGFR ligand that affects EMT. EpCAM is expressed on circulating tumor and cancer stem cells undergoing EMT and modulates metastases and cancer treatment responses. Future research exploring EpCAM’s role in EMT may reveal additional therapeutic opportunities.
Collapse
|
27
|
Boix-Montesinos P, Soriano-Teruel PM, Armiñán A, Orzáez M, Vicent MJ. The past, present, and future of breast cancer models for nanomedicine development. Adv Drug Deliv Rev 2021; 173:306-330. [PMID: 33798642 PMCID: PMC8191594 DOI: 10.1016/j.addr.2021.03.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Even given recent advances in nanomedicine development of breast cancer treatment in recent years and promising results in pre-clinical models, cancer nanomedicines often fail at the clinical trial stage. Limitations of conventional in vitro models include the lack of representation of the stromal population, the absence of a three-dimensional (3D) structure, and a poor representation of inter-tumor and intra-tumor heterogeneity. Herein, we review those cell culture strategies that aim to overcome these limitations, including cell co-cultures, advanced 3D cell cultures, patient-derived cells, bioprinting, and microfluidics systems. The in vivo evaluation of nanomedicines must consider critical parameters that include the enhanced permeability and retention effect, the host's immune status, and the site of tumor implantation. Here, we critically discuss the advantages and limitations of current in vivo models and report how the improved selection and application of breast cancer models can improve the clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Paula M Soriano-Teruel
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain; Centro de Investigación Príncipe Felipe, Targeted Therapies on Cancer and Inflammation Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Ana Armiñán
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Mar Orzáez
- Centro de Investigación Príncipe Felipe, Targeted Therapies on Cancer and Inflammation Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Centro de Investigación Príncipe Felipe, Polymer Therapeutics Laboratory, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
28
|
Carmona-Ule N, González-Conde M, Abuín C, Cueva JF, Palacios P, López-López R, Costa C, Dávila-Ibáñez AB. Short-Term Ex Vivo Culture of CTCs from Advance Breast Cancer Patients: Clinical Implications. Cancers (Basel) 2021; 13:cancers13112668. [PMID: 34071445 PMCID: PMC8198105 DOI: 10.3390/cancers13112668] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Circulating tumor cells (CTCs) are responsible for metastasis, they represent tumor biology and have also predictive value for therapy monitoring and prognosis of metastatic breast cancer patients. In the blood, CTCs are found in low frequency and a small percentage of them survive. Therefore, achieving their expansion in vitro will allow performing characterization and functional analysis. In this work, we used growth factors and Nanoemulsions to support CTCs culture. We have seen that the CTCs subpopulation capable of ex vivo expanding presented mesenchymal and stem characteristics and loss of epithelial markers. Besides, CTC culture predicted progression-free survival. Abstract Background: Circulating tumor cells (CTC) have relevance as prognostic markers in breast cancer. However, the functional properties of CTCs or their molecular characterization have not been well-studied. Experimental models indicate that only a few cells can survive in the circulation and eventually metastasize. Thus, it is essential to identify these surviving cells capable of forming such metastases. Methods: We isolated viable CTCs from 50 peripheral blood samples obtained from 35 patients with advanced metastatic breast cancer using RosetteSepTM for ex vivo culture. The CTCs were seeded and monitored on plates under low adherence conditions and with media supplemented with growth factors and Nanoemulsions. Phenotypic analysis was performed by immunofluorescence and gene expression analysis using RT-PCR and CTCs counting by the Cellsearch® system. Results: We found that in 75% of samples the CTC cultures lasted more than 23 days, predicting a shorter Progression-Free Survival in these patients, independently of having ≥5 CTC by Cellsearch®. We also observed that CTCs before and after culture showed a different gene expression profile. Conclusions: the cultivability of CTCs is a predictive factor. Furthermore, the subset of cells capable of growing ex vivo show stem or mesenchymal features and may represent the CTC population with metastatic potential in vivo.
Collapse
Affiliation(s)
- Nuria Carmona-Ule
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (N.C.-U.); (M.G.-C.); (C.A.); (R.L.-L.)
| | - Miriam González-Conde
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (N.C.-U.); (M.G.-C.); (C.A.); (R.L.-L.)
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain; (J.F.C.); (P.P.)
| | - Carmen Abuín
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (N.C.-U.); (M.G.-C.); (C.A.); (R.L.-L.)
| | - Juan F. Cueva
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain; (J.F.C.); (P.P.)
- Translational Medical Oncology Group (Oncomet), Medical Oncology Department, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Patricia Palacios
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain; (J.F.C.); (P.P.)
- Translational Medical Oncology Group (Oncomet), Medical Oncology Department, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Rafael López-López
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (N.C.-U.); (M.G.-C.); (C.A.); (R.L.-L.)
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain; (J.F.C.); (P.P.)
- Translational Medical Oncology Group (Oncomet), Medical Oncology Department, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Clotilde Costa
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (N.C.-U.); (M.G.-C.); (C.A.); (R.L.-L.)
- CIBERONC, Centro de Investigación Biomédica en Red Cáncer, 28029 Madrid, Spain; (J.F.C.); (P.P.)
- Correspondence: (C.C.); (A.B.D.-I.); Tel.: +34-981-955-602 (C.C.)
| | - Ana Belén Dávila-Ibáñez
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain; (N.C.-U.); (M.G.-C.); (C.A.); (R.L.-L.)
- Correspondence: (C.C.); (A.B.D.-I.); Tel.: +34-981-955-602 (C.C.)
| |
Collapse
|
29
|
Kang K, Zhou X, Zhang Y, Zhu N, Li G, Yi Q, Wu Y. Cell-Released Magnetic Vesicles Capturing Metabolic Labeled Rare Circulating Tumor Cells Based on Bioorthogonal Chemistry. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007796. [PMID: 33749110 DOI: 10.1002/smll.202007796] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/24/2021] [Indexed: 06/12/2023]
Abstract
Capture of circulating tumor cells (CTCs) with high efficiency and high purity holds great value for potential clinical applications. Besides the existing problems of contamination from blood cells and plasma proteins, unknown/down-regulated expression of targeting markers (e.g., antigen, receptor, etc.) of CTCs have questioned the reliability and general applicability of current CTCs capture methodologies based on immune/aptamer-affinity. Herein, a cell-engineered strategy is designed to break down such barriers by employing the cell metabolism as the leading force to solve key problems. Generally, through an extracellular vesicle generation way, the cell-released magnetic vesicles inherited parent cellular membrane characteristics are produced, and then functionalized with dibenzoazacyclooctyne to target and isolate the metabolic labeled rare CTCs. This strategy offers good reliability and broader possibilities to capture different types of tumor cells, as proven by the capture efficiency above 84% and 82% for A549 and HepG2 cell lines as well as an extremely low detection limitation of 5 cells. Moreover, it enabled high purity enrichment of CTCs from 1 mL blood samples of tumor-bearing mice, only ≈5-757 white blood cells are non-specific caught, ignoring the potential phenotypic fluctuation associated with the cancer progression.
Collapse
Affiliation(s)
- Ke Kang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xiaoxi Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yujia Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Nanhang Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Guohao Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
30
|
Clinical Relevance of Mesenchymal- and Stem-Associated Phenotypes in Circulating Tumor Cells Isolated from Lung Cancer Patients. Cancers (Basel) 2021; 13:cancers13092158. [PMID: 33947159 PMCID: PMC8124761 DOI: 10.3390/cancers13092158] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/16/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Lung cancer is the most frequent malignancy in the world. Most lung cancer patients are diagnosed at an advanced stage. To make matters worse, the survival of patients is very poor. Circulating tumor cells (CTCs), albeit rare, have been portrayed as essential players in the progression of lung cancer. It is definitely not easy being a CTC. First, they escape from the primary tumor, then they travel in the bloodstream, have to survive really harsh conditions, and finally, they form metastases. The adoption of epithelial-to-mesenchymal transition as well as cancer stem cell features has been suggested to allow CTCs to survive and metastasize. This review will focus on how these features can be used to estimate the prognosis of lung cancer patients. Abstract Lung cancer is the leading cause of cancer-related mortality globally. Among the types of lung cancer, non-small-cell lung cancer (NSCLC) is more common, while small-cell lung cancer (SCLC) is less frequent yet more aggressive. Circulating tumor cells (CTCs), albeit rare, have been portrayed as essential players in the progression of lung cancer. CTCs are considered to adopt an epithelial-to-mesenchymal transition (EMT) phenotype and characteristics of cancer stem cells (CSCs). This EMT (or partial) phenotype affords these cells the ability to escape from the primary tumor, travel into the bloodstream, and survive extremely adverse conditions, before colonizing distant foci. Acquisition of CSC features, such as self-renewal, differentiation, and migratory potential, further reflect CTCs’ invasive potential. CSCs have been identified in lung cancer, and expression of EMT markers has previously been correlated with poor clinical outcomes. Thus far, a vast majority of studies have concentrated on CTC detection and enumeration as a prognostic tools of patients’ survival or for monitoring treatment efficacy. In this review, we highlight EMT and CSC markers in CTCs and focus on the clinical significance of these phenotypes in the progression of both non-small- and small-cell lung cancer.
Collapse
|
31
|
Smit DJ, Pantel K, Jücker M. Circulating tumor cells as a promising target for individualized drug susceptibility tests in cancer therapy. Biochem Pharmacol 2021; 188:114589. [PMID: 33932470 DOI: 10.1016/j.bcp.2021.114589] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023]
Abstract
Circulating tumor cells (CTCs) play a crucial role in metastasis and became an emerging topic in today's cancer research. In addition, the analysis of CTCs in liquid biopsies will be a valuable tool for prognosis prediction and real time therapy monitoring. The characterization of CTCs may open up a new field of treatment strategy to prevent metastasis or maintain a stable disease. In 2013, the first cell cultures of CTCs have been established in vitro. Additionally, functional studies have been successfully performed over the last years. Meanwhile, more than 300 short-term CTC cultures and 42 long-term CTC cultures from a variety of tumor entities have been described. More than 45 inhibitors have already been tested for their efficacy to target CTCs in several studies in vitro as well as in xenograft mouse models in vivo. Here, we summarize the currently available data of these inhibition experiments and their effects in targeting CTCs. The results suggest that CTCs may be useful for individualized drug susceptibility testing.
Collapse
Affiliation(s)
- Daniel J Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
32
|
Ward MP, E Kane L, A Norris L, Mohamed BM, Kelly T, Bates M, Clarke A, Brady N, Martin CM, Brooks RD, Brooks DA, Selemidis S, Hanniffy S, Dixon EP, A O'Toole S, J O'Leary J. Platelets, immune cells and the coagulation cascade; friend or foe of the circulating tumour cell? Mol Cancer 2021; 20:59. [PMID: 33789677 PMCID: PMC8011144 DOI: 10.1186/s12943-021-01347-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer cells that transit from primary tumours into the circulatory system are known as circulating tumour cells (CTCs). These cancer cells have unique phenotypic and genotypic characteristics which allow them to survive within the circulation, subsequently extravasate and metastasise. CTCs have emerged as a useful diagnostic tool using "liquid biopsies" to report on the metastatic potential of cancers. However, CTCs by their nature interact with components of the blood circulatory system on a constant basis, influencing both their physical and morphological characteristics as well as metastatic capabilities. These properties and the associated molecular profile may provide critical diagnostic and prognostic capabilities in the clinic. Platelets interact with CTCs within minutes of their dissemination and are crucial in the formation of the initial metastatic niche. Platelets and coagulation proteins also alter the fate of a CTC by influencing EMT, promoting pro-survival signalling and aiding in evading immune cell destruction. CTCs have the capacity to directly hijack immune cells and utilise them to aid in CTC metastatic seeding processes. The disruption of CTC clusters may also offer a strategy for the treatment of advance staged cancers. Therapeutic disruption of these heterotypical interactions as well as direct CTC targeting hold great promise, especially with the advent of new immunotherapies and personalised medicines. Understanding the molecular role that platelets, immune cells and the coagulation cascade play in CTC biology will allow us to identify and characterise the most clinically relevant CTCs from patients. This will subsequently advance the clinical utility of CTCs in cancer diagnosis/prognosis.
Collapse
Affiliation(s)
- Mark P Ward
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland.
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland.
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland.
| | - Laura E Kane
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Lucy A Norris
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin 8, Ireland
| | - Bashir M Mohamed
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Tanya Kelly
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Mark Bates
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Andres Clarke
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Nathan Brady
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Cara M Martin
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Robert D Brooks
- Cancer Research Institute, University of South Australia, 5001, Adelaide, Australia
| | - Doug A Brooks
- Cancer Research Institute, University of South Australia, 5001, Adelaide, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Victoria, 3083, Bundoora, Australia
| | | | - Eric P Dixon
- BD Technologies and Innovation, Research Triangle Park, NC, USA
| | - Sharon A O'Toole
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin 8, Ireland
| | - John J O'Leary
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
33
|
Hu X, Zang X, Lv Y. Detection of circulating tumor cells: Advances and critical concerns. Oncol Lett 2021; 21:422. [PMID: 33850563 PMCID: PMC8025150 DOI: 10.3892/ol.2021.12683] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Metastasis is the main cause of cancer-related death and the major challenge in cancer treatment. Cancer cells in circulation are termed circulating tumor cells (CTCs). Primary tumor metastasis is likely due to CTCs released into the bloodstream. These CTCs extravasate and form fatal metastases in different organs. Analyses of CTCs are clarifying the biological understanding of metastatic cancers. These data are also helpful to monitor disease progression and to inform the development of personalized cancer treatment-based liquid biopsy. However, CTCs are a rare cell population with 1-10 CTCs per ml and are difficult to isolate from blood. Numerous approaches to detect CTCs have been developed based on the physical and biological properties of the cells. The present review summarizes the progress made in detecting CTCs.
Collapse
Affiliation(s)
- Xiuxiu Hu
- School of Medical Technology, Jiangsu College of Nursing, Huai'an, Jiangsu 22300, P.R. China
| | - Xiaojuan Zang
- Department of Ultrasonography, Huai'an Maternity and Child Health Care Hospital, Huai'an, Jiangsu 223002, P.R. China
| | - Yanguan Lv
- Clinical Medical Laboratory, Huai'an Maternity and Child Health Care Hospital, Huai'an, Jiangsu 223002, P.R. China
| |
Collapse
|
34
|
Zhang W, Wang H, Sun M, Deng X, Wu X, Ma Y, Li M, Shuoa SM, You Q, Miao L. CXCL5/CXCR2 axis in tumor microenvironment as potential diagnostic biomarker and therapeutic target. Cancer Commun (Lond) 2021; 40:69-80. [PMID: 32237072 PMCID: PMC7163794 DOI: 10.1002/cac2.12010] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
The components of the tumor microenvironment (TME) in solid tumors, especially chemokines, are currently attracting much attention from scientists. C-X-C motif chemokine ligand 5 (CXCL5) is one of the important chemokines in TME. Overexpression of CXCL5 is closely related to the survival time, recurrence and metastasis of cancer patients. In TME, CXCL5 binds to its receptors, such as C-X-C motif chemokine receptor 2 (CXCR2), to participate in the recruitment of immune cells and promote angiogenesis, tumor growth, and metastasis. The CXCL5/CXCR2 axis can act as a bridge between tumor cells and host cells in TME. Blocking the transmission of CXCL5/CXCR2 signals can increase the sensitivity and effectiveness of immunotherapy and slow down tumor progression. CXCL5 and CXCR2 are also regarded as biomarkers for predicting prognosis and molecular targets for customizing the treatment. In this review, we summarized the current literature regarding the biological functions and clinical significance of CXCL5/CXCR2 axis in TME. The possibility to use CXCL5 and CXCR2 as potential prognostic biomarkers and therapeutic targets in cancer is also discussed.
Collapse
Affiliation(s)
- Wen Zhang
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Huishan Wang
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Mingyang Sun
- Department of Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Xueting Deng
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Xueru Wu
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Yilan Ma
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Mengjing Li
- Department of Biotherapy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China.,Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Said Maisam Shuoa
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Qiang You
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China.,Department of Biotherapy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China.,Key Laboratory for Aging and Disease, Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| | - Lin Miao
- Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, Jiangsu, P. R. China
| |
Collapse
|
35
|
Que ZJ, Yang Y, Liu HT, Shang-Guan WJ, Yu P, Zhu LH, Li HG, Liu HM, Tian JH. Jinfukang regulates integrin/Src pathway and anoikis mediating circulating lung cancer cells migration. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113473. [PMID: 33068649 DOI: 10.1016/j.jep.2020.113473] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/30/2020] [Accepted: 10/11/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Metastasis is the main cause of death in lung cancer patients. Circulating tumor cells (CTCs) may be an important target of metastasis intervention. Previous studies have shown that Jinfukang could prevent the recurrence and metastasis of lung cancer, and we have established a circulating lung tumor cell line CTC-TJH-01. However, whether Jinfukang inhibition of lung cancer metastasis is related to CTCs is still unknown. AIM OF THE STUDY To further explore the mechanism of Jinfukang in anti-metastasis of lung cancer from the perspective of intervention of CTCs. MATERIALS AND METHODS CTC-TJH-01 and H1975 cells were treated with Jinfukang. Cell viability was detected by CCK8, and the cell apoptosis was detected by flow cytometry. Transwell was used to detected cell migration and invasion. Cell anoikis was detected by anoikis detection kit. Protein expression was analysis by Western blot. RESULTS Jinfukang could inhibit the proliferation, migration and invasion of CTC-TJH-01 and H1975 cells. Besides, Jinfukang could also induce anoikis in CTC-TJH-01 and H1975 cells. Analysis of the mRNA expression profile showed ECM-receptor interaction and focal adhesion were regulated by Jinfukang. Moreover, it was also find that Jinfukang significantly inhibited integrin/Src pathway in CTC-TJH-01 and H1975 cells. When suppress the expression of integrin with ATN-161, it could promote Jinfukang to inhibit migration and induce anoikis in CTC-TJH-01 and H1975 cells. CONCLUSIONS Our results indicate that the migration and invasion of CTCs are inhibited by Jinfukang, and the mechanism may involve the suppression of integrin/Src axis to induce anoikis. These data suggest that Jinfukang exerts anti-metastatic effects in lung cancer may through anoikis.
Collapse
Affiliation(s)
- Zu-Jun Que
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yun Yang
- Department of Oncology, Shanghai Traditional Chinese Medicine-Intergrated Hospital, Shanghai, China.
| | - Hai-Tao Liu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wen-Ji Shang-Guan
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Pan Yu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Li-Hua Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - He-Gen Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Huai-Min Liu
- Department of Integrative Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Province, China.
| | - Jian-Hui Tian
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
36
|
Hynds RE, Frese KK, Pearce DR, Grönroos E, Dive C, Swanton C. Progress towards non-small-cell lung cancer models that represent clinical evolutionary trajectories. Open Biol 2021; 11:200247. [PMID: 33435818 PMCID: PMC7881177 DOI: 10.1098/rsob.200247] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide. Although advances are being made towards earlier detection and the development of impactful targeted therapies and immunotherapies, the 5-year survival of patients with advanced disease is still below 20%. Effective cancer research relies on pre-clinical model systems that accurately reflect the evolutionary course of disease progression and mimic patient responses to therapy. Here, we review pre-clinical models, including genetically engineered mouse models and patient-derived materials, such as cell lines, primary cell cultures, explant cultures and xenografts, that are currently being used to interrogate NSCLC evolution from pre-invasive disease through locally invasive cancer to the metastatic colonization of distant organ sites.
Collapse
Affiliation(s)
- Robert E. Hynds
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Kristopher K. Frese
- Cancer Research UK Lung Cancer Centre of Excellence, University of Manchester, Manchester, UK
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Alderley Park, Macclesfield, UK
| | - David R. Pearce
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, UK
| | - Eva Grönroos
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Caroline Dive
- Cancer Research UK Lung Cancer Centre of Excellence, University of Manchester, Manchester, UK
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Alderley Park, Macclesfield, UK
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
37
|
Guo QR, Zhang LL, Liu JF, Li Z, Li JJ, Zhou WM, Wang H, Li JQ, Liu DY, Yu XY, Zhang JY. Multifunctional microfluidic chip for cancer diagnosis and treatment. Nanotheranostics 2021; 5:73-89. [PMID: 33391976 PMCID: PMC7738943 DOI: 10.7150/ntno.49614] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Microfluidic chip is not a chip in the traditional sense. It is technologies that control fluids at the micro level. As a burgeoning biochip, microfluidic chips integrate multiple disciplines, including physiology, pathology, cell biology, biophysics, engineering mechanics, mechanical design, materials science, and so on. The application of microfluidic chip has shown tremendous promise in the field of cancer therapy in the past three decades. Various types of cell and tissue cultures, including 2D cell culture, 3D cell culture and tissue organoid culture could be performed on microfluidic chips. Patient-derived cancer cells and tissues can be cultured on microfluidic chips in a visible, controllable, and high-throughput manner, which greatly advances the process of personalized medicine. Moreover, the functionality of microfluidic chip is greatly expanding due to the customizable nature. In this review, we introduce its application in developing cancer preclinical models, detecting cancer biomarkers, screening anti-cancer drugs, exploring tumor heterogeneity and producing nano-drugs. We highlight the functions and recent development of microfluidic chip to provide references for advancing cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Qiao-Ru Guo
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Ling-Ling Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Ji-Fang Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Zhen Li
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R.China
| | - Jia-Jun Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Wen-Min Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Hui Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, P.R.China
| | - Jing-Quan Li
- The First Affiliated Hospital, Hainan Medical University, Haikou, P.R.China
| | - Da-Yu Liu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, P.R.China
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China
| | - Jian-Ye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, P.R.China.,The First Affiliated Hospital, Hainan Medical University, Haikou, P.R.China
| |
Collapse
|
38
|
Liu P, Jonkheijm P, Terstappen LWMM, Stevens M. Magnetic Particles for CTC Enrichment. Cancers (Basel) 2020; 12:cancers12123525. [PMID: 33255978 PMCID: PMC7760229 DOI: 10.3390/cancers12123525] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary For the enrichment of very rare cells, such as Circulating Tumor Cells (CTCs), immunomagnetic enrichment is frequently used. For this purpose, magnetic nanoparticles (MNPs) coated with specific antibodies directed against cancer cells are used. In this review, we look at the properties such a particle needs to have in order to be used successfully, and describe the different methods used in the production of such a particle as well as the methods for their separation. Additionally, an overview is given of the antibodies that could potentially be used for this purpose. Abstract Here, we review the characteristics and synthesis of magnetic nanoparticles (MNPs) and place these in the context of their usage in the immunomagnetic enrichment of Circulating Tumor Cells (CTCs). The importance of the different characteristics is explained, the need for a very specific enrichment is emphasized and different (commercial) magnetic separation techniques are shown. As the specificity of an MNP is in a large part dependent on the antibody coated onto the particle, different strategies in the coupling of specific antibodies as well as an overview of the available antibodies is given.
Collapse
Affiliation(s)
- Peng Liu
- Department of Medical Cell BioPhysics, University of Twente, 7522 NB Enschede, The Netherlnds; (P.L.); (L.W.M.M.T.)
- Department of Molecular Nanofabrication, University of Twente, 7522 NB Enschede, The Netherlands;
| | - Pascal Jonkheijm
- Department of Molecular Nanofabrication, University of Twente, 7522 NB Enschede, The Netherlands;
| | - Leon W. M. M. Terstappen
- Department of Medical Cell BioPhysics, University of Twente, 7522 NB Enschede, The Netherlnds; (P.L.); (L.W.M.M.T.)
| | - Michiel Stevens
- Department of Medical Cell BioPhysics, University of Twente, 7522 NB Enschede, The Netherlnds; (P.L.); (L.W.M.M.T.)
- Correspondence: ; Tel.: +31-53-489-4101
| |
Collapse
|
39
|
Smit DJ, Cayrefourcq L, Haider MT, Hinz N, Pantel K, Alix-Panabières C, Jücker M. High Sensitivity of Circulating Tumor Cells Derived from a Colorectal Cancer Patient for Dual Inhibition with AKT and mTOR Inhibitors. Cells 2020; 9:cells9092129. [PMID: 32962206 PMCID: PMC7566012 DOI: 10.3390/cells9092129] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/24/2022] Open
Abstract
Circulating tumor cells (CTCs) are cells shed from the primary tumor into the bloodstream. While many studies on solid tumor cells exist, data on CTCs are scarce. The mortality of cancer is mostly associated with metastasis and recent research identified CTCs as initiators of metastasis. The PI3K/AKT/mTOR signaling pathway is an intracellular pathway that regulates essential functions including protein biosynthesis, cell growth, cell cycle control, survival and migration. Importantly, activating oncogenic mutations and amplifications in this pathway are frequently observed in a wide variety of cancer entities, underlining the significance of this signaling pathway. In this study, we analyzed the functional role of the PI3K/AKT/mTOR signaling pathway in the CTC-MCC-41 line, derived from a patient with metastatic colorectal cancer. One striking finding in our study was the strong sensitivity of this CTC line against AKT inhibition using MK2206 and mTOR inhibition using RAD001 within the nanomolar range. This suggests that therapies targeting AKT and mTOR could have been beneficial for the patient from which the CTC line was isolated. Additionally, a dual targeting approach of AKT/mTOR inside the PI3K/AKT/mTOR signaling pathway in the colorectal CTCs showed synergistic effects in vitro. Depending on the phenotypical behavior of CTC-MCC-41 in cell culture (adherent vs. suspension), we identified altered phosphorylation levels inside the PI3K/AKT/mTOR pathway. We observed a downregulation of the PI3K/AKT/mTOR signaling pathway, but not of the RAS/RAF/MAPK pathway, in CTCs growing in suspension in comparison to adherent CTCs. Our results highlight distinct functions of AKT isoforms in CTC-MCC-41 cells with respect to cell proliferation. Knockdown of AKT1 and AKT2 leads to significantly impaired proliferation of CTC-MCC-41 cells in vitro. Therefore, our data demonstrate that the PI3K/AKT/mTOR signaling pathway plays a key role in the proliferation of CTC-MCC-41.
Collapse
Affiliation(s)
- Daniel J. Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; (D.J.S.); (N.H.)
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Center of Montpellier, 34093 Montpellier, France; (L.C.); (C.A.-P.)
| | - Marie-Therese Haider
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Nico Hinz
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; (D.J.S.); (N.H.)
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany;
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Center of Montpellier, 34093 Montpellier, France; (L.C.); (C.A.-P.)
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; (D.J.S.); (N.H.)
- Correspondence: ; Tel.: +49-(0)-40-7410-56339
| |
Collapse
|
40
|
Eslami-S Z, Cortés-Hernández LE, Alix-Panabières C. Epithelial Cell Adhesion Molecule: An Anchor to Isolate Clinically Relevant Circulating Tumor Cells. Cells 2020; 9:cells9081836. [PMID: 32764280 PMCID: PMC7464831 DOI: 10.3390/cells9081836] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
In the last few decades, the epithelial cell adhesion molecule (EpCAM) has received increased attention as the main membrane marker used in many enrichment technologies to isolate circulating tumor cells (CTCs). Although there has been a great deal of progress in the implementation of EpCAM-based CTC detection technologies in medical settings, several issues continue to limit their clinical utility. The biology of EpCAM and its role are not completely understood but evidence suggests that the expression of this epithelial cell-surface protein is crucial for metastasis-competent CTCs and may not be lost completely during the epithelial-to-mesenchymal transition. In this review, we summarize the most significant advantages and disadvantages of using EpCAM as a marker for CTC enrichment and its potential biological role in the metastatic cascade.
Collapse
|
41
|
Que ZJ, Luo B, Wang CT, Qian FF, Jiang Y, Li Y, Han XH, Li HG, Liu JX, Tian JH. Proteomics analysis of tumor exosomes reveals vital pathways of Jinfukang inhibiting circulating tumor cells metastasis in lung cancer. JOURNAL OF ETHNOPHARMACOLOGY 2020; 256:112802. [PMID: 32240782 DOI: 10.1016/j.jep.2020.112802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jinfukang has long been used for the clinical treatment of lung cancer. Previous studies have shown that Jinfukang can induce the apoptosis of circulating tumor cells by intervening ROS-mediated DNA damage pathway. However, whether Jinfukang can inhibit the metastasis of circulating tumor cells and its mechanism are still unclear. AIM OF THE STUDY To further investigate the mechanism of Jinfukang in anti-metastasis of lung cancer from the perspective of intervention of tumor exosomes. MATERIALS AND METHODS The invadopodia formation was determined with immunofluorescence. Invasion and migration were detected using the Transwell assay. Ultracentrifugation was used to isolate exosomes. Exosomes were characterized by electron microscopy, nanoparticle tracking analysis and immunoblotting, and the protein profile was evaluated by proteomic analysis. The molecular functions, biological processes and signaling pathway enrichment analysis were performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Key differentially expressed proteins were verified by Western blot. RESULTS Jinfukang can inhibit the expression of MMP14, cortactin, Tks5 and the formation of invadopodia of CTC-TJH-01 cells. Furthermore, Jinfukang can significantly inhibit the invasion and migration of CTC-TJH-01 cells. The diameter of exosomes extracted from the CTC-TJH-01 cells treated by Jinfukang was 30-100 nm, and the exosomal markers CD63, CD81 and TSG101 were expressed. We identified 680 deferentially expressed proteins. Gene oncology analysis indicated that exosomes were mostly derived from plasma membrane and mainly involved in protein localization and intracellular signaling. The ingenuity pathway analysis showed that the EGF pathway was significantly inhibited, whereas the GP6 signaling pathway was significantly activated. We also confirmed that Jinfukang inhibited the expression of EGF pathway-related proteins in CTC-TJH-01 cells. Besides, when EGF was used to activate EGF signaling pathway, the inhibition of Jinfukang on CTC cell metastasis was reversed. CONCLUSION Jinfukang inhibits the metastasis of CTC-TJH-01 cells through the EGF pathway.
Collapse
Affiliation(s)
- Zu-Jun Que
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Bin Luo
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Chen-Tong Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Fang-Fang Qian
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Yi Jiang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Yan Li
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Xiang-Hui Han
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - He-Gen Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Jia-Xiang Liu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Jian-Hui Tian
- Institute of Traditional Chinese Medicine Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China; Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
42
|
Genna A, Vanwynsberghe AM, Villard AV, Pottier C, Ancel J, Polette M, Gilles C. EMT-Associated Heterogeneity in Circulating Tumor Cells: Sticky Friends on the Road to Metastasis. Cancers (Basel) 2020; 12:E1632. [PMID: 32575608 PMCID: PMC7352430 DOI: 10.3390/cancers12061632] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transitions (EMTs) generate hybrid phenotypes with an enhanced ability to adapt to diverse microenvironments encountered during the metastatic spread. Accordingly, EMTs play a crucial role in the biology of circulating tumor cells (CTCs) and contribute to their heterogeneity. Here, we review major EMT-driven properties that may help hybrid Epithelial/Mesenchymal CTCs to survive in the bloodstream and accomplish early phases of metastatic colonization. We then discuss how interrogating EMT in CTCs as a companion biomarker could help refine cancer patient management, further supporting the relevance of CTCs in personalized medicine.
Collapse
Affiliation(s)
- Anthony Genna
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Aline M. Vanwynsberghe
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Amélie V. Villard
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Charles Pottier
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
- Department of Medical Oncology, University Hospital of Liège, 4000 Liège, Belgium
| | - Julien Ancel
- CHU (Centre Hopitalier Universitaire) de Reims, Hôpital Maison Blanche, Service de Pneumologie, 51092 Reims, France;
- INSERM, UMR (Unité Mixte de Recherche)-S1250, SFR CAP-SANTE, Université de Reims Champagne-Ardenne, 51097 Reims, France;
| | - Myriam Polette
- INSERM, UMR (Unité Mixte de Recherche)-S1250, SFR CAP-SANTE, Université de Reims Champagne-Ardenne, 51097 Reims, France;
- CHU de Reims, Hôpital Maison Blanche, Laboratoire de Pathologie, 51092 Reims, France
| | - Christine Gilles
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| |
Collapse
|
43
|
Circulating tumor cell as the functional aspect of liquid biopsy to understand the metastatic cascade in solid cancer. Mol Aspects Med 2020; 72:100816. [PMID: 31377345 DOI: 10.1016/j.mam.2019.07.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/19/2022]
|
44
|
Advances in the Characterization of Circulating Tumor Cells in Metastatic Breast Cancer: Single Cell Analyses and Interactions, and Patient-Derived Models for Drug Testing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1220:61-80. [PMID: 32304080 DOI: 10.1007/978-3-030-35805-1_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastasis is the major cause of breast cancer death worldwide. In metastatic breast cancer, circulating tumor cells (CTCs) can be captured from patient blood samples sequentially over time and thereby serve as surrogates to assess the biology of surviving cancer cells that may still persist in solitary or multiple metastatic sites following treatment. CTCs may thus function as potential real-time decision-making guides for selecting appropriate therapies during the course of disease or for the development and testing of new treatments. The heterogeneous nature of CTCs warrants the use of single cell platforms to better inform our understanding of these cancer cells. Current techniques for single cell analyses and techniques for investigating interactions between cancer and immune cells are discussed. In addition, methodologies for growing patient-derived CTCs in vitro or propagating them in vivo to facilitate CTC drug testing are reviewed. We advocate the use of CTCs in appropriate microenvironments to appraise the effectiveness of cancer chemotherapies, immunotherapies, and for the development of new cancer treatments, fundamental to personalizing and improving the clinical management of metastatic breast cancer.
Collapse
|
45
|
Fakiruddin KS, Lim MN, Nordin N, Rosli R, Zakaria Z, Abdullah S. Targeting of CD133+ Cancer Stem Cells by Mesenchymal Stem Cell Expressing TRAIL Reveals a Prospective Role of Apoptotic Gene Regulation in Non-Small Cell Lung Cancer. Cancers (Basel) 2019; 11:cancers11091261. [PMID: 31466290 PMCID: PMC6770521 DOI: 10.3390/cancers11091261] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are emerging as vehicles for anti-tumor cytotherapy; however, investigation on its efficacy to target a specific cancer stem cell (CSC) population in non-small cell lung cancer (NSCLC) is lacking. Using assays to evaluate cell proliferation, apoptosis, and gene expression, we investigated the efficacy of MSCs expressing tumour necrosis factor (TNF)-related apoptosis inducing ligand (MSC-TRAIL) to target and destroy CD133+ (prominin-1 positive) NSCLC-derived CSCs. Characterization of TRAIL death receptor 5 (DR5) revealed that it was highly expressed in the CD133+ CSCs of both H460 and H2170 cell lines. The human MSC-TRAIL generated in the study maintained its multipotent characteristics, and caused significant tumor cell inhibition in NSCLC-derived CSCs in a co-culture. The MSC-TRAIL induced an increase in annexin V expression, an indicator of apoptosis in H460 and H2170 derived CD133+ CSCs. Through investigation of mitochondria membrane potential, we found that MSC-TRAIL was capable of inducing intrinsic apoptosis to the CSCs. Using pathway-specific gene expression profiling, we uncovered candidate genes such as NFKB1, BAG3, MCL1, GADD45A, and HRK in CD133+ CSCs, which, if targeted, might increase the sensitivity of NSCLC to MSC-TRAIL-mediated inhibition. As such, our findings add credibility to the utilization of MSC-TRAIL for the treatment of NSCLC through targeting of CD133+ CSCs.
Collapse
Affiliation(s)
- Kamal Shaik Fakiruddin
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor 43400, Malaysia.
- Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), National Institutes of Health (NIH), Ministry of Health Malaysia, Shah Alam 40170, Malaysia.
| | - Moon Nian Lim
- Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), National Institutes of Health (NIH), Ministry of Health Malaysia, Shah Alam 40170, Malaysia
| | - Norshariza Nordin
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia
| | - Rozita Rosli
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor 43400, Malaysia
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia
| | - Zubaidah Zakaria
- Haematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), National Institutes of Health (NIH), Ministry of Health Malaysia, Shah Alam 40170, Malaysia
| | - Syahril Abdullah
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Selangor 43400, Malaysia
- Medical Genetics Laboratory, Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia
| |
Collapse
|
46
|
Que Z, Zhou Z, Luo B, Dong C, Jiang Y, Li H, Tian J. Jingfukang induces anti-cancer activity through oxidative stress-mediated DNA damage in circulating human lung cancer cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:204. [PMID: 31391058 PMCID: PMC6686466 DOI: 10.1186/s12906-019-2601-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 07/18/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND Metastasis is the main cause of lung cancer death. As a seed of metastasis, circulating tumor cells are an important target for metastasis intervention. The traditional Chinese medicine, Jinfukang, has been clinically available for the treatment of non-small cell lung cancer (NSCLC). In this study, we investigated the action and underlying mechanisms of Jinfukang against circulating lung tumor cells. METHODS The cell counting kit-8 (CCK-8), colony formation and cell cycle assays were used to study the cell proliferation ability. Flow cytometry was used to detect the apoptosis and the expression level of ROS and Caspase-3. Comet and TUNEL assays were used to detect DNA damage. DNA damage related pathway protein was detected by western blot. RESULTS Jinfukang significantly inhibits the proliferation of CTC-TJH-01 cells by inducing G1 phase arrest and inhibits their colony formation in a dose-dependent manner. Moreover, Jinfukang induces apoptosis in CTC-TJH-01 cells through the ROS-mediated ATM/ATR-p53 pathway and DNA damage. CONCLUSIONS Our findings suggest that Jinfukang may be a potential drug for lung cancer metastasis.
Collapse
Affiliation(s)
- Zujun Que
- Oncology Institute of Traditional Chinese Medicine, Shanghai Research Institute of Traditional Chinese Medicine, No. 725, South Wanping Road, Shanghai, 200032 China
| | - Zhiyi Zhou
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No. 725, South Wanping Road, Shanghai, 200032 China
| | - Bin Luo
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No. 725, South Wanping Road, Shanghai, 200032 China
| | - Changsheng Dong
- Oncology Institute of Traditional Chinese Medicine, Shanghai Research Institute of Traditional Chinese Medicine, No. 725, South Wanping Road, Shanghai, 200032 China
| | - Yi Jiang
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No. 725, South Wanping Road, Shanghai, 200032 China
| | - Hegen Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No. 725, South Wanping Road, Shanghai, 200032 China
| | - Jianhui Tian
- Oncology Institute of Traditional Chinese Medicine, Shanghai Research Institute of Traditional Chinese Medicine, No. 725, South Wanping Road, Shanghai, 200032 China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No. 725, South Wanping Road, Shanghai, 200032 China
| |
Collapse
|
47
|
Zheng W, Zhang Y, Guo L, Wang S, Fang M, Mao W, Lou J. Evaluation of therapeutic efficacy with CytoSorter ® circulating tumor cell-capture system in patients with locally advanced head and neck squamous cell carcinoma. Cancer Manag Res 2019; 11:5857-5869. [PMID: 31303792 PMCID: PMC6603285 DOI: 10.2147/cmar.s208409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/21/2019] [Indexed: 01/20/2023] Open
Abstract
Objective: This study aimed to investigate the feasibility of utilizing CytoSorter® system to detect circulating tumor cells (CTCs) and clinical value of CTCs in patients with locally advanced head and neck squamous cell carcinoma (LAHNSCC). Methods: 31 patients with LAHNSCC, 12 healthy volunteers, and 6 patients with benign tumor serving as controls were enrolled in this study. CTCs were enriched with the CytoSorter®, a microfluidic based immune capture system. CTC detection was performed before and after induction chemotherapy, as well as after surgery and/or radiotherapy. Correlations between CTC enumerations at different time points and survival outcome and recurrence risk were evaluated. The correlation between CTCs and clinicopathological characteristics was appraised. Follow-up of patients continued until March 2019. Results: While CTCs were not found in the controls, they were detected in 24 of 31 LAHNSCC patients. CTCs could be used to distinguish diseased people from the healthy (P<0.0001). CTCs were statistically associated with patient age (P=0.037, >60 years old vs<60 years old) and lymph node metastasis (P= 0.034, N0N1 VS N2N3). Most patients had significantly reduced CTCs at the end of treatment. Patients with partial remission of tumor after induction therapy had more CTCs than those with complete remission of tumor. Patients with higher CTCs counts prior to treatment had higher chance of developing local recurrence of tumor after treatment (P=0.0187). Conclusion: CTCs were successfully isolated in LAHNSCC patients using CytoSorter® system with better sensibility. CTCs can be used to differentiate LAHNSCC patients from those with benign HNSCC tumor or healthy volunteers, and as markers to monitor patient’s response to treatment and predict the local tumor recurrence after treatment. CTC detection at baseline has the greatest prognostic potency in LAHNSCC patients.
Collapse
Affiliation(s)
- Weihui Zheng
- Center of Oncology, the First Affiliated Hospital of Soochow University, Suzhou 215000, People's Republic of China.,Department of Head and Neck Surgery, Zhejiang Cancer Hospital & Creative Laboratory of Head and Neck Oncology in Zhejiang Province, Hangzhou 310022, People's Republic of China
| | - Yibiao Zhang
- Department of Clinical Laboratory, Jinhua Guangfu Hospital, Jinhua 321000, People's Republic of China
| | - Liang Guo
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital & Creative Laboratory of Head and Neck Oncology in Zhejiang Province, Hangzhou 310022, People's Republic of China
| | - Shengye Wang
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou 310022, People's Republic of China
| | - Meiyu Fang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, People's Republic of China
| | - Weimin Mao
- Center of Oncology, the First Affiliated Hospital of Soochow University, Suzhou 215000, People's Republic of China
| | - Jianlin Lou
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital & Creative Laboratory of Head and Neck Oncology in Zhejiang Province, Hangzhou 310022, People's Republic of China
| |
Collapse
|
48
|
Ireson CR, Alavijeh MS, Palmer AM, Fowler ER, Jones HJ. The role of mouse tumour models in the discovery and development of anticancer drugs. Br J Cancer 2019; 121:101-108. [PMID: 31231121 PMCID: PMC6738037 DOI: 10.1038/s41416-019-0495-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 05/09/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
Our understanding of cancer biology has increased substantially over the past 30 years. Despite this, and an increasing pharmaceutical company expenditure on research and development, the approval of novel oncology drugs during the past decade continues to be modest. In addition, the attrition of agents during clinical development remains high. This attrition can be attributed, at least in part, to the clinical development being underpinned by the demonstration of predictable efficacy in experimental models of human tumours. This review will focus on the range of models available for the discovery and development of anticancer drugs, from traditional subcutaneous injection of tumour cell lines to mice genetically engineered to spontaneously give rise to tumours. It will consider the best time to use the models, along with practical applications and shortcomings. Finally, and most importantly, it will describe how these models reflect the underlying cancer biology and how well they predict efficacy in the clinic. Developing a line of sight to the clinic early in a drug discovery project provides clear benefit, as it helps to guide the selection of appropriate preclinical models and facilitates the investigation of relevant biomarkers.
Collapse
Affiliation(s)
| | - Mo S Alavijeh
- Pharmidex Pharmaceutical Services, 14 Hanover Street, London, W1S 1YH, UK
| | - Alan M Palmer
- Reading School of Pharmacy, Whiteknights, Reading, RG6 6A, UK
| | - Emily R Fowler
- Pharmidex Pharmaceutical Services, 14 Hanover Street, London, W1S 1YH, UK.,Wellcome Centre for Cell Biology and Institute of Cell Biology, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH9 3BF, Scotland, UK
| | | |
Collapse
|