1
|
Yu J, Liu QC, Lu SY, Wang S, Zhang H. Detecting plasma SHOX2, HOXA9, SEPTIN9, and RASSF1A methylation and circulating cancer cells for cholangiocarcinoma clinical diagnosis and monitoring. World J Gastrointest Oncol 2025; 17:104253. [DOI: 10.4251/wjgo.v17.i4.104253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/17/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA), also known as bile duct cancer, is a devastating malignancy primarily affecting the biliary tract.
AIM To assess their performance in clinical diagnosis and monitoring of CCA, plasma methylation and circulating tumor cells were detected.
METHODS Plasma samples were collected from Hubei Cancer Hospital (n = 156). Plasma DNA was tested to detect SHOX2, HOXA9, SEPTIN9, and RASSF1A methylation using TaqMan PCR. Circulating tumor cells (CTCs) were detected in the peripheral blood of patients using the United States Food and Drug Administration-approved cell search system before and after clinical therapy. The CCA diagnostic value was estimated using the area under the curve. The independent prognosis risk factors for patients with CCA were estimated using Cox and logistic regression analyses.
RESULTS The sensitivity and specificity of the four DNA plasma methylations exhibited 64.74% sensitivity and 93.88% specificity for detecting CCA. The receiver operating characteristic curve of the combined value for CCA diagnosis in plasma was 0.828 ± 0.032. RASSF1A plasma methylation was related to the prognosis of patients with CCA. We determined the prognostic hazard ratio for CCA using CTC count, tumor stage, methylation, and carbohydrate antigen 19-9 levels as key factors. Our overall survival nomogram achieved a C-index of 0.705 (0.605-0.805).
CONCLUSION SHOX2, HOXA9, SEPTIN9, and RASSF1A plasma methylation demonstrated increased sensitivity for diagnosing CCA. RASSF1A plasma methylation and CTCs were valuable predictors to assess CCA prognosis and recurrence.
Collapse
Affiliation(s)
- Jing Yu
- Department of Laboratory, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan's TCWM Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
- Department of Laboratory, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan 442008, Hubei Province, China
| | - Qiu-Chen Liu
- Department of Laboratory, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, Hubei Province, China
| | - Shuang-Yan Lu
- Department of Blood Transfusion, Wuhan Chinese and Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Shun Wang
- Department of Laboratory, Wuhan Chinese and Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Hua Zhang
- Department of Laboratory, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan 442008, Hubei Province, China
| |
Collapse
|
2
|
Jiang T, Mei L, Yang X, Sun T, Wang Z, Ji Y. Biomarkers of gastric cancer: current advancement. Heliyon 2022; 8:e10899. [PMID: 36247151 PMCID: PMC9561735 DOI: 10.1016/j.heliyon.2022.e10899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/22/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
Gastric cancer (GC) is one of the most prevalent malignant types worldwide, especially in East Asia. Due to its frequently advanced stage at diagnosis, the mortality from GC is high and the prognosis is still unsatisfactory. Thus, early detection using effective screening approaches is vital to decrease the morbidity and mortality of GC. Interestingly, biomarkers can be used for diagnosis, prediction of sensitivity to treatment, and prognosis in GC. The potential biomarkers detectable in liquid biopsies such as circulating tumor cells (CTCs), long non-coding RNAs (lncRNAs), cell-free DNA (cfDNA), microRNAs, and exosomes reveal numerous information regarding the early prediction and the outcomes for GC patients. Additionally, using the novel serum biomarkers has opened up new opportunities for diagnosing and monitoring patients with GC. This review mainly summarizes the novel progress and approaches in GC biomarkers, which could be potentially used for early diagnosis and therapy monitoring. Meanwhile, we also discussed the advantages, disadvantages, and future perspectives of GC biomarkers.
Collapse
Affiliation(s)
- Tiantian Jiang
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Lin Mei
- Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Xiao Yang
- Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Tingkai Sun
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Zhidong Wang
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Yuanyuan Ji
- Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
3
|
Han L, Wan Q, Zheng A, Guo Y, Chen Y. Demonstration of a Flexible Graphene-Based Biosensor for Sensitive and Rapid Detection of Ovarian Cancer Cells. NANOSCALE RESEARCH LETTERS 2021; 16:181. [PMID: 34940924 PMCID: PMC8702589 DOI: 10.1186/s11671-021-03633-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 11/21/2021] [Indexed: 06/14/2023]
Abstract
It is significant to develop an efficient early detection and prediction method for ovarian cancer via a facile and low-cost approach. To address such issues, herein, we develop a novel circulating tumor cell (CTC) detection method to sensitively detect ovarian cancer by using a flexible graphene-based biosensor on polyethylene terephthalate (PET) substrate. The results show that the graphene-based flexible biosensor demonstrates sensitive and rapid detection for ovarian cancer cells: it delivers obvious different responses for cell culture medium and cancer solution, different cancer cells and cancer cell solution with different concentrations; it demonstrates high sensitivity for detecting several tens of ovarian cancer cells per ml; moreover, the flexible graphene biosensor is very suitable for rapid and sensitive detection of ovarian cancer cells within 5 s. This work provides a low-cost and facile graphene biosensor fabrication strategy to sensitively and rapidly detect / identify CTC ovarian cancer cells.
Collapse
Affiliation(s)
- Ling Han
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, People's Republic of China
| | - Qi Wan
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, People's Republic of China
| | - Ai Zheng
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, People's Republic of China
| | - Yunchuan Guo
- Chengdu Ginkgo Electronics Technology Co., Ltd., Chengdu, 610213, People's Republic of China
| | - Yali Chen
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
4
|
Sequential Isolation and Characterization of Single CTCs and Large CTC Clusters in Metastatic Colorectal Cancer Patients. Cancers (Basel) 2021; 13:cancers13246362. [PMID: 34944983 PMCID: PMC8699456 DOI: 10.3390/cancers13246362] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary The presence of cancer cells clusters is a frequent event capable of increasing their aptitude to survive in the bloodstream. Consistently, clusters ranging from 2–50 cancer cells are detected in about 50% of patients with metastatic cancers, including colorectal carcinoma. Although a deepened analysis of clusters might certainly offer new insights into the complexity of metastatic cascade, research in this field has come to a halt, since most circulating tumor cells isolation techniques are not compatible with large-sized clusters isolation. In the present study, we describe a sequential method to simultaneously isolate single and clustered circulating tumor cells from a single blood draw, opening new scenarios for an ever more precise characterization of colorectal cancer metastatic cascade. Abstract Circulating tumor cells (CTCs) detach from a primary tumor or its metastases and circulate in the bloodstream. The vast majority of CTCs are deemed to die into the bloodstream, with only few cells representing viable metastatic precursors. Particularly, single epithelial CTCs do not survive long in the circulation due to the loss of adhesion-dependent survival signals. In metastatic colorectal cancer, the generation of large CTC clusters is a very frequent occurrence, able to increase the aptitude of CTCs to survive in the bloodstream. Although a deepened analysis of large-sized CTC clusters might certainly offer new insights into the complexity of the metastatic cascade, most CTC isolation techniques are unfortunately not compatible with large-sized CTC clusters isolation. The inappropriateness of standard CTC isolation devices for large clusters isolation and the scarce availability of detection methods able to specifically isolate and characterize both single CTCs and CTC clusters finally prevented in-depth studies on the prognostic and predictive value of clusters in clinical practice, unlike that which has been described for single CTCs. In the present study, we validated a new sequential filtration method for the simultaneous isolation of large CTC clusters and single CTCs in patients with metastatic colorectal cancer at failure of first-line treatments. The new method might allow differential downstream analyses for single and clustered CTCs starting from a single blood draw, opening new scenarios for an ever more precise characterization of colorectal cancer metastatic cascade.
Collapse
|
5
|
Dunphy K, O’Mahoney K, Dowling P, O’Gorman P, Bazou D. Clinical Proteomics of Biofluids in Haematological Malignancies. Int J Mol Sci 2021; 22:ijms22158021. [PMID: 34360786 PMCID: PMC8348619 DOI: 10.3390/ijms22158021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Since the emergence of high-throughput proteomic techniques and advances in clinical technologies, there has been a steady rise in the number of cancer-associated diagnostic, prognostic, and predictive biomarkers being identified and translated into clinical use. The characterisation of biofluids has become a core objective for many proteomic researchers in order to detect disease-associated protein biomarkers in a minimally invasive manner. The proteomes of biofluids, including serum, saliva, cerebrospinal fluid, and urine, are highly dynamic with protein abundance fluctuating depending on the physiological and/or pathophysiological context. Improvements in mass-spectrometric technologies have facilitated the in-depth characterisation of biofluid proteomes which are now considered hosts of a wide array of clinically relevant biomarkers. Promising efforts are being made in the field of biomarker diagnostics for haematologic malignancies. Several serum and urine-based biomarkers such as free light chains, β-microglobulin, and lactate dehydrogenase are quantified as part of the clinical assessment of haematological malignancies. However, novel, minimally invasive proteomic markers are required to aid diagnosis and prognosis and to monitor therapeutic response and minimal residual disease. This review focuses on biofluids as a promising source of proteomic biomarkers in haematologic malignancies and a key component of future diagnostic, prognostic, and disease-monitoring applications.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Kelly O’Mahoney
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland; (K.O.); (P.O.)
- Correspondence:
| |
Collapse
|
6
|
Gaston C, De Beco S, Doss B, Pan M, Gauquelin E, D'Alessandro J, Lim CT, Ladoux B, Delacour D. EpCAM promotes endosomal modulation of the cortical RhoA zone for epithelial organization. Nat Commun 2021; 12:2226. [PMID: 33850145 PMCID: PMC8044225 DOI: 10.1038/s41467-021-22482-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 03/11/2021] [Indexed: 01/13/2023] Open
Abstract
At the basis of cell shape and behavior, the organization of actomyosin and its ability to generate forces are widely studied. However, the precise regulation of this contractile network in space and time is unclear. Here, we study the role of the epithelial-specific protein EpCAM, a contractility modulator, in cell shape and motility. We show that EpCAM is required for stress fiber generation and front-rear polarity acquisition at the single cell level. In fact, EpCAM participates in the remodeling of a transient zone of active RhoA at the cortex of spreading epithelial cells. EpCAM and RhoA route together through the Rab35/EHD1 fast recycling pathway. This endosomal pathway spatially organizes GTP-RhoA to fine tune the activity of actomyosin resulting in polarized cell shape and development of intracellular stiffness and traction forces. Impairment of GTP-RhoA endosomal trafficking either by silencing EpCAM or by expressing Rab35/EHD1 mutants prevents proper myosin-II activity, stress fiber formation and ultimately cell polarization. Collectively, this work shows that the coupling between co-trafficking of EpCAM and RhoA, and actomyosin rearrangement is pivotal for cell spreading, and advances our understanding of how biochemical and mechanical properties promote cell plasticity.
Collapse
Affiliation(s)
- Cécile Gaston
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, Paris, France
| | - Simon De Beco
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, Paris, France
| | - Bryant Doss
- Mechanobiology Institute, T-lab, Singapore, Singapore
| | - Meng Pan
- Mechanobiology Institute, T-lab, Singapore, Singapore
| | - Estelle Gauquelin
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, Paris, France
| | - Joseph D'Alessandro
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, Paris, France
| | | | - Benoit Ladoux
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, Paris, France
| | - Delphine Delacour
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, Paris, France.
| |
Collapse
|
7
|
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: 33] [Impact Index Per Article: 8.3] [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
|
8
|
Li C, Wang J, Lu X, Ge H, Jin X, Guan Q, su Y, Pan R, Li P, Cai W, Zhu X. Hydrogen peroxide-response nanoprobe for CD44-targeted circulating tumor cell detection and H2O2 analysis. Biomaterials 2020; 255:120071. [DOI: 10.1016/j.biomaterials.2020.120071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022]
|
9
|
Wang D, Ge C, Liang W, Yang Q, Liu Q, Ma W, Shi L, Wu H, Zhang Y, Wu Z, Wei C, Huang L, Fang Z, Liu L, Bao S, Zhang H. In Vivo Enrichment and Elimination of Circulating Tumor Cells by Using a Black Phosphorus and Antibody Functionalized Intravenous Catheter. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000940. [PMID: 32995123 PMCID: PMC7507385 DOI: 10.1002/advs.202000940] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/02/2020] [Indexed: 06/11/2023]
Abstract
The circulating tumor cell (CTC) count is closely related to cancer recurrence and metastasis. The technology that can in vivo destroy CTCs may bring great benefits to patients, which is an urgent clinical demand. Here, a minimally invasive therapeutic intravenous catheter for in vivo enriching and photothermal killing of CTCs is developed. The surface of catheter is modified with anti-EpCAM antibody and the interior is filled with black phosphorus nanosheets (BPNSs). CTCs in the peripheral blood are captured by the catheter continually with the aid of circulation. The captured CTCs are used for downstream analyses or in vivo eliminated by the near-infrared (NIR) photothermal effect of BPNSs. A capture efficiency of 2.1% is obtained during the 5 min of treatment, and 100% of the captured CTCs are killed by following NIR light irradiation in both an in vitro closed-loop circulation system and an in vivo rabbit model. This cost-effective modality for lowering the CTCs burden can be a good supplement to traditional therapies, which holds great promise as an effective clinical intervention for cancer patients.
Collapse
Affiliation(s)
- Dou Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
- Department of Biomedical EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Chenchen Ge
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
- Integrated Chinese and Western Medicine Postdoctoral research stationJinan UniversityGuangzhou510632China
| | - Weiyuan Liang
- Shenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen University and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong ProvinceShenzhen UniversityShenzhen518060China
| | - Qinhe Yang
- School of Traditional Chinese MedicineJinan UniversityGuangzhou510632China
| | - Quan Liu
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
- Integrated Chinese and Western Medicine Postdoctoral research stationJinan UniversityGuangzhou510632China
| | - Wei Ma
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Lulin Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Hong Wu
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Yuhua Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Zongze Wu
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Chaoying Wei
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Luodan Huang
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Zhiyuan Fang
- School of Biomedical and Pharmaceutical SciencesGuangdong University of TechnologyGuangzhou510006China
| | - Liping Liu
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Shiyun Bao
- Department of Hepatobiliary and Pancreatic SurgeryThe 2nd Clinical medical College (Shenzhen People's Hospital) of Jinan UniversityShenzhen518020China
| | - Han Zhang
- Shenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen University and Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong ProvinceShenzhen UniversityShenzhen518060China
| |
Collapse
|
10
|
Cui JJ, Wang LY, Tan ZR, Zhou HH, Zhan X, Yin JY. MASS SPECTROMETRY-BASED PERSONALIZED DRUG THERAPY. MASS SPECTROMETRY REVIEWS 2020; 39:523-552. [PMID: 31904155 DOI: 10.1002/mas.21620] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/09/2019] [Indexed: 06/10/2023]
Abstract
Personalized drug therapy aims to provide tailored treatment for individual patient. Mass spectrometry (MS) is revolutionarily involved in this area because MS is a rapid, customizable, cost-effective, and easy to be used high-throughput method with high sensitivity, specificity, and accuracy. It is driving the formation of a new field, MS-based personalized drug therapy, which currently mainly includes five subfields: therapeutic drug monitoring (TDM), pharmacogenomics (PGx), pharmacomicrobiomics, pharmacoepigenomics, and immunopeptidomics. Gas chromatography-MS (GC-MS) and liquid chromatography-MS (LC-MS) are considered as the gold standard for TDM, which can be used to optimize drug dosage. Matrix-assisted laser desorption ionization-time of flight-MS (MALDI-TOF-MS) significantly improves the capability of detecting biomacromolecule, and largely promotes the application of MS in PGx. It is becoming an indispensable tool for genotyping, which is used to discover and validate genetic biomarkers. In addition, MALDI-TOF-MS also plays important roles in identity of human microbiome whose diversity can explain interindividual differences of drug response. Pharmacoepigenetics is to study the role of epigenetic factors in individualized drug treatment. MS can be used to discover and validate pharmacoepigenetic markers (DNA methylation, histone modification, and noncoding RNA). For the emerging cancer immunotherapy, personalized cancer vaccine has effective immunotherapeutic activity in the clinic. MS-based immunopeptidomics can effectively discover and screen neoantigens. This article systematically reviewed MS-based personalized drug therapy in the above mentioned five subfields. © 2020 John Wiley & Sons Ltd. Mass Spec Rev.
Collapse
Affiliation(s)
- Jia-Jia Cui
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, P. R. China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, P. R. China
| | - Lei-Yun Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, P. R. China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, P. R. China
| | - Zhi-Rong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, P. R. China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, P. R. China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, P. R. China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, P. R. China
| | - Xianquan Zhan
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, P. R. China
- Department of Oncology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, P. R. China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha, 410078, P. R. China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha, 410078, P. R. China
- National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha, 410008, Hunan, P. R. China
- Hunan Provincial Gynecological Cancer Diagnosis and Treatment Engineering Research Center, Changsha, Hunan, 410078, P. R. China
- Hunan Key Laboratory of Precise Diagnosis and Treatment of Gastrointestinal Tumor, Changsha, Hunan, 410078, P. R. China
| |
Collapse
|
11
|
Liang N, Liu L, Li P, Xu Y, Hou Y, Peng J, Song Y, Bing Z, Wang Y, Wang Y, Jia Z, Yang X, Li D, Xu H, Yu Q, Li S, Hu Z, Yang Y. Efficient isolation and quantification of circulating tumor cells in non-small cell lung cancer patients using peptide-functionalized magnetic nanoparticles. J Thorac Dis 2020; 12:4262-4273. [PMID: 32944338 PMCID: PMC7475553 DOI: 10.21037/jtd-20-1026a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Circulating tumor cells (CTCs) carry a wealth of information on primary and metastatic tumors critical for enhancing the understanding of the occurrence, progression and metastasis of non-small cell lung cancer (NSCLC). However, the low sensitivity of traditional tumor detection methods limits the application of CTCs in the treatment and disease surveillance of NSCLC. Therefore, CTCs isolation and detection with high sensitivity is highly desired especially for NSCLC patients, which is significant because of high occurrence and mortality. While it is very challenging because of the lower expression of CTC positive biomarkers such as epithelial cell adhesion molecules and cytokeratins (EpCAM and CKs), herein we report a method based on peptide-functionalized magnetic nanoparticles with high CTC capture efficiency, which demonstrates superiority in NSCLC clinical applications. Methods For analysis and comparison of the peptide-functionalized magnetic nanoparticles (TumorFisher, Nanopep Corp.) and the antibody-modified magnetic beads (CellSearch, Janssen Diagnostics, LLC), two NSCLC cell lines, A549 and NCI-H1975 were chosen to measure the binding affinity and capture efficiency. In order to compare the effect of the clinical application of these two detection systems, 7 early stage patients with NSCLC were enrolled in this study. To further explore the clinical utility of CTC counting in different stages, 81 NSCLC patients in stage I–IV were enrolled for CTC enumeration and statistical analysis. Results The binding affinities of the recognition peptide to A549 and NCI-H1975 are 76.7%±11.0% and 70.1%±4.8%, respectively, which is similar with the positive control group (anti-EpCAM antibodies). CTCs were captured in 5/7 (71.4%) of early stage NSCLC patients with NSCLC in TumorFisher system, which is higher than CellSearch, and the false negative of TumorFisher is much lower than CellSearch. In a larger clinical cohort, the CTC numbers of NSCLC patients varied in different stages and the overall detection rate of TumorFisher was 59/81 (72.8%), with the similar proportion in stage I (21/29, 72.4%), II (17/22, 77.3%) and III (16/21, 76.2%). Conclusions Highly efficient CTC isolation technique based on peptide-magnetic nanoparticles was firstly applied in NSCLC patients. Compared with the antibody-based the technique, the higher CTC detection rates (71.4%) in NSCLC patient blood samples were demonstrated for the patients in different stages, I–IV, especially in early stages. This indicates the feasibility of the clinical utility of this new technique in early stage screening, prognosis and therapy evaluation of NSCLC.
Collapse
Affiliation(s)
- Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ping Li
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Yuan Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | | | - Jiaxi Peng
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Yang Song
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanyu Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziqi Jia
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoying Yang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Da Li
- Nanopep Biotech Co., Beijing, China
| | | | - Qian Yu
- Nanopep Biotech Co., Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiyuan Hu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.,Nanopep Biotech Co., Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yanlian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.,Nanopep Biotech Co., Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
12
|
Zeng H, Veeramootoo JS, Ma G, Jiang Y, Wang J, Xia T, Liu X. Clinical value and feasibility of ISET in detecting circulating tumor cells in early breast cancer. Transl Cancer Res 2020; 9:4297-4305. [PMID: 35117796 PMCID: PMC8799038 DOI: 10.21037/tcr-19-2662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 06/03/2020] [Indexed: 12/24/2022]
Abstract
Background Patients with operable breast cancer have a better prognosis for recovery. However, once distant organ metastasis occurs, the chance of a long-term survival or a cure is limited. The collection and counting of circulating tumor cells (CTCs) by reliable detection techniques are of increasing importance in the diagnosis of early metastasis and prognosis of disease progression. Isolation by size of epithelial tumor cells (ISET) has the advantage of simplicity of operation and high homogeneity. It is practical for large-scale clinical detection showing cell abundance. The value of ISET in the detection of circulating breast cancers in the blood has not been determined. The purpose of this study is to explore the feasibility of applying ISET to detect CTCs by determining the detection rate of ISET in operable breast cancer and by evaluating the correlation between detection rate, cell count and clinical factors such as molecular typing and pathological staging. Methods The experiment included 193 breast cancer patients who were diagnosed by core needle biopsy before the operation. 10 mL of venous blood was collected from the patients preoperatively, and CTCs in their blood samples were counted and analyzed by ISET. Results Patients were divided into groups according to pathology and immunohistochemistry. The overall detection rate of CTCs by ISET was 41.24%. The detection rate, the number of overall CTCs and the average number of CTCs in each group were analyzed individually. No significant differences were observed between the different groups. Conclusions Although ISET has a relatively good detection rate for circulating breast cancer cells, it fails to provide more information on pathological staging, molecular classification and so forth.
Collapse
Affiliation(s)
- Hanling Zeng
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | | | - Ge Ma
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yi Jiang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jingyi Wang
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center For Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Russell LM, Liu CH, Grodzinski P. Nanomaterials innovation as an enabler for effective cancer interventions. Biomaterials 2020; 242:119926. [PMID: 32169771 DOI: 10.1016/j.biomaterials.2020.119926] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Nanomedicines have been developing very rapidly and have started to play a significant role in several cancer therapeutic modalities. Early on, the nanomedicine field focused on optimizing pharmacokinetics, toxicity, and/or biodistribution of an agent through nanoparticle formulation. In other cases, where materials science is employed more decisively, nanomedicine can include the creation of new agents that take advantage of nanoscale materials properties to enhance treatment efficacy through unique mode of action, molecular targeting, or controlled drug release. Both current and future nanomedicines will seek to contribute to the therapeutic and diagnostic landscape through creative leveraging of mechanical, electrical, optical, magnetic, and biological nanomaterial properties. In this work, we discuss how by modulating these material properties, one can design more diverse and more effective cancer interventions. We focus on six areas in cancer management, including in vitro diagnostics, clinical imaging, theranostics, combination therapy, immunotherapy, and gene therapy.
Collapse
Affiliation(s)
- Luisa M Russell
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina H Liu
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
14
|
Mansouri S, Mokhtari-Hesari P, Naghavi-Al-Hosseini F, Majidzadeh-A K, Farahmand L. The Prognostic Value of Circulating Tumor Cells in Primary Breast Cancer Prior to any Systematic Therapy: A Systematic Review. Curr Stem Cell Res Ther 2020; 14:519-529. [PMID: 30843493 DOI: 10.2174/1574888x14666190306103759] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/04/2019] [Accepted: 02/15/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Numerous studies have defined the outstanding role of circulating tumor cells (CTC) in the management of cancer, particularly the ones in association with primary tumor metastases. OBJECTIVE The overall aim of the present study was to investigate whether CTCs may serve as a clinical prognostic marker for survival in primary breast cancer. METHODS Articles Published from June 2011 to July 2017 in PubMed, EMBase, and Cochrane library databases were thoroughly screened for selecting the ones meeting the inclusion criteria. RESULT Studies applying CellSearch® method demonstrated the risk ratios (RR) of 2.51 (95% CI: 1.78- 3.54), 3.98 (95% CI: 2.28- 6.95), 5.59 (95% CI: 3.29- 9.51), and 3.38 (95% CI: 1.88- 6.06) for death rate and relapse rates of 2.48 (95% CI: 1.89 - 3.26), 3.62 (95% CI: 2.37 - 5.51), 4.45 (95% CI: 2.94 - 6.73), and 2.88 (95 % CI: 1.99 - 4.17) at four CTC positive cut points (≥ 1, ≥ 2, ≥ 3, and ≥ 5 CTCs/7.5 ml). Two studies applying the AdnaTest® also documented increased death (RR: 1.38, 95 % CI: 0.42- 4.49) and relapse rates (RR: 2.97, 95 % CI: 1.23 - 7.18)). CONCLUSION Results of this meta-analysis allude CTCs as potent prognostic markers in primary breast cancers prior to any systemic therapy especially when it is studied via CellSearch® administration, considering that the more the CTCs, the greater the death and relapse rates.
Collapse
Affiliation(s)
- Sepideh Mansouri
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Parisa Mokhtari-Hesari
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Fatemeh Naghavi-Al-Hosseini
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
15
|
Li N, Zuo H, Chen L, Liu H, Zhou J, Yao Y, Xu B, Gong H, Weng Y, Hu Q, Song Q, Peng M, Cheng Y. Circulating Tumor Cell Detection In Epithelial Ovarian Cancer Using Dual-Component Antibodies Targeting EpCAM And FRα. Cancer Manag Res 2019; 11:10939-10948. [PMID: 32021417 PMCID: PMC6978676 DOI: 10.2147/cmar.s211455] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 10/02/2019] [Indexed: 12/25/2022] Open
Abstract
Purpose Circulating tumor cell (CTC) detection methods based on epithelial cell adhesion molecule (EpCAM) have low detection rates in epithelial ovarian cancer (EOC). Meanwhile, folate receptor alpha (FRα) has high expression in EOC cells. We explored the feasibility of combining FRα and EpCAM as CTC capture targets in EOC. Patients and methods EpCAM and FRα antibodies were linked to magnetic nanospheres (MNs) using the principle of carbodiimide chemistry. Blood samples from healthy donor spiked with A2780 ovarian cancer cells were used for detecting the capture rate. Ninety-five blood samples from 30 patients with EOC were used for comparing the positive rate of detection when using anti-EpCAM-MNs alone with that when using combination of anti-EpCAM-MNs and anti-FRα-MNs. Samples from 28 patients initially diagnosed with EOC and 20 patients with ovarian benign disease were used for evaluating the sensitivity and specificity of combination of anti-EpCAM-MNs and anti-FRα-MNs. Results Regression analysis between the number of recovered and that of spiked A2780 cells revealed yEpCAM = 0.535x (R2 = 0.99), yFRα = 0.901x (R2 = 0.99), and yEpCAM+FRα = 0.928x (R2 = 0.99). In mixtures of A2780 and MCF7 cells, the capture rate was 92% using the combination of anti-EpCAM-MNs and anti-FRα-MNs, exceeding the rate when using anti-EpCAM-MNs or anti-FRα-MNs alone by approximately 20% (P < 0.01). The combination of anti-EpCAM-MNs and anti-FRα-MNs showed a significantly increased positive rate of CTC detection in EOC patients compared with anti-EpCAM-MNs alone (χ2 = 14.45, P < 0.001). Sensitivity values were 0.536 and 0.75 and specificity values were 0.9 and 0.85 when using anti-EpCAM-MNs alone and when using the combination of anti-EpCAM-MNs and anti-FRα-MNs, respectively. Conclusion The combination of FRα and EpCAM is feasible as a CTC capture target of CTC detection in patients with EOC.
Collapse
Affiliation(s)
- Na Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Hao Zuo
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Luojun Chen
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Huali Liu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Jin Zhou
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yi Yao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Bin Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Hongyun Gong
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yiming Weng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Qibin Song
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
16
|
Duffy MJ, McDermott EW, Crown J. Blood-based biomarkers in breast cancer: From proteins to circulating tumor cells to circulating tumor DNA. Tumour Biol 2018; 40:1010428318776169. [DOI: 10.1177/1010428318776169] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Biomarkers are the key to personalized treatment in patients with breast cancer. While tissue biomarkers are most useful in determining prognosis and upfront predicting response to therapy, circulating protein biomarkers such as CA 15-3 and carcinoembryonic antigen are mainly used in monitoring response to endocrine or chemotherapy in patients with advanced disease. Although several centers measure biomarkers in asymptomatic patients following curative surgery for primary breast cancer, the clinical utility of this practice is unclear. Promising new biomarkers for breast cancer include circulating tumor DNA and circulating tumor cells. In contrast to circulating protein biomarkers, measurement of circulating tumor DNA–based biomarkers is potentially useful in identifying mechanisms of resistance to ongoing therapies as well as identifying new targets for further treatment. To increase clinical utility, both the established and emerging circulating biomarkers should where possible be incorporated into randomized trials evaluating new therapies in patients with breast cancer.
Collapse
Affiliation(s)
- Michael J Duffy
- Clinical Research Centre, St. Vincent’s University Hospital, Dublin, Ireland
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Enda W McDermott
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - John Crown
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Ireland
| |
Collapse
|