1
|
Bolideei M, Barzigar R, Gahrouei RB, Mohebbi E, Haider KH, Paul S, Paul MK, Mehran MJ. Applications of Gene Editing and Nanotechnology in Stem Cell-Based Therapies for Human Diseases. Stem Cell Rev Rep 2025:10.1007/s12015-025-10857-0. [PMID: 40014250 DOI: 10.1007/s12015-025-10857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Stem cell research is a dynamic and fast-advancing discipline with great promise for the treatment of diverse human disorders. The incorporation of gene editing technologies, including ZFNs, TALENs, and the CRISPR/Cas system, in conjunction with progress in nanotechnology, is fundamentally transforming stem cell therapy and research. These innovations not only provide a glimmer of optimism for patients and healthcare practitioners but also possess the capacity to radically reshape medical treatment paradigms. Gene editing and nanotechnology synergistically enhance stem cell-based therapies' precision, efficiency, and applicability, offering transformative potential for treating complex diseases and advancing regenerative medicine. Nevertheless, it is important to acknowledge that these technologies also give rise to ethical considerations and possible hazards, such as inadvertent genetic modifications and the development of genetically modified organisms, therefore creating a new age of designer infants. This review emphasizes the crucial significance of gene editing technologies and nanotechnology in the progress of stem cell treatments, particularly for degenerative pathologies and injuries. It emphasizes their capacity to restructure and comprehensively revolutionize medical treatment paradigms, providing fresh hope and optimism for patients and healthcare practitioners.
Collapse
Affiliation(s)
- Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Rambod Barzigar
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India
| | - Razieh Bahrami Gahrouei
- Department of Pharmacy PES College, Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka, India
| | - Elham Mohebbi
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois School of Medicine, Springfield, IL, USA
| | - Khawaja Husnain Haider
- Sulaiman AlRajhi Medical School, Al Bukayriyah, AlQaseem, 52726, Kingdom of Saudi Arabia
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA.
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Mohammad Javad Mehran
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India.
| |
Collapse
|
2
|
Pipatwatcharadate C, Iyer PR, Pissuwan D. Recent Update Roles of Magnetic Nanoparticles in Circulating Tumor Cell (CTC)/Non-CTC Separation. Pharmaceutics 2023; 15:2482. [PMID: 37896242 PMCID: PMC10610106 DOI: 10.3390/pharmaceutics15102482] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Metastasis of cancer is a major cause of death worldwide. Circulating tumor cells (CTCs) are important in the metastatic process of cancer. CTCs are able to circulate in the bloodstream. Therefore, they can be used as biomarkers of metastasis. However, CTCs are rare when compared to a large number of blood cells in the blood. Many CTC detection methods have been developed to increase CTC detection efficiency. Magnetic nanoparticles (MNPs) have attracted immense attention owing to their potential medical applications. They are particularly appealing as a tool for cell separation. Because of their unique properties, MNPs are of considerable interest for the enrichment of CTCs through CTC or non-CTC separation. Herein, we review recent developments in the application of MNPs to separate CTCs or non-CTCs in samples containing CTCs. This review provides information on new approaches that can be used to detect CTCs in blood samples. The combination of MNPs with other particles for magnetic-based cell separation for CTC detection is discussed. Furthermore, different approaches for synthesizing MNPs are included in this review.
Collapse
Affiliation(s)
- Chawapon Pipatwatcharadate
- Nanobiotechnology and Nanobiomaterials Research (N-BMR) Laboratory, School of Materials Science and Innovation, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (C.P.); (P.R.I.)
| | - Poornima Ramesh Iyer
- Nanobiotechnology and Nanobiomaterials Research (N-BMR) Laboratory, School of Materials Science and Innovation, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (C.P.); (P.R.I.)
- Materials Science and Engineering Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Dakrong Pissuwan
- Nanobiotechnology and Nanobiomaterials Research (N-BMR) Laboratory, School of Materials Science and Innovation, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (C.P.); (P.R.I.)
- Materials Science and Engineering Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Center of Excellence on Medical Biotechnology (CEMB), Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
3
|
Bhattacharya S, Majumdar Nee Paul S. Application of conventional metallic nanoparticles on male reproductive system - challenges and countermeasures. Syst Biol Reprod Med 2023; 69:32-49. [PMID: 36427189 DOI: 10.1080/19396368.2022.2140087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The application of nanotechnology in the present era has substantial impact on different industrial and medical fields. However, the advancement in nanotechnology for potential therapeutic and consumer benefits has been an anxious cause regarding the probable hazardous consequences of these molecules in biological systems and the environment. The toxic effects can perturb the physiologic system broadly and reproductive function and fertility specifically. Despite engineered nanomaterials (ENMs) having a wide range of applications, toxicological investigations of the probable ramifications of ENMs on the reproductive systems of mammals and fertility remains in its nascence. Complication in the male reproductive system is quite a pertinent issue in today's world which comprises of benign prostatic enlargement, prostate cancer, and unhealthy sperm production. The therapeutic drugs should not only be active in minimum dose but also site-specific in action, criteria being met by nanomedicines. Nanomedicine therapy is promising but encompasses the chances of adverse effects of being cytotoxic and generating oxidative stress. These hurdles can be overcome by creating coated nanoparticles with organic substances, modification of shape and size, and synthesizing biocompatible green nanoparticles. This review attempts to look into the applications of most widely used metals like zinc, titanium, silver, and gold nanoparticles in the therapy of the male reproductive system, their prospective harmful effects, and the way out to create a safe therapeutic system by specific modifications of these metal and metal oxide nanoparticles.
Collapse
Affiliation(s)
- Sonali Bhattacharya
- Department of Zoology (Post Graduate Studies), Rishi Bankim Chandra College, West Bengal State University, Naihati, West Bengal, India
| | - Sudipta Majumdar Nee Paul
- Department of Zoology (Post Graduate Studies), Rishi Bankim Chandra College, West Bengal State University, Naihati, West Bengal, India
| |
Collapse
|
4
|
Isolation, Detection and Analysis of Circulating Tumour Cells: A Nanotechnological Bioscope. Pharmaceutics 2023; 15:pharmaceutics15010280. [PMID: 36678908 PMCID: PMC9864919 DOI: 10.3390/pharmaceutics15010280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/17/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Cancer is one of the dreaded diseases to which a sizeable proportion of the population succumbs every year. Despite the tremendous growth of the health sector, spanning diagnostics to treatment, early diagnosis is still in its infancy. In this regard, circulating tumour cells (CTCs) have of late grabbed the attention of researchers in the detection of metastasis and there has been a huge surge in the surrounding research activities. Acting as a biomarker, CTCs prove beneficial in a variety of aspects. Nanomaterial-based strategies have been devised to have a tremendous impact on the early and rapid examination of tumor cells. This review provides a panoramic overview of the different nanotechnological methodologies employed along with the pharmaceutical purview of cancer. Initiating from fundamentals, the recent nanotechnological developments toward the detection, isolation, and analysis of CTCs are comprehensively delineated. The review also includes state-of-the-art implementations of nanotechnological advances in the enumeration of CTCs, along with future challenges and recommendations thereof.
Collapse
|
5
|
Bharti S, Anant PS, Kumar A. Nanotechnology in stem cell research and therapy. JOURNAL OF NANOPARTICLE RESEARCH 2023; 25:6. [DOI: 10.1007/s11051-022-05654-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2025]
|
6
|
Alhaj-Suliman SO, Wafa EI, Salem AK. Engineering nanosystems to overcome barriers to cancer diagnosis and treatment. Adv Drug Deliv Rev 2022; 189:114482. [PMID: 35944587 DOI: 10.1016/j.addr.2022.114482] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 01/24/2023]
Abstract
Over the past two decades, multidisciplinary investigations into the development of nanoparticles for medical applications have continually increased. However, nanoparticles are still subject to biological barriers and biodistribution challenges, which limit their overall clinical potential. This has motivated the implementation of innovational modifications to a range of nanoparticle formulations designed for cancer imaging and/or cancer treatment to overcome specific barriers and shift the accumulation of payloads toward the diseased tissues. In recent years, novel technological and chemical approaches have been employed to modify or functionalize the surface of nanoparticles or manipulate the characteristics of nanoparticles. Combining these approaches with the identification of critical biomarkers provides new strategies for enhancing nanoparticle specificity for both cancer diagnostic and therapeutic applications. This review discusses the most recent advances in the design and engineering of nanoparticles as well as future directions for developing the next generation of nanomedicines.
Collapse
Affiliation(s)
- Suhaila O Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States; Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, United States.
| |
Collapse
|
7
|
Zhao J, Zhang C, Wang W, Li C, Mu X, Hu K. Current progress of nanomedicine for prostate cancer diagnosis and treatment. Biomed Pharmacother 2022; 155:113714. [PMID: 36150309 DOI: 10.1016/j.biopha.2022.113714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/02/2022] Open
Abstract
Prostate cancer (PCa) is the most common new cancer case and the second most fatal malignancy in men. Surgery, endocrine therapy, radiotherapy and chemotherapy are the main clinical treatment options for PCa. However, most prostate cancers can develop into castration-resistant prostate cancer (CRPC), and due to the invasiveness of prostate cancer cells, they become resistant to different treatments and activate tumor-promoting signaling pathways, thereby inducing chemoresistance, radioresistance, ADT resistance, and immune resistance. Nanotechnology, which can combine treatment with diagnostic imaging tools, is emerging as a promising treatment modality in prostate cancer therapy. Nanoparticles can not only promote their accumulation at the pathological site through passive targeting techniques for enhanced permeability and retention (EPR), but also provide additional advantages for active targeting using different ligands. This property results in a reduced drug dose to achieve the desired effect, a longer duration of action within the tumor and fewer side effects on healthy tissues. In addition, nanotechnology can create good synergy with radiotherapy, chemotherapy, thermotherapy, photodynamic therapy and gene therapy to enhance their therapeutic effects with greater scope, and reduce the resistance of prostate cancer. In this article, we intend to review and discuss the latest technologies regarding the use of nanomaterials as therapeutic and diagnostic tools for prostate cancer.
Collapse
Affiliation(s)
- Jiang Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Chi Zhang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Weihao Wang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Chen Li
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun 130033, China.
| | - Kebang Hu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
8
|
Hu Y, Chen D, Napoleon JV, Srinivasarao M, Singhal S, Savran CA, Low PS. Efficient capture of circulating tumor cells with low molecular weight folate receptor-specific ligands. Sci Rep 2022; 12:8555. [PMID: 35595733 PMCID: PMC9122947 DOI: 10.1038/s41598-022-12118-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/29/2022] [Indexed: 11/09/2022] Open
Abstract
Retrieval of circulating tumor cells (CTC) has proven valuable for assessing a patient's cancer burden, evaluating response to therapy, and analyzing which drug might treat a cancer best. Although most isolation methods retrieve CTCs based on size, shape, or capture by tumor-specific antibodies, we explore here the use of small molecule tumor-specific ligands linked to magnetic beads for CTC capture. We have designed folic acid-biotin conjugates with different linkers for the capture of folate receptor (FR) + tumor cells spiked into whole blood, and application of the same technology to isolate FR + CTCs from the peripheral blood of both tumor-bearing mice and non-small cell lung patients. We demonstrate that folic acid linked via a rigid linker to a flexible PEG spacer that is in turn tethered to a magnetic bead enables optimal CTC retrieval, reaching nearly 100% capture when 100 cancer cells are spiked into 1 mL of aqueous buffer and ~ 90% capture when the same quantity of cells is diluted into whole blood. In a live animal model, the same methodology is shown to efficiently retrieve CTCs from tumor-bearing mice, yielding cancer cell counts that are proportional to total tumor burden. More importantly, the same method is shown to collect ~ 29 CTCs/8 mL peripheral blood from patients with non-small cell lung cancer. Since the ligand-presentation strategy optimized here should also prove useful in targeting other nanoparticles to other cells, the methods described below should have general applicability in the design of nanoparticles for cell-specific targeting.
Collapse
Affiliation(s)
- Yingwen Hu
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Danyang Chen
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - John V Napoleon
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Madduri Srinivasarao
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cagri A Savran
- School of Mechanical Engineering, Birck Nanotechnology Center, Purdue Center for Cancer Research, Purdue University, 1205 W. State St., West Lafayette, IN, 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute for Drug Discovery, Purdue University, 720 Clinic Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
9
|
Li F, Xu H, Zhao Y. Magnetic particles as promising circulating tumor cell catchers assisting liquid biopsy in cancer diagnosis: A review. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
10
|
Nanostructure Materials: Efficient Strategies for Circulating Tumor Cells Capture, Release, and Detection. BIOTECHNOL BIOPROC E 2021. [DOI: 10.1007/s12257-020-0257-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
11
|
Zamani Kouhpanji MR, Nemati Z, Mahmoodi MM, Um J, Modiano J, Franklin R, Stadler B. Selective Detection of Cancer Cells Using Magnetic Nanowires. ACS APPLIED MATERIALS & INTERFACES 2021; 13:21060-21066. [PMID: 33904709 DOI: 10.1021/acsami.1c04628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The main bottleneck for implementing magnetic nanowires (MNWs) in cell-biology research for multimodal therapeutics is the inapplicability of the current state of the art for selective detection and stimulation of MNWs. Here, we introduce a methodology for selective detection of MNWs in platforms that have multiple magnetic signals, such as future multimodal therapeutics. After characterizing the signatures of MNWs, MNWs were surface-functionalized and internalized into canine osteosarcoma (OSCA-8) cancer cells for cell labeling, manipulation, and separation. We also prepared and characterized magnetic biopolymers as multimodal platforms for future use in controlling the movement, growth, and division of cancer cells. First, it is important to have methods for distinguishing the magnetic signature of the biopolymer from the magnetically labeled cells. For this purpose, we use the projection method to selectively detect and demultiplex the magnetic signatures of MNWs inside cells from those inside magnetic biopolymers. We show that tailoring the irreversible switching field of MNWs by tuning their coercivity is a highly effective approach for generating distinct magnetic biolabels for selective detection of cancer cells. These findings open up new possibilities for selective stimulation of MNWs in multimodal therapeutic platforms for drug delivery, hyperthermia cancer therapy, and mitigating cancer cell movement and proliferation.
Collapse
Affiliation(s)
- Mohammad Reza Zamani Kouhpanji
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Zohreh Nemati
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Joseph Um
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jaime Modiano
- Masonic Cancer Research Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota 55108, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rhonda Franklin
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Bethanie Stadler
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
12
|
Wen J, Tang T, Kanwal S, Lu Y, Tao C, Zheng L, Zhang D, Gu Z. Detection and Classification of Multi-Type Cells by Using Confocal Raman Spectroscopy. Front Chem 2021; 9:641670. [PMID: 33912538 PMCID: PMC8071986 DOI: 10.3389/fchem.2021.641670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/19/2021] [Indexed: 11/25/2022] Open
Abstract
Tumor cells circulating in the peripheral blood are the prime cause of cancer metastasis and death, thus the identification and discrimination of these rare cells are crucial in the diagnostic of cancer. As a label-free detection method without invasion, Raman spectroscopy has already been indicated as a promising method for cell identification. This study uses a confocal Raman spectrometer with 532 nm laser excitation to obtain the Raman spectrum of living cells from the kidney, liver, lung, skin, and breast. Multivariate statistical methods are applied to classify the Raman spectra of these cells. The results validate that these cells can be distinguished from each other. Among the models built to predict unknown cell types, the quadratic discriminant analysis model had the highest accuracy. The demonstrated analysis model, based on the Raman spectrum of cells, is propitious and has great potential in the field of biomedical for classifying circulating tumor cells in the future.
Collapse
Affiliation(s)
- Jing Wen
- Engineering Research Center of Optical Instrument and Systems, Ministry of Education and Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Tianchen Tang
- Engineering Research Center of Optical Instrument and Systems, Ministry of Education and Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Saima Kanwal
- Engineering Research Center of Optical Instrument and Systems, Ministry of Education and Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Yongzheng Lu
- Engineering Research Center of Optical Instrument and Systems, Ministry of Education and Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Chunxian Tao
- Engineering Research Center of Optical Instrument and Systems, Ministry of Education and Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Lulu Zheng
- Engineering Research Center of Optical Instrument and Systems, Ministry of Education and Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Dawei Zhang
- Engineering Research Center of Optical Instrument and Systems, Ministry of Education and Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Zhengqin Gu
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Wang YH, Song Z, Hu XY, Wang HS. Circulating tumor DNA analysis for tumor diagnosis. Talanta 2021; 228:122220. [PMID: 33773726 DOI: 10.1016/j.talanta.2021.122220] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/05/2021] [Accepted: 02/13/2021] [Indexed: 01/10/2023]
Abstract
Tumor is a kind of abnormal organism generated by the proliferation and differentiation of cells in the body under the action of various initiating and promoting factors, which seriously threatens human life and health. Tumorigenesis is a gradual process that involves multistage reactions and the accumulation of mutations. Gene mutation usually occurs during tumorigenesis, and can be used for tumor diagnosis. Early diagnosis is the most effective way to improve the cure rate and reduce the mortality rate. Among the peripheral blood circulating tumor DNA (ctDNA), gene mutation in keeping with tumor cells can be detected, which can potentially replace tumor tissue section for early diagnosis. It has been considered as a liquid biopsy marker with good clinical application prospect. However, the high fragmentation and low concentration of ctDNA in blood result in the difficulty of tumor stage determination. Therefore, high sensitive and specific mutation detection methods have been developed to detect trace mutant ctDNA. At present, the approaches include digital PCR (dPCR), Bead, Emulsion, Amplification and Magnetic (BEAMing), Next Generation Sequencing (NGS), Amplification Refractory Mutation System (ARMS), etc. In this paper, the principle, characteristics, latest progress and application prospects of these methods are reviewed, which will facilitate researchers to choose appropriate ctDNA detection approaches.
Collapse
Affiliation(s)
- Yi-Hui Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing, 210009, China; Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhen Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing, 210009, China; Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Xin-Yuan Hu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing, 210009, China; Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China
| | - Huai-Song Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), China Pharmaceutical University, Nanjing, 210009, China; Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
14
|
Singh B, Arora S, D'Souza A, Kale N, Aland G, Bharde A, Quadir M, Calderón M, Chaturvedi P, Khandare J. Chemo-specific designs for the enumeration of circulating tumor cells: advances in liquid biopsy. J Mater Chem B 2021; 9:2946-2978. [PMID: 33480960 DOI: 10.1039/d0tb02574g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Advanced materials and chemo-specific designs at the nano/micrometer-scale have ensured revolutionary progress in next-generation clinically relevant technologies. For example, isolating a rare population of cells, like circulating tumor cells (CTCs) from the blood amongst billions of other blood cells, is one of the most complex scientific challenges in cancer diagnostics. The chemical tunability for achieving this degree of exceptional specificity for extra-cellular biomarker interactions demands the utility of advanced entities and multistep reactions both in solution and in the insoluble state. Thus, this review delineates the chemo-specific substrates, chemical methods, and structure-activity relationships (SARs) of chemical platforms used for isolation and enumeration of CTCs in advancing the relevance of liquid biopsy in cancer diagnostics and disease management. We highlight the synthesis of cell-specific, tumor biomarker-based, chemo-specific substrates utilizing functionalized linkers through chemistry-based conjugation strategies. The capacity of these nano/micro substrates to enhance the cell interaction specificity and efficiency with the targeted tumor cells is detailed. Furthermore, this review accounts for the importance of CTC capture and other downstream processes involving genotypic and phenotypic CTC analysis in real-time for the detection of the early onset of metastases progression and chemotherapy treatment response, and for monitoring progression free-survival (PFS), disease-free survival (DFS), and eventually overall survival (OS) in cancer patients.
Collapse
Affiliation(s)
- Balram Singh
- Actorius Innovations and Research Pvt. Ltd, Pune, 411057, India.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Gessner I, Fries JWU, Brune V, Mathur S. Magnetic nanoparticle-based amplification of microRNA detection in body fluids for early disease diagnosis. J Mater Chem B 2020; 9:9-22. [PMID: 33179710 DOI: 10.1039/d0tb02165b] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Circulating biomarkers such as microRNAs (miRNAs), short noncoding RNA strands, represent prognostic and diagnostic indicators for a variety of physiological disorders making their detection and quantification an attractive approach for minimally invasive early disease diagnosis. However, highly sensitive and selective detection methods are required given the generally low abundance of miRNAs in body fluids together with the presence of large amounts of other potentially interfering biomolecules. Although a variety of miRNA isolation and detection methods have been established in clinics, they usually require trained personnel and often constitute labor-, time- and cost-intensive approaches. During the last years, nanoparticle-based biosensors have received increasing attention due to their superior detection efficiency even in very low concentration regimes. This is based on their unique physicochemical properties in combination with their high surface area that allows for the immobilization of multiple recognition sites resulting in fast and effective recognition of analytes. Among various materials, magnetic nanoparticles have been identified as useful tools for the separation, concentration, and detection of miRNAs. Here, we review state-of-the-art technology with regard to magnetic particle-based miRNA detection from body fluids, critically discussing challenges and future perspective of such biosensors while comparing their handling, sensitivity as well as selectivity against the established miRNA isolation and detection methods.
Collapse
Affiliation(s)
- Isabel Gessner
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | | | | | | |
Collapse
|
16
|
Dianat-Moghadam H, Mahari A, Heidarifard M, Parnianfard N, Pourmousavi-Kh L, Rahbarghazi R, Amoozgar Z. NK cells-directed therapies target circulating tumor cells and metastasis. Cancer Lett 2020; 497:41-53. [PMID: 32987138 DOI: 10.1016/j.canlet.2020.09.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/28/2020] [Accepted: 09/20/2020] [Indexed: 12/15/2022]
Abstract
Metastasis is the major cause of cancer-related deaths. Invasive primary cancers often metastasize after circulating tumor cells (CTCs) enter the bloodstream or lymph node to colonize adjacent tissue or distant anatomical locations. CTCs interact with immune cells and metastatic microenvironments, survival signaling, and chemotherapeutic resistance. Among immune cells, natural killer (NK) cells can, directly and indirectly, interact with CTCs to control cancer metastasis. Understanding the molecular mechanisms that drive NK cells mediated recognition and elimination of CTCs may pave the way for a new generation of anti-CTC molecularly targeted immunotherapies. In this review, we will discuss i) the role of CTCs in metastases, ii) CTCs in the context of the tumor microenvironment, iii) CTCs immune escape, and finally, iv) the potentials of NK cell-based therapies alone, or in combination with nanomedicine for targeted-immunotherapies of metastatic diseases.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mahari
- Department of Chemical Engineering, Islamic Azad University, Ahar Branch, Ahar, Iran
| | - Maryam Heidarifard
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Parnianfard
- Department of Internal Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laleh Pourmousavi-Kh
- Department of Exercise Physiology, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard, Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Barani M, Sabir F, Rahdar A, Arshad R, Kyzas GZ. Nanotreatment and Nanodiagnosis of Prostate Cancer: Recent Updates. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1696. [PMID: 32872181 PMCID: PMC7559844 DOI: 10.3390/nano10091696] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022]
Abstract
The fabrication and development of nanomaterials for the treatment of prostate cancer have gained significant appraisal in recent years. Advancements in synthesis of organic and inorganic nanomaterials with charge, particle size, specified geometry, ligand attachment etc have resulted in greater biocompatibility and active targeting at cancer site. Despite all of the advances made over the years in discovering drugs, methods, and new biomarkers for cancer of the prostate (PCa), PCa remains one of the most troubling cancers among people. Early on, effective diagnosis is an essential part of treating prostate cancer. Prostate-specific antigen (PSA) or serum prostate-specific antigen is the best serum marker widely accessible for diagnosis of PCa. Numerous efforts have been made over the past decade to design new biosensor-based strategies for biomolecules detection and PSA miniaturization biomarkers. The growing nanotechnology is expected to have a significant effect in the immediate future on scientific research and healthcare. Nanotechnology is thus predicted to find a way to solve one of the most and long-standing problem, "early cancer detection". For early diagnosis of PCa biomarkers, different nanoparticles with different approaches have been used. In this review, we provide a brief description of the latest achievements and advances in the use of nanoparticles for PCa biomarker diagnosis.
Collapse
Affiliation(s)
- Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 7616914111, Iran;
| | - Fakhara Sabir
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-98615, Iran
| | - Rabia Arshad
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - George Z. Kyzas
- Department of Chemistry, International Hellenic University, 65404 Kavala, Greece
| |
Collapse
|
18
|
Miccio L, Cimmino F, Kurelac I, Villone MM, Bianco V, Memmolo P, Merola F, Mugnano M, Capasso M, Iolascon A, Maffettone PL, Ferraro P. Perspectives on liquid biopsy for label‐free detection of “circulating tumor cells” through intelligent lab‐on‐chips. VIEW 2020. [DOI: 10.1002/viw.20200034] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Lisa Miccio
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | | | - Ivana Kurelac
- Dipartimento di Scienze Mediche e Chirurgiche Università di Bologna Bologna Italy
- Centro di Ricerca Biomedica Applicata (CRBA) Università di Bologna Bologna Italy
| | - Massimiliano M. Villone
- Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale Università degli Studi di Napoli “Federico II” Napoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Vittorio Bianco
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Pasquale Memmolo
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Francesco Merola
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Martina Mugnano
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Mario Capasso
- CEINGE Biotecnologie Avanzate Naples Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche Università degli Studi di Napoli Federico II Naples Italy
| | - Achille Iolascon
- CEINGE Biotecnologie Avanzate Naples Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche Università degli Studi di Napoli Federico II Naples Italy
| | - Pier Luca Maffettone
- Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale Università degli Studi di Napoli “Federico II” Napoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| | - Pietro Ferraro
- CNR‐ISASI Institute of Applied Sciences and Intelligent Systems E. Caianiello Pozzuoli Italy
- NEAPoLIS, Numerical and Experimental Advanced Program on Liquids and Interface Systems Joint Research Center CNR ‐ Università degli Studi di Napoli “Federico II” Napoli Italy
| |
Collapse
|
19
|
Zamani Kouhpanji MR, Stadler BJH. A Guideline for Effectively Synthesizing and Characterizing Magnetic Nanoparticles for Advancing Nanobiotechnology: A Review. SENSORS (BASEL, SWITZERLAND) 2020; 20:E2554. [PMID: 32365832 PMCID: PMC7248791 DOI: 10.3390/s20092554] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023]
Abstract
The remarkable multimodal functionalities of magnetic nanoparticles, conferred by their size and morphology, are very important in resolving challenges slowing the progression of nanobiotechnology. The rapid and revolutionary expansion of magnetic nanoparticles in nanobiotechnology, especially in nanomedicine and therapeutics, demands an overview of the current state of the art for synthesizing and characterizing magnetic nanoparticles. In this review, we explain the synthesis routes for tailoring the size, morphology, composition, and magnetic properties of the magnetic nanoparticles. The pros and cons of the most popularly used characterization techniques for determining the aforementioned parameters, with particular focus on nanomedicine and biosensing applications, are discussed. Moreover, we provide numerous biomedical applications and highlight their challenges and requirements that must be met using the magnetic nanoparticles to achieve the most effective outcomes. Finally, we conclude this review by providing an insight towards resolving the persisting challenges and the future directions. This review should be an excellent source of information for beginners in this field who are looking for a groundbreaking start but they have been overwhelmed by the volume of literature.
Collapse
Affiliation(s)
- Mohammad Reza Zamani Kouhpanji
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bethanie J. H. Stadler
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
20
|
Chen X, Wang L, Lou J. Nanotechnology Strategies for the Analysis of Circulating Tumor DNA: A Review. Med Sci Monit 2020; 26:e921040. [PMID: 32200389 PMCID: PMC7111132 DOI: 10.12659/msm.921040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Circulating tumor DNA (ctDNA) describes the fragmented DNA released from tumor cells into the blood. The ctDNA may have the same genetic changes as the primary tumor. Currently, ctDNA has become a popular biomarker for diagnosis, treatment, real-time clinical response monitoring, and prognosis, for solid tumors. Detection of ctDNA is minimally invasive, and repeat sampling can easily be performed. However, due to its low quality and short DNA fragment length, ctDNA detection still faces challenges and requires highly sensitive analytical techniques. Recently, liquid biopsies for the analysis of circulating tumor cells (CTCs) and circulating tumor-derived exosomes have been studied, and nanotechnology techniques have rapidly developed. Compared to traditional analytical methods, these nanotechnology-based platforms have the advantages of sensitivity, multiplex detection, simplicity, miniaturization, and automation, which support their potential use in clinical practice. This review aims to discuss the recent nanotechnological strategies for ctDNA analysis and the design of reliable techniques for ctDNA detection and to identify the potential clinical applications.
Collapse
Affiliation(s)
- Xiaomin Chen
- Nano Biomedical Research Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China (mainland).,Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Lin Wang
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China (mainland)
| | - Jiatao Lou
- Department of Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China (mainland)
| |
Collapse
|
21
|
Siemer S, Wünsch D, Khamis A, Lu Q, Scherberich A, Filippi M, Krafft MP, Hagemann J, Weiss C, Ding GB, Stauber RH, Gribko A. Nano Meets Micro-Translational Nanotechnology in Medicine: Nano-Based Applications for Early Tumor Detection and Therapy. NANOMATERIALS 2020; 10:nano10020383. [PMID: 32098406 PMCID: PMC7075286 DOI: 10.3390/nano10020383] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/03/2020] [Accepted: 02/15/2020] [Indexed: 02/07/2023]
Abstract
Nanomaterials have great potential for the prevention and treatment of cancer. Circulating tumor cells (CTCs) are cancer cells of solid tumor origin entering the peripheral blood after detachment from a primary tumor. The occurrence and circulation of CTCs are accepted as a prerequisite for the formation of metastases, which is the major cause of cancer-associated deaths. Due to their clinical significance CTCs are intensively discussed to be used as liquid biopsy for early diagnosis and prognosis of cancer. However, there are substantial challenges for the clinical use of CTCs based on their extreme rarity and heterogeneous biology. Therefore, methods for effective isolation and detection of CTCs are urgently needed. With the rapid development of nanotechnology and its wide applications in the biomedical field, researchers have designed various nano-sized systems with the capability of CTCs detection, isolation, and CTCs-targeted cancer therapy. In the present review, we summarize the underlying mechanisms of CTC-associated tumor metastasis, and give detailed information about the unique properties of CTCs that can be harnessed for their effective analytical detection and enrichment. Furthermore, we want to give an overview of representative nano-systems for CTC isolation, and highlight recent achievements in microfluidics and lab-on-a-chip technologies. We also emphasize the recent advances in nano-based CTCs-targeted cancer therapy. We conclude by critically discussing recent CTC-based nano-systems with high therapeutic and diagnostic potential as well as their biocompatibility as a practical example of applied nanotechnology.
Collapse
Affiliation(s)
- Svenja Siemer
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Désirée Wünsch
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Aya Khamis
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Qiang Lu
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Arnaud Scherberich
- Laboratory of Tissue Engineering, Universitätspital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland (M.F.)
| | - Miriam Filippi
- Laboratory of Tissue Engineering, Universitätspital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland (M.F.)
| | - Marie Pierre Krafft
- Institut Charles Sadron (CNRS), University of Strasbourg, 23 rue du Loess, 67034 Strasbourg Cedex, France
| | - Jan Hagemann
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Carsten Weiss
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Postfach 3640, 76021 Karlsruhe, Germany
| | - Guo-Bin Ding
- Institute for Biotechnology, Shanxi University, No. 92 Wucheng Road, 030006 Taiyuan, China
| | - Roland H. Stauber
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
- Institute for Biotechnology, Shanxi University, No. 92 Wucheng Road, 030006 Taiyuan, China
- Correspondence: (R.H.S.); (A.G.); Tel.: +49-6131-176030 (A.G.)
| | - Alena Gribko
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
- Correspondence: (R.H.S.); (A.G.); Tel.: +49-6131-176030 (A.G.)
| |
Collapse
|
22
|
Wu X, Bai Z, Wang L, Cui G, Yang M, Yang Q, Ma B, Song Q, Tian D, Ceyssens F, Puers R, Kraft M, Zhao W, Wen L. Magnetic Cell Centrifuge Platform Performance Study with Different Microsieve Pore Geometries. SENSORS (BASEL, SWITZERLAND) 2019; 20:E48. [PMID: 31861791 PMCID: PMC6983067 DOI: 10.3390/s20010048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022]
Abstract
The detection and analysis of circulating tumor cells (CTCs) plays a crucial role in clinical practice. However, the heterogeneity and rarity of CTCs make their capture and separation from peripheral blood very difficult while maintaining their structural integrity and viability. We previously demonstrated the effectiveness of the Magnetic Cell Centrifuge Platform (MCCP), which combined the magnetic-labeling cell separation mechanism with the size-based method. In this paper, a comparison of the effectiveness of different microsieve pore geometries toward MCCP is demonstrated to improve the yield of the target cell capture. Firstly, models of a trapped cell with rectangular and circular pore geometries are presented to compare the contact force using finite element numerical simulations. The device performance is then evaluated with both constant pressure and constant flow rate experimental conditions. In addition, the efficient isolation of magnetically labeled Hela cells with red fluorescent proteins (target cells) from Hela cells with green fluorescent protein (background cells) is validated. The experimental results show that the circular sieves yield 97% purity of the target cells from the sample with a throughput of up to 2 μL/s and 66-fold sample enrichment. This finding will pave the way for the design of a higher efficient MCCP systems.
Collapse
Affiliation(s)
- Xinyu Wu
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Zhongyang Bai
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Lin Wang
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Guangchao Cui
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Mengzheng Yang
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Qing Yang
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Bo Ma
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
| | - Qinglin Song
- Qingdao Goertek Microelectronics Research Institute Co., Ltd., Qingdao 266104, China
| | - Dewen Tian
- Qingdao Goertek Microelectronics Research Institute Co., Ltd., Qingdao 266104, China
| | - Frederik Ceyssens
- ESAT-MICAS, KU Leuven, Kasteelpark Arenberg 10, B-3001 Leuven, Belgium
| | - Robert Puers
- ESAT-MICAS, KU Leuven, Kasteelpark Arenberg 10, B-3001 Leuven, Belgium
| | - Michael Kraft
- ESAT-MICAS, KU Leuven, Kasteelpark Arenberg 10, B-3001 Leuven, Belgium
| | - Weisheng Zhao
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| | - Lianggong Wen
- School of Microelectronics, Beihang University, Beijing 100191, China; (X.W.)
- Beihang-Goertek Joint Microelectronics Institute, Qingdao Research Institute, Beihang University, Qingdao 266104, China
| |
Collapse
|
23
|
Wang P, Sun S, Ma H, Sun S, Zhao D, Wang S, Liang X. Treating tumors with minimally invasive therapy: A review. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 108:110198. [PMID: 31923997 DOI: 10.1016/j.msec.2019.110198] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 09/01/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022]
Abstract
With high level of morbidity and mortality, tumor is one of the deadliest diseases worldwide. Aiming to tackle tumor, researchers have developed a lot of strategies. Among these strategies, the minimally invasive therapy (MIT) is very promising, for its capability of targeting tumor cells and resulting in a small incision or no incisions. In this review, we will first illustrate some mechanisms and characteristics of tumor metastasis from the primary tumor to the secondary tumor foci. Then, we will briefly introduce the history, characteristics, and advantages of some of the MITs. Finally, emphasis will be, respectively, focused on an overview of the state-of-the-art of the HIFU-, PDT-, PTT-and SDT-based anti-tumor strategies on each stage of tumor metastasis.
Collapse
Affiliation(s)
- Ping Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Huide Ma
- Ordos Center Hospital, Ordos, Inner Mongolia, 017000, China
| | - Sujuan Sun
- Ordos Center Hospital, Ordos, Inner Mongolia, 017000, China
| | - Duo Zhao
- Ordos Center Hospital, Ordos, Inner Mongolia, 017000, China
| | - Shumin Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
24
|
Farahavar G, Abolmaali SS, Gholijani N, Nejatollahi F. Antibody-guided nanomedicines as novel breakthrough therapeutic, diagnostic and theranostic tools. Biomater Sci 2019; 7:4000-4016. [PMID: 31355391 DOI: 10.1039/c9bm00931k] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent advances in nanotechnology, such as the development of various types of nanoparticles and hybrid nanomaterials, have revolutionized nanomedicine. The small size, customizable surface, enhanced solubility, and multi-functionality endow the nanoparticles with an ability to interact with complex cellular and biological functions in new ways. Furthermore, these systems can deliver drugs to specific tissues and provide a targeted therapy. For this purpose, different categories of molecules, particularly antibodies, have been used as ligands. Antibody-conjugated nanomaterials can significantly enhance the efficiency of nanomedicines, especially in the field of cancer. This review is focused on three major medical applications of antibody-conjugated nanomaterials, namely, therapeutic, diagnostic and theranostic applications. To provide comprehensive information on the topic and an overview of these hybrid nanomaterials for biomedical applications, a brief summary of nanomaterials and antibodies is given. Moreover, the review has depicted the potential applications of antibody-conjugated nanomaterials in different fields and their capabilities to empower nanomedicine, particularly in relation to the treatment and detection of malignancies.
Collapse
Affiliation(s)
- Ghazal Farahavar
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Nasser Gholijani
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Foroogh Nejatollahi
- Shiraz HIV/AIDS research center, Institute of health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
25
|
Liu C, Yang B, Chen X, Hu Z, Dai Z, Yang D, Zheng X, She X, Liu Q. Capture and separation of circulating tumor cells using functionalized magnetic nanocomposites with simultaneous in situ chemotherapy. NANOTECHNOLOGY 2019; 30:285706. [PMID: 30849773 DOI: 10.1088/1361-6528/ab0e25] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Circulating tumor cells (CTCs) are a type of rare cell that are firstly shed from solid tumors and then exist in the bloodstream. The effective capture and separation of CTCs has significant meaning in cancer diagnosis and prognosis. In this study, novel Fe3O4-FePt magnetic nanocomposites (Fe3O4-FePt MNCs) were constructed by integrating face centered cubic (fcc) FePt nanoparticles (NPs) onto the surface of the Fe3O4@SiO2 core. After further modification with NH2-PEG-COOH and the tumor-targeting molecule tLyP-1, the acquired Fe3O4-FePt MNCs possesses excellent biocompatibility and stability and could efficiently target and capture tLyP-1 receptor-positive CTCs. Based on the acidic microenvironment within cancer cells, the FePt layer could rapidly release active Fe2+ ions, which could catalyze H2O2 into reactive oxygen species (ROS) and further induce in situ apoptosis in cancer cells while having no distinct cytotoxicity to normal cells. Moreover, the Fe3O4@SiO2 core with its intrinsic magnetism has huge potential for the bioseparation of CTCs. The in vitro ROS fluorescence imaging experiments and cell capture and separation experiments indicated that the Fe3O4-FePt MNCs could specifically capture and separate cancer cells in the CTCs model and further induce in situ apoptosis. Therefore, the Fe3O4-FePt MNCs could serve as a promising multifunctional nanoseparator for efficiently capturing CTCs and simultaneously inducing in situ chemotherapy.
Collapse
Affiliation(s)
- Chunmiao Liu
- School of Materials Science and Engineering, Qingdao University, Qingdao 266071, People's Republic of China. Key Laboratory of Functional Nanomaterials and Technology in Universities of Shandong, Linyi University, Linyi 276000, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lu J, Pang J, Chen Y, Dong Q, Sheng J, Luo Y, Lu Y, Lin B, Liu T. Application of Microfluidic Chips in Separation and Analysis of Extracellular Vesicles in Liquid Biopsy for Cancer. MICROMACHINES 2019; 10:mi10060390. [PMID: 31212643 PMCID: PMC6630239 DOI: 10.3390/mi10060390] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/01/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are becoming a promising biomarker in liquid biopsy of cancer. Separation EV from cell culture medium or biofluids with high purity and quality remains a technique challenge. EV manipulation techniques based on microfluidics have been developed in the last decade. Microfluidic-based EV separation techniques developed so far can be classified into two categories: surface biomarker-dependent and size-dependent approaches. Microfluidic techniques allow the integration of EV separation and analysis on a single chip. Integrated EV separation and on-chip analysis have shown great potential in cancer diagnosis and monitoring treatment of responses. In this review, we discuss the development of microfluidic chips for EV separation and analysis. We also detail the clinical application of these microfluidic chips in the liquid biopsy of various cancers.
Collapse
Affiliation(s)
- Jin Lu
- College of Stomatology, Dalian Medical University, Dalian 116044, China.
| | - Jiushen Pang
- College of Stomatology, Dalian Medical University, Dalian 116044, China.
| | - Ying Chen
- College of Stomatology, Dalian Medical University, Dalian 116044, China.
| | - Qi Dong
- College of Stomatology, Dalian Medical University, Dalian 116044, China.
| | - Jiahao Sheng
- College of Stomatology, Dalian Medical University, Dalian 116044, China.
| | - Yong Luo
- Faculty of Chemical, Environmental and Biological Science and Technology, Dalian Technology University, Dalian 116044, China.
| | - Yao Lu
- Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116044, China.
| | - Bingcheng Lin
- Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116044, China.
| | - Tingjiao Liu
- College of Stomatology, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
27
|
Gribko A, Künzel J, Wünsch D, Lu Q, Nagel SM, Knauer SK, Stauber RH, Ding GB. Is small smarter? Nanomaterial-based detection and elimination of circulating tumor cells: current knowledge and perspectives. Int J Nanomedicine 2019; 14:4187-4209. [PMID: 31289440 PMCID: PMC6560927 DOI: 10.2147/ijn.s198319] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are disseminated cancer cells. The occurrence and circulation of CTCs seem key for metastasis, still the major cause of cancer-associated deaths. As such, CTCs are investigated as predictive biomarkers. However, due to their rarity and heterogeneous biology, CTCs’ practical use has not made it into the clinical routine. Clearly, methods for the effective isolation and reliable detection of CTCs are urgently needed. With the development of nanotechnology, various nanosystems for CTC isolation and enrichment and CTC-targeted cancer therapy have been designed. Here, we summarize the relationship between CTCs and tumor metastasis, and describe CTCs’ unique properties hampering their effective enrichment. We comment on nanotechnology-based systems for CTC isolation and recent achievements in microfluidics and lab-on-a-chip technologies. We discuss recent advances in CTC-targeted cancer therapy exploiting the unique properties of nanomaterials. We conclude by introducing developments in CTC-directed nanosystems and other advanced technologies currently in (pre)clinical research.
Collapse
Affiliation(s)
- Alena Gribko
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Julian Künzel
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Désirée Wünsch
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Qiang Lu
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Sophie Madeleine Nagel
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Shirley K Knauer
- Department of Molecular Biology II, Center for Medical Biotechnology (ZMB)/Center for Nanointegration (CENIDE), University Duisburg-Essen, Essen 45117, Germany
| | - Roland H Stauber
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Guo-Bin Ding
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ; .,Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, People's Republic of China,
| |
Collapse
|
28
|
Petanidis S, Kioseoglou E, Salifoglou A. Metallodrugs in Targeted Cancer Therapeutics: Aiming at Chemoresistance- related Patterns and Immunosuppressive Tumor Networks. Curr Med Chem 2019; 26:607-623. [DOI: 10.2174/0929867324666171116125908] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022]
Abstract
Tumor cell chemoresistance is a major challenge in cancer therapeutics. Major
select metal-based drugs are potent anticancer mediators yet they exhibit adverse sideeffects
and are efficient against limited types of malignancies. A need, therefore, arises
for novel metallodrugs with improved efficacy and decreased toxicity. Enhancement of
antitumor drugs based on anticancer metals is currently a very active research field, with
considerable efforts having been made toward elucidating the mechanisms of immune action
of complex metalloforms and optimizing their immunoregulatory bioactivity through
appropriate synthetic structural modification(s) and encapsulation in suitable nanocarriers,
thereby enhancing their selectivity, specificity, stability, and bioactivity. In that respect,
comprehending the molecular factors involved in drug resistance and immune response
may help us develop new approaches toward more promising chemotherapies, reducing
the rate of relapse and overcoming chemoresistance. In this review, a) molecular immunerelated
mechanisms in the tumor microenvironment, responsible for lower drug sensitivity
and tumor relapse, along with b) strategies for reversing drug resistance and targeting
immunosuppressive tumor networks, while concurrently optimizing the design of complex
metalloforms bearing anti-tumor activity, are discussed in an effort to identify and
overcome chemoresistance mechanisms for effective tumor immunotherapeutic approaches.
Collapse
Affiliation(s)
- Savvas Petanidis
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Efrosini Kioseoglou
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Athanasios Salifoglou
- Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| |
Collapse
|
29
|
Li F, Xu H, Sun P, Hu Z, Aguilar ZP. Size effects of magnetic beads in circulating tumour cells magnetic capture based on streptavidin-biotin complexation. IET Nanobiotechnol 2019; 13:6-11. [PMID: 30964030 PMCID: PMC8675959 DOI: 10.1049/iet-nbt.2018.5104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 05/09/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2024] Open
Abstract
Circulating tumour cells (CTCs) draw significant attention as a promising biomarker for cancer prognosis, status monitoring, and metastasis diagnosis. However, the concentration of CTCs in peripheral blood is usually extremely low, thereby requiring enrichment followed by isolation of CTCs prior to detection. An immunomagnetic separation is a promising tool for CTCs enrichment. In this study, a cost-effective magnetic separation method, based on streptavidin-biotin complexation, was developed and the effects of magnetic beads' size in CTCs capture were compared. Magnetic nanobeads which were 25 nm in diameter lead to highest capture efficiency (82.2%) compared with 150 nm magnetic beads and 1 µm microbeads. Based on the streptavidin-biotin system, 25 nm magnetic nanobeads could capture model CTCs over 80% efficiency even at concentrations as low as ∼25 cells/mL that may represent the actual level of CTCs in peripheral blood of cancer patients. Furthermore, the isolated cells remained robust and healthy showing insignificant changes in morphology and behaviour when cultured for 24 h immediately after capture and isolation. The magnetic nanobeads based on streptavidin-biotin complexation showed promise for the easy and efficient capture and isolation of healthy CTCs for further diagnosis and analysis.
Collapse
Affiliation(s)
- Fulai Li
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, People's Republic of China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, People's Republic of China.
| | - Pingfeng Sun
- The Obstetrics and Gynecology Department of Second Affiliated Hospital of Nanchang University, Nanchang University, 2 Min De Road, Nanchang 330006, People's Republic of China
| | - Zhibin Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, People's Republic of China
| | | |
Collapse
|
30
|
Zhang Y, Mi X, Tan X, Xiang R. Recent Progress on Liquid Biopsy Analysis using Surface-Enhanced Raman Spectroscopy. Theranostics 2019; 9:491-525. [PMID: 30809289 PMCID: PMC6376192 DOI: 10.7150/thno.29875] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/30/2018] [Indexed: 12/12/2022] Open
Abstract
Traditional tissue biopsy is limited in understanding heterogeneity and dynamic evolution of tumors. Instead, analyzing circulating cancer markers in various body fluids, commonly referred to as "liquid biopsy", has recently attracted remarkable interest for their great potential to be applied in non-invasive early cancer screening, tumor progression monitoring and therapy response assessment. Among the various approaches developed for liquid biopsy analysis, surface-enhanced Raman spectroscopy (SERS) has emerged as one of the most powerful techniques based on its high sensitivity, specificity, tremendous spectral multiplexing capacity for simultaneous target detection, as well as its unique capability for obtaining intrinsic fingerprint spectra of biomolecules. In this review, we will first briefly explain the mechanism of SERS, and then introduce recently reported SERS-based techniques for detection of circulating cancer markers including circulating tumor cells, exosomes, circulating tumor DNAs, microRNAs and cancer-related proteins. Cancer diagnosis based on SERS analysis of bulk body fluids will also be included. In the end, we will summarize the "state of the art" technologies of SERS-based platforms and discuss the challenges of translating them into clinical settings.
Collapse
Affiliation(s)
- Yuying Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, 300071 Tianjin, China
| | | | | | | |
Collapse
|
31
|
Kwizera EA, O'Connor R, Vinduska V, Williams M, Butch ER, Snyder SE, Chen X, Huang X. Molecular Detection and Analysis of Exosomes Using Surface-Enhanced Raman Scattering Gold Nanorods and a Miniaturized Device. Theranostics 2018; 8:2722-2738. [PMID: 29774071 PMCID: PMC5957005 DOI: 10.7150/thno.21358] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 01/09/2018] [Indexed: 12/24/2022] Open
Abstract
Exosomes are a potential source of cancer biomarkers. Probing tumor-derived exosomes can offer a potential non-invasive way to diagnose cancer, assess cancer progression, and monitor treatment responses. Novel molecular methods would facilitate exosome analysis and accelerate basic and clinical exosome research. Methods: A standard gold-coated glass microscopy slide was used to develop a miniaturized affinity-based device to capture exosomes in a target-specific manner with the assistance of low-cost 3-D printing technology. Gold nanorods coated with QSY21 Raman reporters were used as the label agent to quantitatively detect the target proteins based on surface enhanced Raman scattering spectroscopy. The expressions of several surface protein markers on exosomes from conditioned culture media of breast cancer cells and from HER2-positive breast cancer patients were quantitatively measured. The data was statistically analyzed and compared with healthy controls. Results: A miniaturized 17 × 5 Au array device with 2-mm well size was fabricated to capture exosomes in a target-specific manner and detect the target proteins on exosomes with surface enhanced Raman scattering gold nanorods. This assay can specifically detect exosomes with a limit of detection of 2×106 exosomes/mL and analyze over 80 purified samples on a single device within 2 h. Using the assay, we have showed that exosomes derived from MDA-MB-231, MDA-MB-468, and SKBR3 breast cancer cells give distinct protein profiles compared to exosomes derived from MCF12A normal breast cells. We have also showed that exosomes in the plasma from HER2-positive breast cancer patients exhibit significantly (P ≤ 0.01) higher level of HER2 and EpCAM than those from healthy donors. Conclusion: We have developed a simple, inexpensive, highly efficient, and portable Raman exosome assay for detection and protein profiling of exosomes. Using the assay and model exosomes from breast cancer cells, we have showed that exosomes exhibit diagnostic surface protein markers, reflecting the protein profile of their donor cells. Through proof-of-concept studies, we have identified HER2 and EpCAM biomarkers on exosomes in plasma from HER2-positive breast cancer patients, suggesting the diagnostic potential of these markers for breast cancer diagnostics. This assay would accelerate exosome research and pave a way to the development of novel cancer liquid biopsy for cancer detection and monitoring.
Collapse
Affiliation(s)
| | - Ryan O'Connor
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| | - Vojtech Vinduska
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| | - Melody Williams
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| | - Elizabeth R. Butch
- Diagnostics Imaging Department, St Jude Children's Research Hospital, Memphis, TN 38105
| | - Scott E. Snyder
- Diagnostics Imaging Department, St Jude Children's Research Hospital, Memphis, TN 38105
| | - Xiang Chen
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN 38105
| | - Xiaohua Huang
- Department of Chemistry, The University of Memphis, Memphis, TN 38152
| |
Collapse
|
32
|
Chen F, Haddour N, Frenea-Robin M, Chevolot Y, Monnier V. Polyamidoamine Dendrimers as Crosslinkers for Efficient Electron Transfer between Redox Probes onto Magnetic Nanoparticles. ChemistrySelect 2018. [DOI: 10.1002/slct.201703135] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Feixiong Chen
- Université de Lyon, Ecole Centrale de Lyon, UMR CNRS 5270; Institut des Nanotechnologies de Lyon, UMR CNRS 5270; Ecully, F- 69130 France
| | - Naoufel Haddour
- Université de Lyon, Ecole Centrale de Lyon, CNRS, UMR 5005; Laboratoire Ampère; Ecully, F- 69130 France
| | - Marie Frenea-Robin
- Université de Lyon, Université Lyon 1, CNRS, UMR 5005; Laboratoire Ampère; Villeurbanne, F- 69622 France
| | - Yann Chevolot
- Université de Lyon, Ecole Centrale de Lyon, UMR CNRS 5270; Institut des Nanotechnologies de Lyon, UMR CNRS 5270; Ecully, F- 69130 France
| | - Virginie Monnier
- Université de Lyon, Ecole Centrale de Lyon, UMR CNRS 5270; Institut des Nanotechnologies de Lyon, UMR CNRS 5270; Ecully, F- 69130 France
| |
Collapse
|
33
|
Rauta PR, Hallur PM, Chaubey A. Gold nanoparticle-based rapid detection and isolation of cells using ligand-receptor chemistry. Sci Rep 2018; 8:2893. [PMID: 29440656 PMCID: PMC5811494 DOI: 10.1038/s41598-018-21068-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/22/2018] [Indexed: 01/07/2023] Open
Abstract
Identification and isolation of low-frequency cells of interest from a heterogeneous cell mixture is an important aspect of many diagnostic applications (including enumeration of circulating tumor cells) and is integral to various assays in (cancer) biology. Current techniques typically require expensive instrumentation and are not amenable to high throughput. Here, we demonstrate a simple and effective platform for cell detection and isolation using gold nanoparticles (Au NPs) conjugated with hyaluronic acid (HA) i.e. Au-PEG-HA NPs. The proposed platform exploits ligand-receptor chemistry to detect/isolate cells with high specificity and efficiency. When the Au-PEG-HA NPs come in contact with cells that express CD44 (the receptor for HA), a clear colorimetric change occurs (along with an accompanying SPR peak shift from 521 nm to 559 nm) in the solution due to NPs-cell interaction. This clearly discernible, colorimetric change can be leveraged by point-of-care devices employed in diagnostic applications. Finally, we show that we can successfully isolate viable cells from a heterogeneous cell population (including from human blood samples) with high specificity, which can be used in further downstream applications. The developed NPs-based platform can be a convenient and cost-efficient alternative for diagnostic applications and for cell isolation or sorting in research laboratories.
Collapse
Affiliation(s)
- Pradipta Ranjan Rauta
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research, NH Health City, Bangalore, 560 099, India
| | - Pavan M Hallur
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research, NH Health City, Bangalore, 560 099, India
| | - Aditya Chaubey
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research, NH Health City, Bangalore, 560 099, India.
| |
Collapse
|
34
|
Austin RG, Huang TJ, Wu M, Armstrong AJ, Zhang T. Clinical utility of non-EpCAM based circulating tumor cell assays. Adv Drug Deliv Rev 2018; 125:132-142. [PMID: 29366804 DOI: 10.1016/j.addr.2018.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/14/2018] [Accepted: 01/17/2018] [Indexed: 01/06/2023]
Abstract
Methods enabling the isolation, detection, and characterization of circulating tumor cells (CTCs) in blood have clear potential to facilitate precision medicine approaches in patients with cancer, not only for prognostic purposes but also for prediction of the benefits of specific therapies in oncology. However, current CTC assays, which capture CTCs based on expression of epithelial cell adhesion molecule (EpCAM), fail to capture cells from de-differentiated tumors and carcinomas undergoing loss of the epithelial phenotype during the invasion/metastatic process. To address this limitation, many groups are developing non-EpCAM based CTC assays that incorporate nanotechnology to improve test sensitivity for rare but important cells that may otherwise go undetected, and therefore may improve upon clinical utility. In this review, we outline emerging non-EpCAM based CTC assays utilizing nanotechnology approaches for CTC capture or characterization, including dendrimers, magnetic nanoparticles, gold nanoparticles, negative selection chip or software-based on-slide methods, and nano-scale substrates. In addition, we address challenges that remain for the clinical translation of these platforms.
Collapse
|
35
|
Kuai JH, Wang Q, Zhang AJ, Zhang JY, Chen ZF, Wu KK, Hu XZ. Epidermal growth factor receptor-targeted immune magnetic liposomes capture circulating colorectal tumor cells efficiently. World J Gastroenterol 2018; 24:351-359. [PMID: 29391757 PMCID: PMC5776396 DOI: 10.3748/wjg.v24.i3.351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/27/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To compare the capacity of newly developed epidermal growth factor receptor (EGFR)-targeted immune magnetic liposomes (EILs) vs epithelial cell adhesion molecule (EpCAM) immunomagnetic beads to capture colorectal circulating tumor cells (CTCs).
METHODS EILs were prepared using a two-step method, and the magnetic and surface characteristics were confirmed. The efficiency of capturing colorectal CTCs as well as the specificity were compared between EILs and EpCAM magnetic beads.
RESULTS The obtained EILs had a lipid nanoparticle structure similar to cell membrane. Improved binding with cancer cells was seen in EILs compared with the method of coupling nano/microspheres with antibody. The binding increased as the contact time extended. Compared with EpCAM immunomagnetic beads, EILs captured more CTCs in peripheral blood from colorectal cancer patients. The captured cells showed consistency with clinical diagnosis and pathology. Mutation analysis showed same results between captured CTCs and cancer tissues.
CONCLUSION EGFR antibody-coated magnetic liposomes show high efficiency and specificity in capturing colorectal CTCs.
Collapse
Affiliation(s)
- Jing-Hua Kuai
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266035, Shandong Province, China
| | - Qing Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266035, Shandong Province, China
| | - Ai-Jun Zhang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266035, Shandong Province, China
| | - Jing-Yu Zhang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266035, Shandong Province, China
| | - Zheng-Feng Chen
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266035, Shandong Province, China
| | - Kang-Kang Wu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Qingdao 266035, Shandong Province, China
| | - Xiao-Zhen Hu
- Department of General Surgery, Qilu Hospital of Shandong University, Qingdao 266035, Shandong Province, China
| |
Collapse
|
36
|
Huang Q, Wang Y, Chen X, Wang Y, Li Z, Du S, Wang L, Chen S. Nanotechnology-Based Strategies for Early Cancer Diagnosis Using Circulating Tumor Cells as a Liquid Biopsy. Nanotheranostics 2018; 2:21-41. [PMID: 29291161 PMCID: PMC5743836 DOI: 10.7150/ntno.22091] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/10/2017] [Indexed: 12/11/2022] Open
Abstract
Circulating tumor cells (CTCs) are cancer cells that shed from a primary tumor and circulate in the bloodstream. As a form of “tumor liquid biopsy”, CTCs provide important information for the mechanistic investigation of cancer metastasis and the measurement of tumor genotype evolution during treatment and disease progression. However, the extremely low abundance of CTCs in the peripheral blood and the heterogeneity of CTCs make their isolation and characterization major technological challenges. Recently, nanotechnologies have been developed for sensitive CTC detection; such technologies will enable better cell and molecular characterization and open up a wide range of clinical applications, including early disease detection and evaluation of treatment response and disease progression. In this review, we summarize the nanotechnology-based strategies for CTC isolation, including representative nanomaterials (such as magnetic nanoparticles, gold nanoparticles, silicon nanopillars, nanowires, nanopillars, carbon nanotubes, dendrimers, quantum dots, and graphene oxide) and microfluidic chip technologies that incorporate nanoroughened surfaces and discuss their key challenges and perspectives in CTC downstream analyses, such as protein expression and genetic mutations that may reflect tumor aggressiveness and patient outcome.
Collapse
Affiliation(s)
- Qinqin Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, and Brain Center, Zhongnan Hospital, and Medical Research Institute, Wuhan University, Wuhan 430072, China
| | - Yin Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, and Brain Center, Zhongnan Hospital, and Medical Research Institute, Wuhan University, Wuhan 430072, China
| | - Xingxiang Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, and Brain Center, Zhongnan Hospital, and Medical Research Institute, Wuhan University, Wuhan 430072, China
| | - Yimeng Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, and Brain Center, Zhongnan Hospital, and Medical Research Institute, Wuhan University, Wuhan 430072, China
| | - Zhiqiang Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, and Brain Center, Zhongnan Hospital, and Medical Research Institute, Wuhan University, Wuhan 430072, China
| | - Shiming Du
- Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Lianrong Wang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, and Brain Center, Zhongnan Hospital, and Medical Research Institute, Wuhan University, Wuhan 430072, China
| | - Shi Chen
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery of Ministry of Education, School of Pharmaceutical Sciences, and Brain Center, Zhongnan Hospital, and Medical Research Institute, Wuhan University, Wuhan 430072, China
| |
Collapse
|
37
|
Yan S, Chen P, Zeng X, Zhang X, Li Y, Xia Y, Wang J, Dai X, Feng X, Du W, Liu BF. Integrated Multifunctional Electrochemistry Microchip for Highly Efficient Capture, Release, Lysis, and Analysis of Circulating Tumor Cells. Anal Chem 2017; 89:12039-12044. [DOI: 10.1021/acs.analchem.7b02469] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Shuangqian Yan
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xuemei Zeng
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xian Zhang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yun Xia
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jie Wang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaofang Dai
- Cancer
Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaojun Feng
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wei Du
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics—Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
38
|
Duan X, Liu Z, Xu S. [Research Progresses of Circulating Tumor Cells in Diagnosis and Treatment of
Early Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2017; 20:703-709. [PMID: 29061218 PMCID: PMC5972993 DOI: 10.3779/j.issn.1009-3419.2017.10.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
循环肿瘤细胞(circulating tumor cells, CTCs)作为液体活检的一种重要类型,在肺癌的筛查诊断、疗效评估、术后监测与预后判断等方面显示出越来越丰富的临床价值。随着对肺癌高危人群筛查工作的进展,大量肺小结节患者被检出,但是肺小结节不等于肺癌,而且据统计良性比例达90%-95%,这使得该部分患者在首次就诊时的良恶性鉴别诊断成为临床医生面临着的新的机遇与挑战。CTCs检测技术的不断进步与完善,是否可以在早期肺癌的鉴别诊断中发挥更大的作用,此外,它是否对早期肺癌手术治疗时的操作具有指导意义,这还需要进一步科研探索,以期将来实现临床转化。
Collapse
Affiliation(s)
- Xinchun Duan
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Zhidong Liu
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Shaofa Xu
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| |
Collapse
|
39
|
Sun D, Chen Z, Wu M, Zhang Y. Nanomaterial-based Microfluidic Chips for the Capture and Detection of Circulating Tumor Cells. Nanotheranostics 2017; 1:389-402. [PMID: 29071201 PMCID: PMC5647762 DOI: 10.7150/ntno.21268] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/25/2017] [Indexed: 01/27/2023] Open
Abstract
Circulating tumor cells (CTCs), a type of cancer cells that spreads from primary or metastatic tumors into the bloodstream, can lead to a new fatal metastasis. As a new type of liquid biopsy, CTCs have become a hot pursuit and detection of CTCs offers the possibility for early diagnosis of cancers, earlier evaluation of chemotherapeutic efficacy and cancer recurrence, and choice of individual sensitive anti-cancer drugs. The fundamental challenges of capturing and characterizing CTCs are the extremely low number of CTCs in the blood and the intrinsic heterogeneity of CTCs. A series of microfluidic devices have been proposed for the analysis of CTCs with automation capability, precise flow behaviors, and significant advantages over the conventional larger scale systems. This review aims to provide in-depth insights into CTCs analysis, including various nanomaterial-based microfluidic chips for the capture and detection of CTCs based on the specific biochemical and physical properties of CTCs. The current developmental trends and promising research directions in the establishment of microfluidic chips for the capture and detection of CTCs are also discussed.
Collapse
Affiliation(s)
- Duanping Sun
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Zuanguang Chen
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| | - Minhao Wu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, P. R. China
| | - Yuanqing Zhang
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, P. R. China
| |
Collapse
|
40
|
Abstract
Circulating tumor cells are a hallmark of cancer metastasis which accounts for approximately 90% of all cancer-related deaths. Their detection and characterization have significant implications in cancer biology and clinical practice. However, CTCs are rare cells and consist of heterogeneous subpopulations, requiring highly sensitive and specific techniques to identify and isolate them with high efficiency. Nanomaterials, with unique structural and functional properties, have shown strong promise to meet the challenging demands. In this review, we discuss CTC capture and therapeutic targeting, emphasizing the significance of the nanomaterials being used for this purpose. The next generation of therapy for metastatic cancer may well involve capturing and even directly neutralizing CTCs using nanomaterials.
Collapse
Affiliation(s)
- Zhenjiang Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235 USA
| | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235 USA
| |
Collapse
|
41
|
Orza A, Wu H, Xu Y, Lu Q, Mao H. One-Step Facile Synthesis of Highly Magnetic and Surface Functionalized Iron Oxide Nanorods for Biomarker-Targeted Applications. ACS APPLIED MATERIALS & INTERFACES 2017; 9:20719-20727. [PMID: 28513139 PMCID: PMC8898331 DOI: 10.1021/acsami.7b02575] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We report a one-step method for facile and sustainable synthesis of magnetic iron oxide nanorods (or IONRs) with mean lengths ranging from 25 to 50 nm and mean diameters ranging from 5 to 8 nm. The prepared IONRs are highly stable in aqueous media and can be surface functionalized for biomarker-targeted applications. This synthetic strategy involves the reaction of iron(III) acetylacetonate with polyethyleneimine in the presence of oleylamine and phenyl ether, followed by thermal decomposition. Importantly, the length and diameter as well as the aspect ratio of the prepared IONRs can be controlled by modulating the reaction parameters. We show that the resultant IONRs exhibit stronger magnetic properties compared to those of the widely used spherical iron oxide nanoparticles (IONPs) at the same iron content. The increased magnetic properties are dependent on the aspect ratio, with the magnetic saturation gradually increasing from 10 to 75 emu g-1 when increasing length of the IONRs, 5 nm in diameter, from 25 to 50 nm. The magnetic resonance imaging (MRI) contrast-enhancing effect, as measured in terms of the transverse relaxivity, r2, increased from 670.6 to 905.5 mM-1 s-1, when increasing the length from 25 to 50 nm. When applied to the immunomagnetic cell separation of the transferrin receptor (TfR)-overexpressed medulloblastoma cells using transferrin (Tf) as the targeting ligand, Tf-conjugated IONRs can capture 92 ± 3% of the targeted cells under a given condition (2.0 × 104 cells/mL, 0.2 mg Fe/mL concentration of magnetic materials, and 2.5 min of incubation time) compared to only 37 ± 2% when using the spherical IONPs, and 14 ± 2% when using commercially available magnetic beads, significantly improving the efficiency of separating the targeted cells.
Collapse
Affiliation(s)
- Anamaria Orza
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30329, United States
- Center for Systems Imaging, Emory University School of Medicine, Atlanta, Georgia 30329, United States
| | - Hui Wu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30329, United States
- Center for Systems Imaging, Emory University School of Medicine, Atlanta, Georgia 30329, United States
| | - Yaolin Xu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30329, United States
- Center for Systems Imaging, Emory University School of Medicine, Atlanta, Georgia 30329, United States
| | - Qiong Lu
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30329, United States
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P. R. China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia 30329, United States
- Center for Systems Imaging, Emory University School of Medicine, Atlanta, Georgia 30329, United States
| |
Collapse
|
42
|
Abstract
Background Immunotherapy consists of activating the patient’s immune system to fight cancer and has the great potential of preventing future relapses thanks to immunological memory. A great variety of strategies have emerged to harness the immune system against tumors, from the administration of immunomodulatory agents that activate immune cells, to therapeutic vaccines or infusion of previously activated cancer-specific T cells. However, despite great recent progress many difficulties still remain, which prevent the widespread use of immunotherapy. Some of these limitations include: systemic toxicity, weak immune cellular responses or persistence over time and most ultimately costly and time-consuming procedures. Main body Synthetic and natural biomaterials hold great potential to address these hurdles providing biocompatible systems capable of targeted local delivery, co-delivery, and controlled and/or sustained release. In this review we discuss some of the bioengineered solutions and approaches developed so far and how biomaterials can be further implemented to help and shape the future of cancer immunotherapy. Conclusion The bioengineering strategies here presented constitute a powerful toolkit to develop safe and successful novel cancer immunotherapies.
Collapse
|
43
|
Wikman H, Pantel K. Nanoplatforms for Circulating Tumor Cell Detection in Lung Cancer. Clin Chem 2017; 63:1318-1320. [PMID: 28588120 DOI: 10.1373/clinchem.2017.272682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/09/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Harriet Wikman
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
44
|
Messaritakis I, Stoltidis D, Kotsakis A, Dermitzaki EK, Koinis F, Lagoudaki E, Koutsopoulos A, Politaki E, Apostolaki S, Souglakos J, Georgoulias V. TTF-1- and/or CD56-positive Circulating Tumor Cells in patients with small cell lung cancer (SCLC). Sci Rep 2017; 7:45351. [PMID: 28349943 PMCID: PMC5368597 DOI: 10.1038/srep45351] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/22/2017] [Indexed: 11/17/2022] Open
Abstract
The aim of the study was to evaluate the phenotypic CTCs heterogeneity (TTF-1+ and/or CD56+) in SCLC patients and correlate it with the CellSearch. Peripheral blood was obtained from 108 consecutive patients. CTCs were detected by CellSearch and double-immunofluorescence using anti-CD45, anti-TTF-1 and anti-CD56 antibodies. Before chemotherapy TTF-1+/CD45−, CD56+/CD45− and TTF-1+/CD56+ CTCs were detected in 66(61.1%), 55(50.9%) and 46(42.6%) patients, respectively; 60.2% of patients were CellSearch+. Among the 22 patients with 0 CTCs/7.5 ml on CellSearch, TTF-1+/CD45−, CD56+/CD45− and TTF-1+/CD56+ CTCs were detected in 8(36.4%), 6(27.3) and 6(27.3%) patients, respectively; no CK+/EpCAM+ or TTF1+/EpCAM+ CTCs were detected in these patients. One-chemotherapy cycle decreased both the number of positive patients (p < 0.001) and their CTC number (p < 0.001), irrespectively of their phenotype and the detection method. The incidence and number of the different CTC subpopulations on PD, was significantly increased at their baseline levels. Multivariate analysis revealed that the increased number of CTCs at baseline and on PD were significantly associated with decreased PFS (p = 0.048) and OS (p = 0.041), respectively. There is an important CTC heterogeneity in such patients according to the expression of TTF-1 and CD56 which could detect EpCAM− CTC subpopulations and, thus, undetectable by CellSearch. These CTC subpopulations are dynamically correlated with treatment efficacy and disease-progression.
Collapse
Affiliation(s)
| | - Dimitris Stoltidis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Athanasios Kotsakis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece.,Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| | | | - Fillipos Koinis
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Eleni Lagoudaki
- Pathology, University General Hospital of Heraklion, Crete, Greece
| | | | - Eleni Politaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - Stella Apostolaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| | - John Souglakos
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece.,Department of Medical Oncology, University General Hospital of Heraklion, Crete, Greece
| | - Vassilis Georgoulias
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete, Crete, Greece
| |
Collapse
|
45
|
Ming Y, Li Y, Xing H, Luo M, Li Z, Chen J, Mo J, Shi S. Circulating Tumor Cells: From Theory to Nanotechnology-Based Detection. Front Pharmacol 2017; 8:35. [PMID: 28203204 PMCID: PMC5285331 DOI: 10.3389/fphar.2017.00035] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/17/2017] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells with stem-cell properties are regarded as tumor initiating cells. Sharing stem-cell properties, circulating tumor cells (CTCs) are responsible for the development of metastasis, which significant affects CTC analysis in clinical practice. Due to their extremely low occurrence in blood, however, it is challenging to enumerate and analyze CTCs. Nanotechnology is able to address the problems of insufficient capture efficiency and low purity of CTCs owing to the unique structural and functional properties of nanomaterials, showing strong promise for CTC isolation and detection. In this review, we discuss the role of stem-like CTCs in metastases, provide insight into recent progress in CTC isolation and detection approaches using various nanoplatforms, and highlight the role of nanotechnology in the advancement of CTC research.
Collapse
Affiliation(s)
- Yue Ming
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University Chongqing, China
| | - Yuanyuan Li
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University Chongqing, China
| | - Haiyan Xing
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University Chongqing, China
| | - Minghe Luo
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University Chongqing, China
| | - Ziwei Li
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University Chongqing, China
| | - Jianhong Chen
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University Chongqing, China
| | - Jingxin Mo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen UniversityGuangzhou, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Sanjun Shi
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University Chongqing, China
| |
Collapse
|
46
|
Li R, Liu B, Gao J. The application of nanoparticles in diagnosis and theranostics of gastric cancer. Cancer Lett 2016; 386:123-130. [PMID: 27845158 DOI: 10.1016/j.canlet.2016.10.032] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/12/2016] [Accepted: 10/22/2016] [Indexed: 02/07/2023]
Abstract
Gastric cancer is the fourth most common cancer and the second leading cause of cancer related death worldwide. For the diagnosis of gastric cancer, apart from regular systemic imaging, the locoregional imaging is also of great importance. Moreover, there are still other ways for the detecting of gastric cancer, including the early detection of gastric cancer by endoscopy, the detection of gastric-cancer related biomarkers and the detection of circulating tumor cells (CTCs) of gastric cancer. However, conventional diagnostic methods are usually lack of specificity and sensitivity. Nanoparticles provide many benefits in the diagnosis of gastric cancer. Besides, nanoparticles are capable of integrating the functions of diagnosis and treatment together (theranostics). In this paper, we reviewed the applications of nanoparticles in diagnosis and theranostics of gastric cancer in the above mentioned aspects.
Collapse
Affiliation(s)
- Rutian Li
- The Comprehensive Cancer Center of Drum-Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, PR China; Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, PR China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum-Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, PR China.
| | - Jiahui Gao
- The Comprehensive Cancer Center of Drum-Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, PR China; Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, PR China
| |
Collapse
|
47
|
Vatansever F, Hamblin MR. Surface-initiated ring-opening metathesis polymerization (SI-ROMP) to attach a tethered organic corona onto CdSe/ZnS core/shell quantum dots. JOURNAL OF NANOPARTICLE RESEARCH : AN INTERDISCIPLINARY FORUM FOR NANOSCALE SCIENCE AND TECHNOLOGY 2016; 18:302. [PMID: 28360819 PMCID: PMC5367471 DOI: 10.1007/s11051-016-3328-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/05/2016] [Indexed: 06/07/2023]
Abstract
Core-shell CdSe/ZnS quantum dots (QDs) are useful as tunable photostable fluorophores for multiple applications in industry, biology, and medicine. However, to achieve the optimum optical properties, the surface of the QDs must be passivated to remove charged sites that might bind extraneous substances and allow aggregation. Here we describe a method of growing an organic polymer corona onto the QD surface using the bottom-up approach of surface-initiated ring-opening metathesis polymerization (SI-ROMP) with Grubbs catalyst. CdSe/ZnS QDs were first coated with mercaptopropionic acid by displacing the original trioctylphosphine oxide layer, and then reacted with 7-octenyl dimethyl chlorosilane. The resulting octenyl double bonds allowed the attachment of ruthenium alkylidene groups as a catalyst. A subsequent metathesis reaction with strained bicyclic monomers (norbornene-dicarbonyl chloride (NDC), and a mixture of NDC and norbornenylethylisobutyl-polyhedral oligomeric silsesquioxane (norbornoPOSS)) allowed the construction of tethered organic homo-polymer or co-polymer layers onto the QD. Compounds were characterized by FT-IR, 1H-NMR, X-ray photoelectron spectroscopy, differential scanning calorimetry, and transmission electron microscopy. Atomic force microscopy showed that the coated QDs were separate and non-aggregated with a range of diameter of 48-53 nm.
Collapse
Affiliation(s)
- Fatma Vatansever
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA. Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA. Harvard-MIT Division of Health Sciences and Technology, Boston, MA 02139, USA
| |
Collapse
|
48
|
Recent insights into the development of nanotechnology to detect circulating tumor cells. Trends Analyt Chem 2016. [DOI: 10.1016/j.trac.2016.05.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
Chaffin E, O'Connor RT, Barr J, Huang X, Wang Y. Dependence of SERS enhancement on the chemical composition and structure of Ag/Au hybrid nanoparticles. J Chem Phys 2016; 145:054706. [PMID: 27497571 PMCID: PMC4975750 DOI: 10.1063/1.4960052] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/18/2016] [Indexed: 02/03/2023] Open
Abstract
Noble metal nanoparticles (NPs) such as silver (Ag) and gold (Au) have unique plasmonic properties that give rise to surface enhanced Raman scattering (SERS). Generally, Ag NPs have much stronger plasmonic properties and, hence, provide stronger SERS signals than Au NPs. However, Ag NPs lack the chemical stability and biocompatibility of comparable Au NPs and typically exhibit the most intense plasmonic resonance at wavelengths much shorter than the optimal spectral region for many biomedical applications. To overcome these issues, various experimental efforts have been devoted to the synthesis of Ag/Au hybrid NPs for the purpose of SERS detections. However, a complete understanding on how the SERS enhancement depends on the chemical composition and structure of these nanoparticles has not been achieved. In this study, Mie theory and the discrete dipole approximation have been used to calculate the plasmonic spectra and near-field electromagnetic enhancements of Ag/Au hybrid NPs. In particular, we discuss how the electromagnetic enhancement depends on the mole fraction of Au in Ag/Au alloy NPs and how one may use extinction spectra to distinguish between Ag/Au alloyed NPs and Ag-Au core-shell NPs. We also show that for incident laser wavelengths between ∼410 nm and 520 nm, Ag/Au alloyed NPs provide better electromagnetic enhancement than pure Ag, pure Au, or Ag-Au core-shell structured NPs. Finally, we show that silica-core Ag/Au alloy shelled NPs provide even better performance than pure Ag/Au alloy or pure solid Ag and pure solid Au NPs. The theoretical results presented will be beneficial to the experimental efforts in optimizing the design of Ag/Au hybrid NPs for SERS-based detection methods.
Collapse
Affiliation(s)
- Elise Chaffin
- Department of Chemistry, The University of Memphis, Memphis, Tennessee 38152, USA
| | - Ryan T O'Connor
- Department of Chemistry, The University of Memphis, Memphis, Tennessee 38152, USA
| | - James Barr
- Department of Chemistry, The University of Memphis, Memphis, Tennessee 38152, USA
| | - Xiaohua Huang
- Department of Chemistry, The University of Memphis, Memphis, Tennessee 38152, USA
| | - Yongmei Wang
- Department of Chemistry, The University of Memphis, Memphis, Tennessee 38152, USA
| |
Collapse
|
50
|
Ieranò C, Portella L, Lusa S, Salzano G, D'Alterio C, Napolitano M, Buoncervello M, Macchia D, Spada M, Barbieri A, Luciano A, Barone MV, Gabriele L, Caraglia M, Arra C, De Rosa G, Scala S. CXCR4-antagonist Peptide R-liposomes for combined therapy against lung metastasis. NANOSCALE 2016; 8:7562-7571. [PMID: 26983756 DOI: 10.1039/c5nr06335c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The chemokine CXCL12 activates CXCR4, initiating multiple pathways that control immune cell trafficking, angiogenesis and embryogenesis; CXCR4 is also overexpressed in multiple tumors affecting metastatic dissemination. While there has been great enthusiasm for exploiting the CXCR4-CXCL12 axis as a target in cancer therapy, to date the promise has yet to be fulfilled. A new class of CXCR4-antagonist cyclic peptides was recently developed and the compound named Peptide R was identified as the most active. With the intent to improve the efficacy and biodistribution of Peptide R, stealth liposomes decorated with Peptide R were developed (PL-Peptide R). In vitro PL-Peptide R efficiently inhibited CXCR4-dependent migration and in vivo it significantly reduced lung metastases and increased overall survival in B16-CXCR4 injected C57BL/6 mice. To evaluate if PL-Peptide R could also be a drug delivery system for CXCR4 expressing tumors, the PL-Peptide R was loaded with doxorubicin (DOX) (PL-Peptide R-DOX). PL-Peptide R-DOX efficiently delivered DOX to CXCR4 expressing cell lines with a consequent decrease in the DOX IC50 efficient dose. In vivo, B16-CXCR4 injected C57BL/6 mice treated with PL-Peptide R-DOX developed fewer lung metastases compared to PL-DOX treated mice. This work provides the proof-of-concept to prevent metastasis by using combined nanomedicine.
Collapse
Affiliation(s)
- Caterina Ieranò
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Luigi Portella
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Sara Lusa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Giuseppina Salzano
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy. and Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Ave, Boston, MA, USA
| | - Crescenzo D'Alterio
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Maria Napolitano
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| | - Maria Buoncervello
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Daniele Macchia
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Massimo Spada
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Antonio Luciano
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Maria Vittoria Barone
- Department of Translational Medical Science and European Laboratory for the Investigation of Food Induced Disease (ELFID), University of Naples, Federico II, Via S. Pansini 5, 80131, Naples, Italy
| | - Lucia Gabriele
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio 7, 80138, Naples, Italy
| | - Claudio Arra
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy.
| | - Stefania Scala
- Molecular Immunology and Immune regulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale"-IRCCS, Italy.
| |
Collapse
|