1
|
Hakala S, Hämäläinen A, Sandelin S, Giannareas N, Närvä E. Detection of Cancer Stem Cells from Patient Samples. Cells 2025; 14:148. [PMID: 39851576 PMCID: PMC11764358 DOI: 10.3390/cells14020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
The existence of cancer stem cells (CSCs) in various tumors has become increasingly clear in addition to their prominent role in therapy resistance, metastasis, and recurrence. For early diagnosis, disease progression monitoring, and targeting, there is a high demand for clinical-grade methods for quantitative measurement of CSCs from patient samples. Despite years of active research, standard measurement of CSCs has not yet reached clinical settings, especially in the case of solid tumors. This is because detecting this plastic heterogeneous population of cells is not straightforward. This review summarizes various techniques, highlighting their benefits and limitations in detecting CSCs from patient samples. In addition, methods designed to detect CSCs based on secreted and niche-associated signaling factors are reviewed. Spatial and single-cell methods for analyzing patient tumor tissues and noninvasive techniques such as liquid biopsy and in vivo imaging are discussed. Additionally, methods recently established in laboratories, preclinical studies, and clinical assays are covered. Finally, we discuss the characteristics of an ideal method as we look toward the future.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Närvä
- Institute of Biomedicine and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, FI-20520 Turku, Finland; (S.H.); (A.H.); (S.S.); (N.G.)
| |
Collapse
|
2
|
Szewczyk K, Jiang L, Khawaja H, Miranti CK, Zohar Y. Microfluidic Applications in Prostate Cancer Research. MICROMACHINES 2024; 15:1195. [PMID: 39459070 PMCID: PMC11509716 DOI: 10.3390/mi15101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Prostate cancer is a disease in which cells in the prostate, a gland in the male reproductive system below the bladder, grow out of control and, among men, it is the second-most frequently diagnosed cancer (other than skin cancer). In recent years, prostate cancer death rate has stabilized and, currently, it is the second-most frequent cause of cancer death in men (after lung cancer). Most deaths occur due to metastasis, as cancer cells from the original tumor establish secondary tumors in distant organs. For a long time, classical cell cultures and animal models have been utilized in basic and applied scientific research, including clinical applications for many diseases, such as prostate cancer, since no better alternatives were available. Although helpful in dissecting cellular mechanisms, these models are poor predictors of physiological behavior mainly because of the lack of appropriate microenvironments. Microfluidics has emerged in the last two decades as a technology that could lead to a paradigm shift in life sciences and, in particular, controlling cancer. Microfluidic systems, such as organ-on-chips, have been assembled to mimic the critical functions of human organs. These microphysiological systems enable the long-term maintenance of cellular co-cultures in vitro to reconstitute in vivo tissue-level microenvironments, bridging the gap between traditional cell cultures and animal models. Several reviews on microfluidics for prostate cancer studies have been published focusing on technology advancement and disease progression. As metastatic castration-resistant prostate cancer remains a clinically challenging late-stage cancer, with no curative treatments, we expanded this review to cover recent microfluidic applications related to prostate cancer research. The review includes discussions of the roles of microfluidics in modeling the human prostate, prostate cancer initiation and development, as well as prostate cancer detection and therapy, highlighting potentially major contributions of microfluidics in the continuous march toward eradicating prostate cancer.
Collapse
Affiliation(s)
- Kailie Szewczyk
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
| | - Hunain Khawaja
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA;
| | - Cindy K. Miranti
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA;
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ 85721, USA; (K.S.); (L.J.)
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
3
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
4
|
Lamichhane A, Tavana H. Three-Dimensional Tumor Models to Study Cancer Stemness-Mediated Drug Resistance. Cell Mol Bioeng 2024; 17:107-119. [PMID: 38737455 PMCID: PMC11082110 DOI: 10.1007/s12195-024-00798-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/01/2024] [Indexed: 05/14/2024] Open
Abstract
Solid tumors often contain genetically different populations of cancer cells, stromal cells, various structural and soluble proteins, and other soluble signaling molecules. The American Cancer society estimated 1,958,310 new cancer cases and 609,820 cancer deaths in the United States in 2023. A major barrier against successful treatment of cancer patients is drug resistance. Gain of stem cell-like states by cancer cells under drug pressure or due to interactions with the tumor microenvironment is a major mechanism that renders therapies ineffective. Identifying approaches to target cancer stem cells is expected to improve treatment outcomes for patients. Most of our understanding of drug resistance and the role of cancer stemness is from monolayer cell cultures. Recent advances in cell culture technologies have enabled developing sophisticated three-dimensional tumor models that facilitate mechanistic studies of cancer drug resistance. This review summarizes the role of cancer stemness in drug resistance and highlights the various tumor models that are used to discover the underlying mechanisms and test potentially novel therapeutics.
Collapse
Affiliation(s)
- Astha Lamichhane
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325 USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325 USA
| |
Collapse
|
5
|
Shen F, Gao J, Zhang J, Ai M, Gao H, Liu Z. Vortex sorting of rare particles/cells in microcavities: A review. BIOMICROFLUIDICS 2024; 18:021504. [PMID: 38571909 PMCID: PMC10987199 DOI: 10.1063/5.0174938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 03/18/2024] [Indexed: 04/05/2024]
Abstract
Microfluidics or lab-on-a-chip technology has shown great potential for the separation of target particles/cells from heterogeneous solutions. Among current separation methods, vortex sorting of particles/cells in microcavities is a highly effective method for trapping and isolating rare target cells, such as circulating tumor cells, from flowing samples. By utilizing fluid forces and inertial particle effects, this passive method offers advantages such as label-free operation, high throughput, and high concentration. This paper reviews the fundamental research on the mechanisms of focusing, trapping, and holding of particles in this method, designs of novel microcavities, as well as its applications. We also summarize the challenges and prospects of this technique with the hope to promote its applications in medical and biological research.
Collapse
Affiliation(s)
- Feng Shen
- Authors to whom correspondence should be addressed: and
| | - Jie Gao
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, People’s Republic of China
| | - Jie Zhang
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, People’s Republic of China
| | - Mingzhu Ai
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing 100124, People’s Republic of China
| | - Hongkai Gao
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing 100853, People’s Republic of China
| | - Zhaomiao Liu
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
6
|
Zaky W, Ragoonanan D, Batth I, Dao L, Wang J, Xia X, Daw NC, Gill JB, Khatua S, Li S. Automated Capture and Analysis of Circulating Tumor Cells in Pediatric, Adolescent and Young Adult Patients with Central Nervous System Tumors. Cancers (Basel) 2023; 15:3853. [PMID: 37568669 PMCID: PMC10417345 DOI: 10.3390/cancers15153853] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Tumors of the central nervous system (CNS) are the most common and lethal childhood malignancy. Detection of residual disease and longitudinal monitoring of treatment response in patients are challenging and rely on serial imaging. This current standard of care fails to detect microscopic disease or provide molecular characteristics of residual tumors. As such, there is dire need for minimally invasive liquid biopsy techniques. We have previously shown the high specificity of using cell surface vimentin (CSV) to identify circulating tumor cells (CTCs) from patients bearing various types of cancers. Here, we describe the first report of CTCs captured from peripheral blood samples in 58 pediatric CNS tumor patients. In this study, we used a CSV-coated cell capture chip, the Abnova CytoQuest automated CTC isolation system, to boost the CTC capture from pediatric patients with CNS tumors. We successfully isolated CTCs in six glioma patients using immunostaining of histone H3 lysine27-to-methionine (H3K27M) mutations which are highly expressed by this tumor. We show that CSV is a viable marker for CNS CTC isolation and that this is a feasible method for detecting microscopic disease. Larger-scale studies focusing on CTCs in pediatric CNS tumors to explore their diagnostic and prognostic value are warranted.
Collapse
Affiliation(s)
- Wafik Zaky
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| | - Dristhi Ragoonanan
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| | - Izhar Batth
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| | - Long Dao
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| | - Jian Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xueqing Xia
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| | - Najat C. Daw
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| | - Jonathan B. Gill
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| | - Soumen Khatua
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77023, USA
| |
Collapse
|
7
|
Pei H, Han Z, Wang Y, Xu C, Li Y, Fan Y, Li L, Tang B. Retraction of "Label-Free Isolation of Low-Adhesion Cells with Stem Properties for Cancer Stem Cell-Specific Drug Evaluation". Anal Chem 2023; 95:6191. [PMID: 36122350 DOI: 10.1021/acs.analchem.2c00444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
8
|
Jiang B, Xie D, Wang S, Li X, Wu G. Advances in early detection methods for solid tumors. Front Genet 2023; 14:1091223. [PMID: 36911396 PMCID: PMC9998680 DOI: 10.3389/fgene.2023.1091223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
During the last decade, non-invasive methods such as liquid biopsy have slowly replaced traditional imaging and invasive pathological methods used to diagnose and monitor cancer. Improvements in the available detection methods have enabled the early screening and diagnosis of solid tumors. In addition, advances in early detection methods have made the continuous monitoring of tumor progression using repeat sampling possible. Previously, the focus of liquid biopsy techniques included the following: 1) the isolation of circulating tumor cells, circulating tumor DNA, and extracellular tumor vesicles from solid tumor cells in the patient's blood; in addition to 2) analyzing genomic and proteomic data contained within the isolates. Recently, there has been a rapid devolvement in the techniques used to isolate and analyze molecular markers. This rapid evolvement in detection techniques improves their accuracy, especially when few samples are available. In addition, there is a tremendous expansion in the acquisition of samples and targets for testing; solid tumors can be detected from blood and other body fluids. Test objects have also expanded from samples taken directly from cancer to include indirect objects affected in cancer development. Liquid biopsy technology has limitations. Even so, this detection technique is the key to a new phase of oncogenetics. This review aims to provide an overview of the current advances in liquid biopsy marker selection, isolation, and detection methods for solid tumors. The advantages and disadvantages of liquid biopsy technology will also be explored.
Collapse
Affiliation(s)
| | | | | | - Xiunan Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Microsecond cell triple-sorting enabled by multiple pulse irradiation of femtosecond laser. Sci Rep 2023; 13:405. [PMID: 36624119 PMCID: PMC9829734 DOI: 10.1038/s41598-022-27229-0] [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: 03/15/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
Femtosecond-laser-assisted cell manipulation, as one of the high throughput cell sorting techniques, is tailored for single-step multiple sorting based on controllable impulsive force. In this paper, femtosecond laser pulses are focused within a pocket structure and they induce an impulse force acting on the flowing objects. The impulsive force is shown to be controllable by a new method to adjust the femtosecond pulse properties. This allows precise streamline manipulation of objects having various physical qualities (e.g., weight and volume). The pulse energy, pulse number, and pulse interval of the femtosecond laser are altered to determine the impulsive force strength. The method is validated in single cell or bead triple-sorting experiments and its capability to perform streamline manipulation in as little as 10 μs is shown. The shift profiles of the beads acting under the impulsive force are studied in order to better understand the sorting mechanism. Additionally, beads and cells with different fluorescence intensities are successfully detected and directed into different microchannels, with maximum success rates of 90% and 64.5%, respectively. To sum up, all results suggest that this method has the potential to sort arbitrary subpopulations by altering the number of femtosecond pulses and that it takes the first step toward developing a single-step multi-selective system.
Collapse
|
10
|
Chen K, George TJ, Fan ZH. Lateral Filter Array Microfluidic Devices for Detecting Circulating Tumor Cells. Methods Mol Biol 2023; 2679:1-13. [PMID: 37300605 DOI: 10.1007/978-1-0716-3271-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Circulating tumor cells (CTCs) are an important liquid biopsy biomarker for next-generation cancer diagnosis and prognosis. However, their clinical usage is hindered by the rarity of CTCs in patient's peripheral blood. Microfluidics has shown unique advantages in CTC isolation and detection. We have developed lateral filter array microfluidic (LFAM) devices for highly efficient CTC isolation. In this chapter, we describe in detail the design and fabrication of the LFAM devices and their applications for CTC enumeration from clinical blood samples.
Collapse
Affiliation(s)
- Kangfu Chen
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Thomas J George
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Chemistry, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
11
|
Comparative application of microfluidic systems in circulating tumor cells and extracellular vesicles isolation; a review. Biomed Microdevices 2022; 25:4. [PMID: 36574057 DOI: 10.1007/s10544-022-00644-w] [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: 11/14/2022] [Indexed: 12/28/2022]
Abstract
Cancer is a prevalent cause of mortality globally, where early diagnosis leads to a reduced death rate. Many researchers' common strategies are based on personalized diagnostic methods with rapid response and high accuracy. This technology was developed by applying liquid biopsy instead of tissue biopsies in the case of tumor cell analysis that facilitates point-of-care testing for cancer diagnosis and treatment. In recent years, significant progress in microfluidic technology led to the successful isolation, analysis, and monitoring of cancer biomarkers in body liquid biopsy with merits like high sensitivity and flexibility, low sample usage, cost effective, and the ability of automation. The most critical and informative markers in body liquid refer to circulating tumor cells (CTCs) and extracellular vesicles derived from tumors (EVs) that carry various biomarkers in their structure (DNAs, proteins, and RNAs) as compared to ctDNA. The released ctDNA has a low half-life and decreased sensitivity due to large amounts of nucleic acid in serum. This review intends to highlight different cancer screening tests with a particular focus on the details regarding the only FDA-approved and awaiting technologies for FDA clearance to isolate CTCs and EVs based on microfluidics systems.
Collapse
|
12
|
Słomka A, Wang B, Mocan T, Horhat A, Willms AG, Schmidt-Wolf IGH, Strassburg CP, Gonzalez-Carmona MA, Lukacs-Kornek V, Kornek MT. Extracellular Vesicles and Circulating Tumour Cells - complementary liquid biopsies or standalone concepts? Theranostics 2022; 12:5836-5855. [PMID: 35966579 PMCID: PMC9373826 DOI: 10.7150/thno.73400] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/06/2022] [Indexed: 12/11/2022] Open
Abstract
Liquid biopsies do promise a lot, but are they keeping it? In the past decade, additional novel biomarkers qualified to be called like that, of which, some took necessary hurdles resulting in FDA approval and clinical use. Some others are since a while around, well known and were once regarded to be a game changer in cancer diagnosis or cancer screening. But, during their clinical use limitations were observed from statistical significance and questions raised regarding their robustness, that eventually led to be dropped from associated clinical guidelines for certain applications including cancer diagnosis. The purpose of this review isn't to give a broad overview of all current liquid biopsy as biomarkers, weight them and promise a brighter future in cancer prevention, but rather to take a deeper look on two of those who do qualify to be called liquid biopsies now or then. These two are probably of greatest interest conceptually and methodically, and likely have the highest chances to be in clinical use soon, with a portfolio extension over their original conceptual usage. We aim to dig deeper beyond cancer diagnosis or cancer screening. Actually, we aim to review in depth extracellular vesicles (EVs) and compare with circulating tumour cells (CTCs). The latter methodology is partially FDA approved and in clinical use. We will lay out similarities as taking advantage of surface antigens on EVs and CTCs in case of characterization and quantification. But drawing readers' attention to downstream application based on capture/isolation methodology and simply on their overall nature, here apparently being living material eventually recoverable as CTCs are vs. dead material with transient effects on recipient cell as in case of EVs. All this we try to bring in perspective, compare and conclude towards which future direction we are aiming for, or should aim for. Do we announce a winner between CTCs vs EVs? No, but we provide good reasons to intensify research on them.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85-067 Bydgoszcz, Poland
| | - Bingduo Wang
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany.,Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Tudor Mocan
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Adelina Horhat
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Arnulf G Willms
- Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany.,Department of General, Visceral and Vascular Surgery, German Armed Forces Hospital Hamburg, 22049 Hamburg, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Christian P Strassburg
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Maria A Gonzalez-Carmona
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Veronika Lukacs-Kornek
- Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Miroslaw T Kornek
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| |
Collapse
|
13
|
Varillas JI, Chen K, Dopico P, Zhang J, George TJ, Fan ZH. Comparison of sample preparation methods for rare cell isolation in microfluidic devices. CAN J CHEM 2022; 100:512-519. [PMID: 36338875 PMCID: PMC9635407 DOI: 10.1139/cjc-2021-0229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2024]
Abstract
The analysis of circulating tumor cells (CTCs) is important for cancer diagnosis and prognosis. Microfluidics has been employed for CTC analysis due to its scaling advantages and high performance. However, pre-analytical methods for CTC sample preparation are often combined with microfluidic platforms because a large sample volume is required to detect extremely rare CTCs. Among pre-analytical methods, Ficoll-Paque™, OncoQuick™, and RosetteSep™ are commonly used to separate cells of interest. To compare their performance, we spiked L3.6pl pancreatic cancer cells into healthy blood samples and then employed each technique to prepare blood samples, followed by using a microfluidic platform to capture and detect L3.6pl cells. We found these three methods have similar performance, though the slight edge of RosetteSep™ over Ficoll-Paque™ is statistically significant. We also studied the effects of the tumor cell concentrations on the performance of the frequently used Ficoll-Paque™ method. Furthermore, we examined the repeatability and variability of each pre-analytical technique and the microfluidics-enabled detection. This study will provide researchers and clinicians with comparative data that can influence the choice of sample preparation method, help estimate CTC loss in each pre-analytical method, and correlate the results of clinical studies that employ different techniques.
Collapse
Affiliation(s)
- Jose I Varillas
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL 32611, USA
| | - Kangfu Chen
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA
| | - Pablo Dopico
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA
| | - Jinling Zhang
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA
| | - Thomas J George
- Department of Medicine, University of Florida, P.O. Box 100278, Gainesville, FL 32610, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, P.O. Box 116250, Gainesville, FL 32611, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL 32611, USA; Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL 32611, USA
| |
Collapse
|
14
|
Bhat MP, Thendral V, Uthappa UT, Lee KH, Kigga M, Altalhi T, Kurkuri MD, Kant K. Recent Advances in Microfluidic Platform for Physical and Immunological Detection and Capture of Circulating Tumor Cells. BIOSENSORS 2022; 12:220. [PMID: 35448280 PMCID: PMC9025399 DOI: 10.3390/bios12040220] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 05/05/2023]
Abstract
CTCs (circulating tumor cells) are well-known for their use in clinical trials for tumor diagnosis. Capturing and isolating these CTCs from whole blood samples has enormous benefits in cancer diagnosis and treatment. In general, various approaches are being used to separate malignant cells, including immunomagnets, macroscale filters, centrifuges, dielectrophoresis, and immunological approaches. These procedures, on the other hand, are time-consuming and necessitate multiple high-level operational protocols. In addition, considering their low efficiency and throughput, the processes of capturing and isolating CTCs face tremendous challenges. Meanwhile, recent advances in microfluidic devices promise unprecedented advantages for capturing and isolating CTCs with greater efficiency, sensitivity, selectivity and accuracy. In this regard, this review article focuses primarily on the various fabrication methodologies involved in microfluidic devices and techniques specifically used to capture and isolate CTCs using various physical and biological methods as well as their conceptual ideas, advantages and disadvantages.
Collapse
Affiliation(s)
- Mahesh Padmalaya Bhat
- Centre for Research in Functional Materials (CRFM), Jain Global Campus, Jain University, Bengaluru 562112, Karnataka, India; (M.P.B.); (V.T.); (M.K.)
- Agricultural Automation Research Center, Chonnam National University, Gwangju 61186, Korea;
| | - Venkatachalam Thendral
- Centre for Research in Functional Materials (CRFM), Jain Global Campus, Jain University, Bengaluru 562112, Karnataka, India; (M.P.B.); (V.T.); (M.K.)
| | | | - Kyeong-Hwan Lee
- Agricultural Automation Research Center, Chonnam National University, Gwangju 61186, Korea;
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Korea
| | - Madhuprasad Kigga
- Centre for Research in Functional Materials (CRFM), Jain Global Campus, Jain University, Bengaluru 562112, Karnataka, India; (M.P.B.); (V.T.); (M.K.)
| | - Tariq Altalhi
- Department of Chemistry, Faculty of Science, Taif University, Taif 21944, Saudi Arabia;
| | - Mahaveer D. Kurkuri
- Centre for Research in Functional Materials (CRFM), Jain Global Campus, Jain University, Bengaluru 562112, Karnataka, India; (M.P.B.); (V.T.); (M.K.)
| | - Krishna Kant
- Departamento de Química Física, Campus Universitario, CINBIO Universidade de Vigo, 36310 Vigo, Spain
| |
Collapse
|
15
|
Dopico PJ, Le MCN, Burgess B, Yang Z, Zhao Y, Wang Y, George TJ, Fan ZH. Longitudinal Study of Circulating Biomarkers in Patients with Resectable Pancreatic Ductal Adenocarcinoma. BIOSENSORS 2022; 12:206. [PMID: 35448266 PMCID: PMC9028387 DOI: 10.3390/bios12040206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 05/12/2023]
Abstract
While patients with resectable pancreatic ductal adenocarcinoma (PDAC) show improved survival compared to their non-resectable counterparts, survival remains low owing to occult metastatic disease and treatment resistance. Liquid biopsy based on circulating tumor cells (CTCs) and cell-free DNA (cfDNA) has been shown to predict recurrence and treatment resistance in various types of cancers, but their utility has not been fully demonstrated in resectable PDAC. We have simultaneously tracked three circulating biomarkers, including CTCs, cfDNA, and circulating tumor DNA (ctDNA), over a period of cancer treatment using a microfluidic device and droplet digital PCR (ddPCR). The microfluidic device is based on the combination of filtration and immunoaffinity mechanisms. We have measured CTCs, cfDNA, and ctDNA in a cohort of seven resectable PDAC patients undergoing neoadjuvant therapy followed by surgery, and each patient was followed up to 10 time points over a period of 4 months. CTCs were detectable in all patients (100%) at some point during treatment but were detectable in only three out of six patients (50%) prior to the start of treatment. Median cfDNA concentrations remained comparable to negative controls throughout treatment. ddPCR was able to find KRAS mutations in six of seven patients (86%); however, these mutations were present in only two of seven patients (29%) prior to treatment. Overall, the majority of circulating biomarkers (81% for CTCs and 91% for cfDNA/ctDNA) were detected after the start of neoadjuvant therapy but before surgery. This study suggests that a longitudinal study of circulating biomarkers throughout treatment provides more useful information than those single time-point tests for resectable PDAC patients.
Collapse
Affiliation(s)
- Pablo J. Dopico
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (P.J.D.); (M.-C.N.L.)
| | - Minh-Chau N. Le
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (P.J.D.); (M.-C.N.L.)
| | - Benjamin Burgess
- UF Health Cancer Center, University of Florida, 2033 Mowry Rd., Gainesville, FL 32610, USA;
| | - Zhijie Yang
- Atila Biosystems, 740 Sierra Vista Ave., Unit E, Mountain View, CA 94043, USA; (Z.Y.); (Y.Z.); (Y.W.)
| | - Yu Zhao
- Atila Biosystems, 740 Sierra Vista Ave., Unit E, Mountain View, CA 94043, USA; (Z.Y.); (Y.Z.); (Y.W.)
| | - Youxiang Wang
- Atila Biosystems, 740 Sierra Vista Ave., Unit E, Mountain View, CA 94043, USA; (Z.Y.); (Y.Z.); (Y.W.)
| | - Thomas J. George
- UF Health Cancer Center, University of Florida, 2033 Mowry Rd., Gainesville, FL 32610, USA;
- Department of Medicine, University of Florida, 1600 SW Archer Rd., Gainesville, FL 32610, USA
| | - Z. Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (P.J.D.); (M.-C.N.L.)
- UF Health Cancer Center, University of Florida, 2033 Mowry Rd., Gainesville, FL 32610, USA;
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
16
|
Park Y, Yoon HJ, Lee SE, Lee LP. Multifunctional Cellular Targeting, Molecular Delivery, and Imaging by Integrated Mesoporous-Silica with Optical Nanocrescent Antenna: MONA. ACS NANO 2022; 16:2013-2023. [PMID: 35041396 DOI: 10.1021/acsnano.1c07015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Multifunctional nanoprobes have attracted significant attention in a wide range of disciplines such as nanomedicine, precision medicine, and cancer diagnosis and treatment. However, integrating multifunctional ability in a nanoscale structure to precisely target, image, and deliver with cellular spatial/temporal resolution is still challenging in cellulo applications. This is because the development of such high-precision resolution needs to be carried out without labeling, photobleaching, and structurally segregating live cells. In this study, we present an integrated nanostructure of a mesoporous-silica nanosphere with an optical nanocrescent antenna (MONA) for multifunctional cellular targeting, drug delivery, and molecular imaging with spatiotemporal resolution. MONA comprises a systematically constructed Au nanocrescent (AuNC) antenna as a nanosensor and optical switch on a mesoporous-silica nanosphere as a cargo to molecular delivery. MONA made of antiepithelial cell adhesion molecules (anti-EpCAM)-conjugated AuNC facilitates the specific targeting of breast cancer cells, resulting in a highly focused photothermal gradient that functions as a molecular emitter. This light-driven molecular, doxorubicin (DOX) delivery function allows rapid apoptosis of breast cancer cells. Since MONA permits the tracking of quantum biological electron-transfer processes, in addition to its role as an on-demand optical switch, it enables the monitoring of the dynamic behavior of cellular cytochrome c pivoting cell apoptosis in response to the DOX delivery. Owing to the integrated functions of molecular actuation and direct sensing at the precisely targeted spot afforded by MONA, we anticipate that this multifunctional optical nanoantenna structure will have an impact in the fields of nanomedicine, cancer theranostics, and basic life sciences.
Collapse
Affiliation(s)
- Younggeun Park
- Department of Bioengineering and Biomolecular Nanotechnology Center, Berkeley Sensor and Actuator Center and University of California, Berkeley, California 94720, United States
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hyeun Joong Yoon
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Somin Eunice Lee
- Department of Electrical & Computer Engineering, Department of Biomedical Engineering, Applied Physics, Biointerfaces Institute, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Luke P Lee
- Department of Bioengineering and Biomolecular Nanotechnology Center, Berkeley Sensor and Actuator Center and University of California, Berkeley, California 94720, United States
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, California 94720, United States
- Harvard Institute of Medicine, Harvard Medical School, Department of Medicine, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts 02115 United States
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
17
|
Exploring the Clinical Utility of Pancreatic Cancer Circulating Tumor Cells. Int J Mol Sci 2022; 23:ijms23031671. [PMID: 35163592 PMCID: PMC8836025 DOI: 10.3390/ijms23031671] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 01/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most frequent pancreatic cancer type, characterized by a dismal prognosis due to late diagnosis, frequent metastases, and limited therapeutic response to standard chemotherapy. Circulating tumor cells (CTCs) are a rare subset of tumor cells found in the blood of cancer patients. CTCs has the potential utility for screening, early and definitive diagnosis, prognostic and predictive assessment, and offers the potential for personalized management. However, a gold-standard CTC detection and enrichment method remains elusive, hindering comprehensive comparisons between studies. In this review, we summarize data regarding the utility of CTCs at different stages of PDAC from early to metastatic disease and discuss the molecular profiling and culture of CTCs. The characterization of CTCs brings us closer to defining the specific CTC subpopulation responsible for metastasis with the potential to uncover new therapies and more effective management options for PDAC.
Collapse
|
18
|
Lin D, Shen L, Luo M, Zhang K, Li J, Yang Q, Zhu F, Zhou D, Zheng S, Chen Y, Zhou J. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther 2021; 6:404. [PMID: 34803167 PMCID: PMC8606574 DOI: 10.1038/s41392-021-00817-8] [Citation(s) in RCA: 444] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cells (CTCs) are tumor cells that have sloughed off the primary tumor and extravasate into and circulate in the blood. Understanding of the metastatic cascade of CTCs has tremendous potential for the identification of targets against cancer metastasis. Detecting these very rare CTCs among the massive blood cells is challenging. However, emerging technologies for CTCs detection have profoundly contributed to deepening investigation into the biology of CTCs and have facilitated their clinical application. Current technologies for the detection of CTCs are summarized herein, together with their advantages and disadvantages. The detection of CTCs is usually dependent on molecular markers, with the epithelial cell adhesion molecule being the most widely used, although molecular markers vary between different types of cancer. Properties associated with epithelial-to-mesenchymal transition and stemness have been identified in CTCs, indicating their increased metastatic capacity. Only a small proportion of CTCs can survive and eventually initiate metastases, suggesting that an interaction and modulation between CTCs and the hostile blood microenvironment is essential for CTC metastasis. Single-cell sequencing of CTCs has been extensively investigated, and has enabled researchers to reveal the genome and transcriptome of CTCs. Herein, we also review the clinical applications of CTCs, especially for monitoring response to cancer treatment and in evaluating prognosis. Hence, CTCs have and will continue to contribute to providing significant insights into metastatic processes and will open new avenues for useful clinical applications.
Collapse
Affiliation(s)
- Danfeng Lin
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lesang Shen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Luo
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinfan Li
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Yang
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fangfang Zhu
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhou
- Department of Surgery, Traditional Chinese Medical Hospital of Zhuji, Shaoxing, China
| | - Shu Zheng
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiding Chen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiaojiao Zhou
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
19
|
Li MM, Yuan J, Guan XY, Ma NF, Liu M. Molecular subclassification of gastrointestinal cancers based on cancer stem cell traits. Exp Hematol Oncol 2021; 10:53. [PMID: 34774101 PMCID: PMC8590337 DOI: 10.1186/s40164-021-00246-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/05/2021] [Indexed: 12/18/2022] Open
Abstract
Human gastrointestinal malignancies are highly heterogeneous cancers. Clinically, heterogeneity largely contributes to tumor progression and resistance to therapy. Heterogeneity within gastrointestinal cancers is defined by molecular subtypes in genomic and transcriptomic analyses. Cancer stem cells (CSCs) have been demonstrated to be a major source of tumor heterogeneity; therefore, assessing tumor heterogeneity by CSC trait-guided classification of gastrointestinal cancers is essential for the development of effective therapies. CSCs share critical features with embryonic stem cells (ESCs). Molecular investigations have revealed that embryonic genes and developmental signaling pathways regulating the properties of ESCs or cell lineage differentiation are abnormally active and might be oncofetal drivers in certain tumor subtypes. Currently, multiple strategies allow comprehensive identification of tumor subtype-specific oncofetal signatures and evaluation of subtype-specific therapies. In this review, we summarize current knowledge concerning the molecular classification of gastrointestinal malignancies based on CSC features and elucidate their clinical relevance. We also outline strategies for molecular subtype identification and subtype-based therapies. Finally, we explore how clinical implementation of tumor classification by CSC subtype might facilitate the development of more effective personalized therapies for gastrointestinal cancers.
Collapse
Affiliation(s)
- Mei-Mei Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou, 511436, China
| | - Jun Yuan
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou, 511436, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- Department of Clinical Oncology, State Key Laboratory of Liver Research, University of Hong Kong, Hong Kong, China
| | - Ning-Fang Ma
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou, 511436, China
| | - Ming Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou, 511436, China.
| |
Collapse
|
20
|
Negative enrichment of circulating tumor cells from unmanipulated whole blood with a 3D printed device. Sci Rep 2021; 11:20583. [PMID: 34663896 PMCID: PMC8523721 DOI: 10.1038/s41598-021-99951-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/23/2021] [Indexed: 01/07/2023] Open
Abstract
Reliable and routine isolation of circulating tumor cells (CTCs) from peripheral blood would allow effective monitoring of the disease and guide the development of personalized treatments. Negative enrichment of CTCs by depleting normal blood cells ensures against a biased selection of a subpopulation and allows the assay to be applied on different tumor types. Here, we report an additively manufactured microfluidic device that can negatively enrich viable CTCs from clinically-relevant volumes of unmanipulated whole blood samples. Our device depletes nucleated blood cells based on their surface antigens and the smaller anucleated cells based on their size. Enriched CTCs are made available off the device in suspension making our technique compatible with standard immunocytochemical, molecular and functional assays. Our device could achieve a ~ 2.34-log depletion by capturing > 99.5% of white blood cells from 10 mL of whole blood while recovering > 90% of spiked tumor cells. Furthermore, we demonstrated the capability of the device to isolate CTCs from blood samples collected from patients (n = 15) with prostate and pancreatic cancers in a pilot study. A universal CTC assay that can differentiate tumor cells from normal blood cells with the specificity of clinically established membrane antigens yet require no label has the potential to enable routine blood-based tumor biopsies at the point-of-care.
Collapse
|
21
|
Perales S, Torres C, Jimenez-Luna C, Prados J, Martinez-Galan J, Sanchez-Manas JM, Caba O. Liquid biopsy approach to pancreatic cancer. World J Gastrointest Oncol 2021; 13:1263-1287. [PMID: 34721766 PMCID: PMC8529923 DOI: 10.4251/wjgo.v13.i10.1263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/18/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) continues to pose a major clinical challenge. There has been little improvement in patient survival over the past few decades, and it is projected to become the second leading cause of cancer mortality by 2030. The dismal 5-year survival rate of less than 10% after the diagnosis is attributable to the lack of early symptoms, the absence of specific biomarkers for an early diagnosis, and the inadequacy of available chemotherapies. Most patients are diagnosed when the disease has already metastasized and cannot be treated. Cancer interception is vital, actively intervening in the malignization process before the development of a full-blown advanced tumor. An early diagnosis of PC has a dramatic impact on the survival of patients, and improved techniques are urgently needed to detect and evaluate this disease at an early stage. It is difficult to obtain tissue biopsies from the pancreas due to its anatomical position; however, liquid biopsies are readily available and can provide useful information for the diagnosis, prognosis, stratification, and follow-up of patients with PC and for the design of individually tailored treatments. The aim of this review was to provide an update of the latest advances in knowledge on the application of carbohydrates, proteins, cell-free nucleic acids, circulating tumor cells, metabolome compounds, exosomes, and platelets in blood as potential biomarkers for PC, focusing on their clinical relevance and potential for improving patient outcomes.
Collapse
Affiliation(s)
- Sonia Perales
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Granada 18071, Spain
| | - Carolina Torres
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Sciences, University of Granada, Granada 18071, Spain
| | - Cristina Jimenez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain
| | - Joaquina Martinez-Galan
- Department of Medical Oncology, Hospital Universitario Virgen de las Nieves, Granada 18011, Spain
| | | | - Octavio Caba
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada 18100, Spain
| |
Collapse
|
22
|
Jia Y, Shen P, Yan T, Zhou W, Sun J, Han X. Microfluidic Tandem Mechanical Sorting System for Enhanced Cancer Stem Cell Isolation and Ingredient Screening. Adv Healthc Mater 2021; 10:e2100985. [PMID: 34486235 DOI: 10.1002/adhm.202100985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/13/2021] [Indexed: 12/13/2022]
Abstract
Robust isolation of cancer stem cells (CSCs) in a high-throughput, label-free manner is critical for understanding tumor heterogeneity and developing therapeutic strategies targeting CSCs. Cell-mechanics-based microfluidic sorting systems provide efficient and specific platforms for investigation of stem cell-like characteristics on the basis of cell deformability and cell-substrate adhesion properties. In the present study, a microfluidic tandem mechanical sorting system is developed to enrich CSCs with high flexibility and low adhesive capacity. In the integrated microfluidic system, cancer cells are driven by hydrodynamic forces to flow continuously through two featured devices, which are functionalized with sequentially variable microbarriers and surface-coated fluid mixing microchannels, respectively. Collected deformable and low-adhesive cancer cells exhibit enhanced stem cell-like properties with higher stemness and metastasis capacity both in vitro and in vivo, compared with each single device separation. Using these devices, bioactive natural compound screening targeting CSCs is performed and a potent therapeutic compound isoliquiritigenin from licorice is identified to inhibit the lung cancer stem cell phenotype. Taken together, this microfluidic tandem mechanical sorting system can facilitate drug screening targeting CSCs and the analysis of signals regulating CSC function in drug resistance.
Collapse
Affiliation(s)
- Yuanyuan Jia
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Peiliang Shen
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Tao Yan
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Weijia Zhou
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Jia Sun
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Xin Han
- Department of Biochemistry and Molecular Biology School of Medicine & Holistic Integrative Medicine Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization Nanjing University of Chinese Medicine Nanjing 210023 China
| |
Collapse
|
23
|
Fu G, Cheng KS, Chen A, Xu Z, Chen X, Tian J, Xu C, Sun Y, Neoh KH, Dai Y, Han RPS, Jin B. Microfluidic Assaying of Circulating Tumor Cells and Its Application in Risk Stratification of Urothelial Bladder Cancer. Front Oncol 2021; 11:701298. [PMID: 34178700 PMCID: PMC8222714 DOI: 10.3389/fonc.2021.701298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Bladder cancer is characterized by its frequent recurrence and progression. Effective treatment strategies need to be based on an accurate risk stratification, in which muscle invasiveness and tumor grade represent the two most important factors. Traditional imaging techniques provide preliminary information about muscle invasiveness but are lacking in terms of accuracy. Although as the gold standard, pathological biopsy is only available after the surgery and cannot be performed longitudinally for long-term surveillance. In this work, we developed a microfluidic approach that interrogates circulating tumor cells (CTCs) in the peripheral blood of bladder cancer patients to reflect the risk stratification of the disease. In a cohort of 48 bladder cancer patients comprising 33 non-muscle invasive bladder cancer (NMIBC) cases and 15 muscle invasive bladder cancer (MIBC) cases, the CTC count was found to be considerably higher in the MIBC group compared with the NMIBC group (4.67 vs. 1.88 CTCs/3 mL, P=0.019), and was significantly higher in high-grade bladder cancer patients verses low-grade bladder cancer patients (3.69 vs. 1.18 CTCs/3mL, P=0.024). This microfluidic assay of CTCs is believed to be a promising complementary tool for the risk stratification of bladder cancer.
Collapse
Affiliation(s)
- Guanghou Fu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kok Suen Cheng
- Jiangzhong Cancer Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Department of Material Science and Engineering, College of Engineering, Peking University, Beijing, China
| | - Anqi Chen
- Department of Material Science and Engineering, College of Engineering, Peking University, Beijing, China
| | - Zhijie Xu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyi Chen
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junjie Tian
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Congcong Xu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yukun Sun
- Department of Material Science and Engineering, College of Engineering, Peking University, Beijing, China
| | - Kuang Hong Neoh
- Department of Material Science and Engineering, College of Engineering, Peking University, Beijing, China
| | - Yun Dai
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ray P. S. Han
- Jiangzhong Cancer Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Baiye Jin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
24
|
Lei Y, Wang X, Sun H, Fu Y, Tian Y, Yang L, Wang J, Xia F. Association of Preoperative NANOG-Positive Circulating Tumor Cell Levels With Recurrence of Hepatocellular Carcinoma. Front Oncol 2021; 11:601668. [PMID: 34123777 PMCID: PMC8190394 DOI: 10.3389/fonc.2021.601668] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 02/22/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) and Circulating tumor cells (CTCs) have been proposed as fundamental causes for the recurrence of hepatocellular carcinoma (HCC). CTCs isolated from patients with HCC illustrate a unique Nanog expression profile analysis. The aim of this study was to enhance the prediction of recurrence and prognosis of the CTC phenotype in patients with HCC by combining Nanog expression into a combined forecasting model. SUBJECTS MATERIALS AND METHODS We collected 320 blood samples from 160 patients with HCC cancer before surgery and used CanPatrol™ CTC enrichment technology and in situ hybridization (ISH) to enrich and detect CTCs and CSCs. Nanog expression in all CTCs was also determined. In addition, RT-PCR and immunohistochemistry were used to study the expression of Nanog, E-Cadherin, and N-Cadherin in liver cancer tissues and to conduct clinical correlation studies. RESULTS The numbers of EpCAM mRNA+ CTCs and Nanog mRNA+ CTCs were strongly correlated with postoperative HCC recurrence (CTC number (P = 0.03), the total number of mixed CTCS (P = 0.02), and Nanog> 6.7 (P = 0.001), with Nanog > 6.7 (P = 0.0003, HR = 2.33) being the most crucial marker. There are significant differences in the expression of Nanog on different types of CTC: most Epithelial CTCs do not express Nanog, while most of Mixed CTC and Mesenchymal CTC express Nanog, and their positive rates are 38.7%, 66.7%, and 88.7%, respectively, (P=0.0001). Moreover, both CTC (≤/> 13.3) and Nanog (≤/>6.7) expression were significantly correlated with BCLC stage, vascular invasion, tumor size, and Hbv-DNA (all P < 0.05). In the young group and the old group, patients with higher Nanog expression had a higher recurrence rate. (P < 0.001). CONCLUSIONS The number of Nanog-positive cells showed positive correlation with the poor prognosis of HCC patients. The detection and analysis of CTC markers (EpCAM and CK8, 18, CD45 Vimentin,Twist and 19) and CSCs markers (NANOG) are of great value in the evaluation of tumor progression.
Collapse
Affiliation(s)
- Yongrong Lei
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Xishu Wang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, the First Hospital Affiliated to AMU (Southwest Hospital), Chongqing, China
| | - Heng Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Yuna Fu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Yichen Tian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ludi Yang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, the First Hospital Affiliated to AMU (Southwest Hospital), Chongqing, China
| | - Jianhua Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Feng Xia
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, the First Hospital Affiliated to AMU (Southwest Hospital), Chongqing, China
| |
Collapse
|
25
|
Chemi F, Mohan S, Guevara T, Clipson A, Rothwell DG, Dive C. Early Dissemination of Circulating Tumor Cells: Biological and Clinical Insights. Front Oncol 2021; 11:672195. [PMID: 34026650 PMCID: PMC8138033 DOI: 10.3389/fonc.2021.672195] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
Circulating tumor cells (CTCs) play a causal role in the development of metastasis, the major cause of cancer-associated mortality worldwide. In the past decade, the development of powerful cellular and molecular technologies has led to a better understanding of the molecular characteristics and timing of dissemination of CTCs during cancer progression. For instance, genotypic and phenotypic characterization of CTCs, at the single cell level, has shown that CTCs are heterogenous, disseminate early and could represent only a minor subpopulation of the primary tumor responsible for disease relapse. While the impact of molecular profiling of CTCs has not yet been translated to the clinic, CTC enumeration has been widely used as a prognostic biomarker to monitor treatment response and to predict disease relapse. However, previous studies have revealed a major challenge: the low abundance of CTCs in the bloodstream of patients with cancer, especially in early stage disease where the identification and characterization of subsequently "lethal" cells has potentially the greatest clinical relevance. The CTC field is rapidly evolving with development of new technologies to improve the sensitivity of CTC detection, enumeration, isolation, and molecular profiling. Here we examine the technical and analytical validity of CTC technologies, we summarize current data on the biology of CTCs that disseminate early and review CTC-based clinical applications.
Collapse
Affiliation(s)
- Francesca Chemi
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Macclesfield, United Kingdom
| | | | | | | | | | - Caroline Dive
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Macclesfield, United Kingdom
| |
Collapse
|
26
|
The Present and Future Role of Microfluidics for Protein and Peptide-Based Therapeutics and Diagnostics. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11094109] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The implementation of peptide-based molecules within the medical field has vast potential, owing to their unique nature and predictable physicochemical profiles. However, peptide therapeutic usage is hindered by delivery-related challenges, meaning that their formulations must be altered to overcome these limitations. This process could be propelled by applying microfluidics (MFs) due to its highly controllable and adaptable attributes; however, therapeutic research within this field is extremely limited. Peptides possess multifunctional roles within therapeutic formulations, ranging from enhancing target specificity to acting as the active component of the medicine. Diagnostically, MFs are well explored in the field of peptides, as MFs provide an unsullied platform to provide fast yet accurate examinations. The capacity to add attributes, such as integrated sensors and microwells, to the MF chip, only enhances the attractiveness of MFs as a diagnostic platform. The structural individuality of peptides makes them prime candidates for diagnostic purposes, for example, antigen detection and isolation. Therefore, this review provides a useful insight into the current applications of MFs for peptide-based therapy and diagnostics and highlights potential gaps in the field that are yet to be explored or optimized.
Collapse
|
27
|
Li M, Zhang W, Yang X, An G, Zhao W. The α2δ1 subunit of the voltage-gated calcium channel acts as a potential candidate for breast cancer tumor initial cells biomarker. Cancer Biomark 2021; 31:295-305. [PMID: 33896833 DOI: 10.3233/cbm-203165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The voltage-gated calcium channel subunit alpha 2 delta 1 (α2δ1) is a functional tumor initial cells (TICs) marker for some solid cancer cells. This study aimed to investigate whether α2δ1 can be used as a potential TIC marker for breast cancer cells. METHODS α2δ1+ and α2δ1- cells were identified and sorted from the breast cancer cell lines MDA-MB-231, MDA-MB-435s and ZR-75-1 by Immunofluorescence (IF) and Fluorescent-activated cell sorting (FACS) analyses. Spheroid formation in vitro and tumorigenesis in NOD/SCID mice were assessed to determine the self-renewal and serial transplantation abilities of these cells. Using a lentivirus infection system for α2δ1 in breast cancer cell lines, we determined the mRNA levels of stemnessassociated genes by quality real-time PCR (qRT-PCR). Boyden chamber and wounding assays were further performed to detect the migration of α2δ1 overexpression cells. Bioinformatics explored the relationship of molecular classification of breast cancer and drug resistance. RESULTS α2δ1 presents on the cytomembrane of breast cancer cells, with a positive rate of 1.5-3%. The α2δ1+ cells in breast cancer cell lines have a stronger self-renewal ability and tumor initiating properties in vitro and in vivo. Overexpressing α2δ1 successfully enhanced the sphere-forming efficiency, and upregulated the expression of stemness-associated genes, and increased cell migration. However, seldom significant was available between estrogen receptor +/- (ER+/-), progesterone receptor (PR+/-), and Her2+/-. CONCLUSIONS Breast cancer cells positive for the α2δ1 charactered tumor initiation, and α2δ1 is a potential TIC marker for breast cancer that further promotes the migration.
Collapse
|
28
|
Cho HY, Choi JH, Lim J, Lee SN, Choi JW. Microfluidic Chip-Based Cancer Diagnosis and Prediction of Relapse by Detecting Circulating Tumor Cells and Circulating Cancer Stem Cells. Cancers (Basel) 2021; 13:1385. [PMID: 33803846 PMCID: PMC8003176 DOI: 10.3390/cancers13061385] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022] Open
Abstract
Detecting circulating tumor cells (CTCs) has been considered one of the best biomarkers in liquid biopsy for early diagnosis and prognosis monitoring in cancer. A major challenge of using CTCs is detecting extremely low-concentrated targets in the presence of high noise factors such as serum and hematopoietic cells. This review provides a selective overview of the recent progress in the design of microfluidic devices with optical sensing tools and their application in the detection and analysis of CTCs and their small malignant subset, circulating cancer stem cells (CCSCs). Moreover, discussion of novel strategies to analyze the differentiation of circulating cancer stem cells will contribute to an understanding of metastatic cancer, which can help clinicians to make a better assessment. We believe that the topic discussed in this review can provide brief guideline for the development of microfluidic-based optical biosensors in cancer prognosis monitoring and clinical applications.
Collapse
Affiliation(s)
- Hyeon-Yeol Cho
- Department of Bio & Fermentation Convergence Technology, Kookmin University, Seoul 02707, Korea;
- Interdisciplinary Program for Bio-health Convergence, Kookmin University, Seoul 02707, Korea
| | - Jin-Ha Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
- School of Chemical Engineering, Jeonbuk National University, Jeonju 54896, Korea
| | - Joungpyo Lim
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
| | - Sang-Nam Lee
- Uniance Gene Inc., 1107 Teilhard Hall, 35 Baekbeom-Ro, Mapo-Gu, Seoul 04107, Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea; (J.-H.C.); (J.L.)
| |
Collapse
|
29
|
Kang Y, Niu Z, Hadlock T, Purcell E, Lo T, Zeinali M, Owen S, Keshamouni VG, Reddy R, Ramnath N, Nagrath S. On-Chip Biogenesis of Circulating NK Cell-Derived Exosomes in Non-Small Cell Lung Cancer Exhibits Antitumoral Activity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003747. [PMID: 33747745 PMCID: PMC7967048 DOI: 10.1002/advs.202003747] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/14/2020] [Indexed: 05/19/2023]
Abstract
As the recognition between natural killer (NK) cells and cancer cells does not require antigen presentation, NK cells are being actively studied for use in adoptive cell therapies in the rapidly evolving armamentarium of cancer immunotherapy. In addition to utilizing NK cells, recent studies have shown that exosomes derived from NK cells also exhibit antitumor properties. Furthermore, these NK cell-derived exosomes exhibit higher stability, greater modification potentials and less immunogenicity compared to NK cells. Therefore, technologies that allow highly sensitive and specific isolation of NK cells and NK cell-derived exosomes can enable personalized NK-mediated cancer therapeutics in the future. Here, a novel microfluidic system to collect patient-specific NK cells and on-chip biogenesis of NK-exosomes is proposed. In a small cohort of non-small cell lung cancer (NSCLC) patients, both NK cells and circulating tumor cells (CTCs) were isolated, and it is found NSCLC patients have high numbers of NK and NK-exosomes compared with healthy donors, and these concentrations show a trend of positive and negative correlations with bloodborne CTC numbers, respectively. It is further demonstrated that the NK-exosomes harvested from NK-graphene oxide chip exhibit cytotoxic effect on CTCs. This versatile system is expected to be used for patient-specific NK-based immunotherapies along with CTCs for potential prognostic/diagnostic applications.
Collapse
Affiliation(s)
- Yoon‐Tae Kang
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Zeqi Niu
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Thomas Hadlock
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Emma Purcell
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Ting‐Wen Lo
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Mina Zeinali
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Sarah Owen
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | | | - Rishindra Reddy
- Michigan Medicine Thoracic Surgery ClinicTaubman Center1500E Medical Center Dr. SPC 5344Ann ArborMI48109USA
| | - Nithya Ramnath
- Department of Internal MedicineUniversity of MichiganAnn ArborMI48109USA
| | - Sunitha Nagrath
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
- Rogel Cancer CenterUniversity of Michigan1500 East Medical Center DriveAnn ArborMI48109USA
| |
Collapse
|
30
|
Pang TCY, Po JW, Becker TM, Goldstein D, Pirola RC, Wilson JS, Apte MV. Circulating tumour cells in pancreatic cancer: A systematic review and meta-analysis of clinicopathological implications. Pancreatology 2021; 21:103-114. [PMID: 33309014 DOI: 10.1016/j.pan.2020.11.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The detection and quantification of circulating tumour cells (CTCs) in pancreatic cancer (PC) has the potential to provide prognostic information. The aim of this review was to provide an overview of the literature surrounding CTCs in PC. METHODS A systematic literature review on CTCs in PC between 2005-2020 was performed. Data based on peripheral vein samples were used to determine the positivity rate of CTCs, their prognostic significance and their relative numbers compared to portal vein (PV) samples. RESULTS The overall CTC detection rate in forty-four articles was 65% (95%CI: 55-75%). Detection rate for CellSearch was 26% (95%CI: 14-38%), which was lower than for both filtration and microfluidic techniques. In nine studies with >50 patients, overall survival was worse with CTC positivity (HR 1.82; 95%CI: 1.61-2.05). Five of seven studies which described PV CTC collection provided patient-level data. PV CTC yield was 7.7-fold (95%CI 1.35-43.9) that of peripheral blood. CONCLUSIONS CTCs were detected in the peripheral circulation of most patients with PC and may be related to prognosis and disease stage. PV blood contains more CTCs than peripheral blood sampling. This review points to the maturation of techniques of CTC enrichment, and its evidence base for eventual clinical deployment.
Collapse
Affiliation(s)
- Tony C Y Pang
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia; Surgical Innovations Unit, Westmead Hospital, Westmead, Australia; Westmead Clinical School, University of Sydney, Westmead, Australia
| | - Joseph W Po
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, South Western Clinical School, University of New South Wales, School of Medicine, Western Sydney University, Australia; Surgical Innovations Unit, Westmead Hospital, Westmead, Australia; Westmead Clinical School, University of Sydney, Westmead, Australia
| | - Therese M Becker
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, South Western Clinical School, University of New South Wales, School of Medicine, Western Sydney University, Australia
| | - David Goldstein
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Romano C Pirola
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Jeremy S Wilson
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia.
| |
Collapse
|
31
|
Ajani JA, Xu Y, Huo L, Wang R, Li Y, Wang Y, Pizzi MP, Scott AW, Harada K, Ma L, Yao X, Jin J, Zhao W, Dong X, Badgwell BD, Shanbhag ND, Tatlonghari G, Estrella JS, Roy Chowdhuri S, Kobayashi M, Vykouka JV, Hanash S, Calin GA, Peng G, Lee JS, Johnson RL, Wang Z, Wang L, Song S. YAP1 mediates gastric adenocarcinoma peritoneal metastases that are attenuated by YAP1 inhibition. Gut 2021; 70:55-66. [PMID: 32345613 PMCID: PMC9832914 DOI: 10.1136/gutjnl-2019-319748] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Peritoneal carcinomatosis (PC; malignant ascites or implants) occurs in approximately 45% of advanced gastric adenocarcinoma (GAC) patients and associated with a poor survival. The molecular events leading to PC are unknown. The yes-associated protein 1 (YAP1) oncogene has emerged in many tumour types, but its clinical significance in PC is unclear. Here, we investigated the role of YAP1 in PC and its potential as a therapeutic target. METHODS Patient-derived PC cells, patient-derived xenograft (PDX) and patient-derived orthotopic (PDO) models were used to study the function of YAP1 in vitro and in vivo. Immunofluorescence and immunohistochemical staining, RNA sequencing (RNA-Seq) and single-cell RNA-Seq (sc-RNA-Seq) were used to elucidate the expression of YAP1 and PC cell heterogeneity. LentiCRISPR/Cas9 knockout of YAP1 and a YAP1 inhibitor were used to dissect its role in PC metastases. RESULTS YAP1 was highly upregulated in PC tumour cells, conferred cancer stem cell (CSC) properties and appeared to be a metastatic driver. Dual staining of YAP1/EpCAM and sc-RNA-Seq revealed that PC tumour cells were highly heterogeneous, YAP1high PC cells had CSC-like properties and easily formed PDX/PDO tumours but also formed PC in mice, while genetic knockout YAP1 significantly slowed tumour growth and eliminated PC in PDO model. Additionally, pharmacologic inhibition of YAP1 specifically reduced CSC-like properties and suppressed tumour growth in YAP1high PC cells especially in combination with cytotoxics in vivo PDX model. CONCLUSIONS YAP1 is essential for PC that is attenuated by YAP1 inhibition. Our data provide a strong rationale to target YAP1 in clinic for GAC patients with PC.
Collapse
Affiliation(s)
- Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,To whom correspondence should be addressed: Shumei Song, PhD, tel.: 713-834-6144, ,; Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA. Jaffer A. Ajani, MD, Tel: 713-792-2828, ; Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Yan Xu
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P.R. China
| | - Longfei Huo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ruiping Wang
- Detartment of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuan Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P.R. China
| | - Ying Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Melissa Pool Pizzi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ailing W. Scott
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kazuto Harada
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lang Ma
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaodan Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaochuan Dong
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Brian D. Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Namita D. Shanbhag
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ghia Tatlonghari
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeannelyn S. Estrella
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sinchita Roy Chowdhuri
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Makoto Kobayashi
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jody V. Vykouka
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George A. Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guang Peng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Randy L. Johnson
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, P.R. China
| | - Linghua Wang
- Detartment of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
32
|
Kim J, Cho H, Kim J, Park JS, Han KH. A disposable smart microfluidic platform integrated with on-chip flow sensors. Biosens Bioelectron 2020; 176:112897. [PMID: 33342692 DOI: 10.1016/j.bios.2020.112897] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 01/27/2023]
Abstract
Microfluidic devices are powerful tools for biological, biomedical, chemical, and pharmaceutical applications, but their commercialization is still hindered by the lack of methods to automatically control fluid flow in a low-cost, simple, accurate, and safe manner. This study introduces a disposable smart microfluidic platform (DIS-μChip), which can be fully automated and utilized for a wide range of applications. On-chip microfluidic flow sensors are integrated with the platform and placed at all inlet and outlet channels, thereby allowing the DIS-μChip to be fully automated with a pressure control system. Furthermore, these confer a self-diagnosis function through monitoring of all the input and output flow rates. The DIS-μChip consists of a disposable polymeric microchannel superstrate and a permanent multifunctional substrate, which could be assembled and disassembled using only vacuum pressure. The superstrate was fabricated by combining a polydimethylsiloxane microchannel structure with a polyethylene terephthalate (PET) thin film. The substrate contains sense electrodes for the on-chip-integrated flow sensors and functional components for creating an energy field, which can penetrate the PET thin film and manipulate the fluid in the microchannels of the superstrate. Owing to the film-chip technique, the superstrate was disposable and could prevent biological cross-contamination, which cannot be realized with conventional flow sensors. The usefulness of the DIS-μChip was demonstrated by using it to isolate circulating tumor cells from the blood of patients with pancreatic cancer and to obtain cancer-specific genetic information from them with droplet digital PCR.
Collapse
Affiliation(s)
- Jinho Kim
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae-si, 50834, South Korea
| | - Hyungseok Cho
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae-si, 50834, South Korea
| | - Junhyeong Kim
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae-si, 50834, South Korea
| | - Joon Seong Park
- Pancreatobiliary Cancer Clinic, Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06229, South Korea
| | - Ki-Ho Han
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae-si, 50834, South Korea.
| |
Collapse
|
33
|
Yee NS, Zhang S, He HZ, Zheng SY. Extracellular Vesicles as Potential Biomarkers for Early Detection and Diagnosis of Pancreatic Cancer. Biomedicines 2020; 8:biomedicines8120581. [PMID: 33297544 PMCID: PMC7762339 DOI: 10.3390/biomedicines8120581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic carcinoma (PC) is highly metastatic, and it tends to be detected at advanced stages. Identifying and developing biomarkers for early detection of PC is crucial for a potentially curative treatment. Extracellular vesicles (EVs) are bilayer lipid membrane-structured nanovesicles found in various human bodily fluids, and they play important roles in tumor biogenesis and metastasis. Cancer-derived EVs are enriched with DNA, RNA, protein, and lipid, and they have emerged as attractive diagnostic biomarkers for early detection of PC. In this article, we provided an overview of the cell biology of EVs and their isolation and analysis, and their roles in cancer pathogenesis and progression. Multiplatform analyses of plasma-based exosomes for genomic DNA, micro RNA, mRNA, circular RNA, and protein for diagnosis of PC were critically reviewed. Numerous lines of evidence demonstrate that liquid biopsy with analysis of EV-based biomarkers has variable performance for diagnosis of PC. Future investigation is indicated to optimize the methodology for isolating and analyzing EVs and to identify the combination of EV-based biomarkers and other clinical datasets, with the goal of improving the predictive value, sensitivity, and specificity of screening tests for early detection and diagnosis of PC.
Collapse
Affiliation(s)
- Nelson S. Yee
- Division of Hematology-Oncology, Department of Medicine, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
- Next-Generation Therapies Program, Penn State Cancer Institute, Hershey, PA 17033, USA
- Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence: (N.S.Y.); (H.-Z.H.); (S.-Y.Z.); Tel.: +1-717-531-8678 (N.S.Y.); +1-949-878-2679 (H.-Z.H.); +1-412-268-3684 (S.-Y.Z.)
| | - Sheng Zhang
- Micro & Nano Integrated Biosystem Laboratory, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Hong-Zhang He
- Micro & Nano Integrated Biosystem Laboratory, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
- Captis Diagnostics, Inc., Pittsburgh, PA 15213, USA
- Correspondence: (N.S.Y.); (H.-Z.H.); (S.-Y.Z.); Tel.: +1-717-531-8678 (N.S.Y.); +1-949-878-2679 (H.-Z.H.); +1-412-268-3684 (S.-Y.Z.)
| | - Si-Yang Zheng
- Micro & Nano Integrated Biosystem Laboratory, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
- Correspondence: (N.S.Y.); (H.-Z.H.); (S.-Y.Z.); Tel.: +1-717-531-8678 (N.S.Y.); +1-949-878-2679 (H.-Z.H.); +1-412-268-3684 (S.-Y.Z.)
| |
Collapse
|
34
|
Ma Y, Yin Y, Ni L, Miao H, Wang Y, Pan C, Tian X, Pan J, You T, Li B, Pan G. Thermo-responsive imprinted hydrogel with switchable sialic acid recognition for selective cancer cell isolation from blood. Bioact Mater 2020; 6:1308-1317. [PMID: 33251380 PMCID: PMC7662873 DOI: 10.1016/j.bioactmat.2020.10.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 01/07/2023] Open
Abstract
In this work, a sialic acid (SA)-imprinted thermo-responsive hydrogel layer was prepared for selective capture and release of cancer cells. The SA-imprinting process was performed at 37 °C using thermo-responsive functional monomer, thus generating switchable SA-recognition sites with potent SA binding at 37 °C and weak binding at a lower temperature (e.g., 25 °C). Since SA is often overexpressed at the glycan terminals of cell membrane proteins or lipids, the SA-imprinted hydrogel layer could be used for selective cancer cell recognition. Our results confirmed that the hydrogel layer could efficiently capture cancer cells from not only the culture medium but also the real blood samples. In addition, the captured cells could be non-invasively released by lowing the temperature. Considering the non-invasive processing mode, considerable capture efficiency, good cell selectivity, as well as the more stable and durable SA-imprinted sites compared to natural antibodies or receptors, this thermo-responsive hydrogel layer could be used as a promising and general platform for cell-based cancer diagnosis. Thermo-responsive sialic acid (SA)-imprinted hydrogel layer was prepared. The hydrogel layer could efficiently and selective capture cancer cells at 37 °C. The captured cancer cells could be released at a lower temperature (e.g., 25 °C). The hydrogel layer could be used for capture and release cancer cells from blood.
Collapse
Affiliation(s)
- Yue Ma
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China.,School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China.,Jiangsu Agrochem Laboratory, Changzhou, Jiangsu 213022, PR China
| | - Yimei Yin
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Li Ni
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Haohan Miao
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Yingjia Wang
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Cheng Pan
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Xiaohua Tian
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Jianming Pan
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Tianyan You
- Key Laboratory of Modern Agricultural Equipment and Technology, Ministry of Education, School of Agricultural Equipment Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Bin Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| |
Collapse
|
35
|
Frappart PO, Hofmann TG. Pancreatic Ductal Adenocarcinoma (PDAC) Organoids: The Shining Light at the End of the Tunnel for Drug Response Prediction and Personalized Medicine. Cancers (Basel) 2020; 12:E2750. [PMID: 32987786 PMCID: PMC7598647 DOI: 10.3390/cancers12102750] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents 90% of pancreatic malignancies. In contrast to many other tumor entities, the prognosis of PDAC has not significantly improved during the past thirty years. Patients are often diagnosed too late, leading to an overall five-year survival rate below 10%. More dramatically, PDAC cases are on the rise and it is expected to become the second leading cause of death by cancer in western countries by 2030. Currently, the use of gemcitabine/nab-paclitaxel or FOLFIRINOX remains the standard chemotherapy treatment but still with limited efficiency. There is an urgent need for the development of early diagnostic and therapeutic tools. To this point, in the past 5 years, organoid technology has emerged as a revolution in the field of PDAC personalized medicine. Here, we are reviewing and discussing the current technical and scientific knowledge on PDAC organoids, their future perspectives, and how they can represent a game change in the fight against PDAC by improving both diagnosis and treatment options.
Collapse
Affiliation(s)
- Pierre-Olivier Frappart
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | | |
Collapse
|
36
|
Chen K, Amontree J, Varillas J, Zhang J, George TJ, Fan ZH. Incorporation of lateral microfiltration with immunoaffinity for enhancing the capture efficiency of rare cells. Sci Rep 2020; 10:14210. [PMID: 32848184 PMCID: PMC7450051 DOI: 10.1038/s41598-020-71041-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/22/2020] [Indexed: 02/03/2023] Open
Abstract
The methods for isolating rare cells such as circulating tumor cells (CTCs) can be generally classified into two categories: those based on physical properties (e.g., size) and methods based on biological properties (e.g., immunoaffinity). CellSearch, the only FDA-approved method for the CTC-based cancer prognosis, relies on immunoaffinity interactions between CTCs and antibodies immobilized on magnetic particles. Immunoaffinity-based CTC isolation has also been employed in microfluidic devices, which show higher capture efficiency than CellSearch. We report here our investigation of combining size-based microfiltration into a microfluidic device with immunoaffinity for enhanced capture efficiency of CTCs. The device consists of four serpentine main channels, and each channel contains an array of lateral filters that create a two-dimensional flow. The main flow is through the serpentine channel, allowing the majority of the sample to pass by while the secondary flow goes through the lateral filters. The device design is optimized to make all fluid particles interact with filters. The filter sizes range from 24 to 12 µm, being slightly larger than or having similar dimension of CTCs. These filters are immobilized with antibodies specific to CTCs and thus they function as gates, allowing normal blood cells to pass by while forcing the interactions between CTCs and antibodies on the filter surfaces. The hydrodynamic force experienced by a CTC was also studied for optimal experimental conditions to ensure immunoaffinity-enabled cell capture. The device was evaluated by capturing two types of tumor cells spiked in healthy blood or a buffer, and we found that their capture efficiency was between 87.2 and 93.5%. The platform was further validated by isolating CTCs from blood samples of patients with metastatic pancreatic cancer.
Collapse
Affiliation(s)
- Kangfu Chen
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Jacob Amontree
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Jose Varillas
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Jinling Zhang
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA
| | - Thomas J George
- Department of Medicine, University of Florida, P.O. Box 100278, Gainesville, FL, 32610, USA
| | - Z Hugh Fan
- Interdisciplinary Microsystems Group (IMG), Department of Mechanical and Aerospace Engineering, University of Florida, P.O. BOX 116250, Gainesville, FL, 32611, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, P.O. Box 116131, Gainesville, FL, 32611, USA.
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA.
| |
Collapse
|
37
|
Jariyal H, Gupta C, Bhat VS, Wagh JR, Srivastava A. Advancements in Cancer Stem Cell Isolation and Characterization. Stem Cell Rev Rep 2020; 15:755-773. [PMID: 31863337 DOI: 10.1007/s12015-019-09912-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Occurrence of stem cells (CSCs) in cancer is well established in last two decades. These rare cells share several properties including presence of common surface markers, stem cell markers, chemo- and radio- resistance and are highly metastatic in nature; thus, considered as valuable prognostic and therapeutic targets in cancer. However, the studies related to CSCs pave number of issues due to rare cell population and difficulties in their isolation ascribed to common stem cell marker. Various techniques including flow cytometry, laser micro-dissection, fluorescent nanodiamonds and microfluidics are used for the isolation of these rare cells. In this review, we have included the advance strategies adopted for the isolation of CSCs using above mentioned techniques. Furthermore, CSCs are primarily found in the core of the solid tumors and their microenvironment plays an important role in maintenance, self-renewal, division and tumor development. Therefore, in vivo tracking and model development become obligatory for functional studies of CSCs. Fluorescence and bioluminescence tagging has been widely used for transplantation assay and lineage tracking experiments to improve our understanding towards CSCs behaviour in their niche. Techniques such as Magnetic resonance imaging (MRI) and Positron emission tomography (PET) have proved useful for tracking of endogenous CSCs which could be helpful in their identification in clinical settings.
Collapse
Affiliation(s)
- Heena Jariyal
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Chanchal Gupta
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Vedika Sandeep Bhat
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Jayant Ramakant Wagh
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Akshay Srivastava
- Department of Medical Device, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India.
| |
Collapse
|
38
|
Kip Ç, Akbay E, Gökçal B, Savaş BO, Onur MA, Tuncel A. Colorimetric determination of tumor cells via peroxidase-like activity of a cell internalizable nanozyme: Hyaluronic acid attached-silica microspheres containing accessible magnetite nanoparticles. Colloids Surf A Physicochem Eng Asp 2020. [DOI: 10.1016/j.colsurfa.2020.124812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
39
|
Genna A, Vanwynsberghe AM, Villard AV, Pottier C, Ancel J, Polette M, Gilles C. EMT-Associated Heterogeneity in Circulating Tumor Cells: Sticky Friends on the Road to Metastasis. Cancers (Basel) 2020; 12:E1632. [PMID: 32575608 PMCID: PMC7352430 DOI: 10.3390/cancers12061632] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transitions (EMTs) generate hybrid phenotypes with an enhanced ability to adapt to diverse microenvironments encountered during the metastatic spread. Accordingly, EMTs play a crucial role in the biology of circulating tumor cells (CTCs) and contribute to their heterogeneity. Here, we review major EMT-driven properties that may help hybrid Epithelial/Mesenchymal CTCs to survive in the bloodstream and accomplish early phases of metastatic colonization. We then discuss how interrogating EMT in CTCs as a companion biomarker could help refine cancer patient management, further supporting the relevance of CTCs in personalized medicine.
Collapse
Affiliation(s)
- Anthony Genna
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Aline M. Vanwynsberghe
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Amélie V. Villard
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| | - Charles Pottier
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
- Department of Medical Oncology, University Hospital of Liège, 4000 Liège, Belgium
| | - Julien Ancel
- CHU (Centre Hopitalier Universitaire) de Reims, Hôpital Maison Blanche, Service de Pneumologie, 51092 Reims, France;
- INSERM, UMR (Unité Mixte de Recherche)-S1250, SFR CAP-SANTE, Université de Reims Champagne-Ardenne, 51097 Reims, France;
| | - Myriam Polette
- INSERM, UMR (Unité Mixte de Recherche)-S1250, SFR CAP-SANTE, Université de Reims Champagne-Ardenne, 51097 Reims, France;
- CHU de Reims, Hôpital Maison Blanche, Laboratoire de Pathologie, 51092 Reims, France
| | - Christine Gilles
- GIGA-Cancer, Laboratory of Tumor and Development Biology, CHU Sart-Tilman, University of Liège, Pathology Tower, 4000 Liège, Belgium; (A.G.); (A.M.V.); (A.V.V.); (C.P.)
| |
Collapse
|
40
|
Zhu L, Hissa B, Győrffy B, Jann JC, Yang C, Reissfelder C, Schölch S. Characterization of Stem-like Circulating Tumor Cells in Pancreatic Cancer. Diagnostics (Basel) 2020; 10:E305. [PMID: 32429174 PMCID: PMC7278018 DOI: 10.3390/diagnostics10050305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/26/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth most frequent cause of death from cancer. Circulating tumor cells (CTCs) with stem-like characteristics lead to distant metastases and thus contribute to the dismal prognosis of PDAC. Our purpose is to investigate the role of stemness in CTCs derived from a genetically engineered mouse model of PDAC and to further explore the potential molecular mechanisms. The publically available RNA sequencing dataset GSE51372 was analyzed, and CTCs with (CTC-S) or without (CTC-N) stem-like features were discriminated based on a principal component analysis (PCA). Differentially expressed genes, weighted gene co-expression network analysis (WGCNA), and further functional enrichment analyses were performed. The prognostic role of the candidate gene (CTNNB1) was assessed in a clinical PDAC patient cohort. Overexpression of the pluripotency marker Klf4 (Krüppel-like factor 4) in CTC-S cells positively correlates with Ctnnb1 (β-Catenin) expression, and their interaction presumably happens via protein-protein binding in the nucleus. As a result, the adherens junction pathway is significantly enriched in CTC-S. Furthermore, the overexpression of Ctnnb1 is a negative prognostic factor for progression-free survival (PFS) and relapse-free survival (RFS) in human PDAC cohort. Overexpression of Ctnnb1 may thus promote the metastatic capabilities of CTCs with stem-like properties via adherens junctions in murine PDAC.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (L.Z.); (B.H.); (C.Y.); (C.R.)
| | - Barbara Hissa
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (L.Z.); (B.H.); (C.Y.); (C.R.)
| | - Balázs Győrffy
- 2nd Department of Pediatrics, Semmelweis University, H-1094 Budapest, Hungary;
- TTK Cancer Biomarker Research Group, Institute of Enzymology, H-1117 Budapest, Hungary
| | - Johann-Christoph Jann
- Department of Medicine III, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Cui Yang
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (L.Z.); (B.H.); (C.Y.); (C.R.)
| | - Christoph Reissfelder
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (L.Z.); (B.H.); (C.Y.); (C.R.)
- German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sebastian Schölch
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (L.Z.); (B.H.); (C.Y.); (C.R.)
- German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
41
|
Lim M, Park J, Lowe AC, Jeong HO, Lee S, Park HC, Lee K, Kim GH, Kim MH, Cho YK. A lab-on-a-disc platform enables serial monitoring of individual CTCs associated with tumor progression during EGFR-targeted therapy for patients with NSCLC. Am J Cancer Res 2020; 10:5181-5194. [PMID: 32373206 PMCID: PMC7196290 DOI: 10.7150/thno.44693] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/14/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: Unlike traditional biopsy, liquid biopsy, which is a largely non-invasive diagnostic and monitoring tool, can be performed more frequently to better track tumors and mutations over time and to validate the efficiency of a cancer treatment. Circulating tumor cells (CTCs) are considered promising liquid biopsy biomarkers; however, their use in clinical settings is limited by high costs and a low throughput of standard platforms for CTC enumeration and analysis. In this study, we used a label-free, high-throughput method for CTC isolation directly from whole blood of patients using a standalone, clinical setting-friendly platform. Methods: A CTC-based liquid biopsy approach was used to examine the efficacy of therapy and emergent drug resistance via longitudinal monitoring of CTC counts, DNA mutations, and single-cell-level gene expression in a prospective cohort of 40 patients with epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer. Results: The change ratio of the CTC counts was associated with tumor response, detected by CT scan, while the baseline CTC counts did not show association with progression-free survival or overall survival. We achieved a 100% concordance rate for the detection of EGFR mutation, including emergence of T790M, between tumor tissue and CTCs. More importantly, our data revealed the importance of the analysis of the epithelial/mesenchymal signature of individual pretreatment CTCs to predict drug responsiveness in patients. Conclusion: The fluid-assisted separation technology disc platform enables serial monitoring of CTC counts, DNA mutations, as well as unbiased molecular characterization of individual CTCs associated with tumor progression during targeted therapy.
Collapse
|
42
|
Dianat-Moghadam H, Azizi M, Eslami-S Z, Cortés-Hernández LE, Heidarifard M, Nouri M, Alix-Panabières C. The Role of Circulating Tumor Cells in the Metastatic Cascade: Biology, Technical Challenges, and Clinical Relevance. Cancers (Basel) 2020; 12:E867. [PMID: 32260071 PMCID: PMC7225923 DOI: 10.3390/cancers12040867] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Metastases and cancer recurrence are the main causes of cancer death. Circulating Tumor Cells (CTCs) and disseminated tumor cells are the drivers of cancer cell dissemination. The assessment of CTCs' clinical role in early metastasis prediction, diagnosis, and treatment requires more information about their biology, their roles in cancer dormancy, and immune evasion as well as in therapy resistance. Indeed, CTC functional and biochemical phenotypes have been only partially characterized using murine metastasis models and liquid biopsy in human patients. CTC detection, characterization, and enumeration represent a promising tool for tailoring the management of each patient with cancer. The comprehensive understanding of CTCs will provide more opportunities to determine their clinical utility. This review provides much-needed insights into this dynamic field of translational cancer research.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran; (H.D.-M.); (M.N.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Mehdi Azizi
- Proteomics Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran;
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, UPRES, EA2415, 34093 Montpellier, France (L.E.C.-H.)
| | - Luis Enrique Cortés-Hernández
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, UPRES, EA2415, 34093 Montpellier, France (L.E.C.-H.)
| | - Maryam Heidarifard
- Drug Applied Research Center, Tabriz University of Medical Sciences, 51368 Tabriz, Iran;
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran; (H.D.-M.); (M.N.)
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, UPRES, EA2415, 34093 Montpellier, France (L.E.C.-H.)
| |
Collapse
|
43
|
Abstract
Electrokinetic separation techniques in microfluidics are a powerful analytical chemistry tool, although an inherent limitation of microfluidics is their low sample throughput. In this article we report a free-flow variant of an electrokinetic focusing method, namely ion concentration polarization focusing (ICPF). The analytes flow continuously through the system via pressure driven flow while they separate and concentrate perpendicularly to the flow by ICPF. We demonstrate free flow ion concentration polarization focusing (FF-ICPF) in two operating modes, namely peak and plateau modes. Additionally, we showed the separation resolution could be improved by the use of an electrophoretic spacer. We report a concentration factor of 10 in human blood plasma in continuous flow at a flow rate of 15 μL min-1.
Collapse
Affiliation(s)
- Vasileios A Papadimitriou
- BIOS Lab on a Chip group, MESA+ Institute for Nanotechnology, Max Planck Centre for Complex Fluid Dynamics and Technical Medical Centre, University of Twente, Enschede 7500 AE, The Netherlands
| | - Loes I Segerink
- BIOS Lab on a Chip group, MESA+ Institute for Nanotechnology, Max Planck Centre for Complex Fluid Dynamics and Technical Medical Centre, University of Twente, Enschede 7500 AE, The Netherlands
| | - Jan C T Eijkel
- BIOS Lab on a Chip group, MESA+ Institute for Nanotechnology, Max Planck Centre for Complex Fluid Dynamics and Technical Medical Centre, University of Twente, Enschede 7500 AE, The Netherlands
| |
Collapse
|
44
|
Wu J, Hu S, Zhang L, Xin J, Sun C, Wang L, Ding K, Wang B. Tumor circulome in the liquid biopsies for cancer diagnosis and prognosis. Theranostics 2020; 10:4544-4556. [PMID: 32292514 PMCID: PMC7150480 DOI: 10.7150/thno.40532] [Citation(s) in RCA: 85] [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: 09/22/2019] [Accepted: 01/07/2020] [Indexed: 12/20/2022] Open
Abstract
Liquid biopsy is a convenient, fast, non-invasive and reproducible sampling method that can dynamically reflect the changes in tumor gene expression profile, and provide a robust basis for individualized therapy and early diagnosis of cancer. Circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs) are the currently approved diagnostic biomarkers for screening cancer patients. In addition, tumor-derived extracellular vesicles (tdEVs), circulating tumor-derived proteins, circulating tumor RNA (ctRNA) and tumor-bearing platelets (TEPs) are other components of liquid biopsies with diagnostic potential. In this review, we have discussed the clinical applications of these biomarkers, and the factors that limit their implementation in routine clinical practice. In addition, the most recent developments in the isolation and analysis of circulating tumor biomarkers have been summarized, and the potential of non-blood liquid biopsies in tumor diagnostics has also been discussed.
Collapse
Affiliation(s)
- Jicheng Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Shen Hu
- Department of Obstetrics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Lihong Zhang
- Department of Biochemistry, College of Biomedical Sciences, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jinxia Xin
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Chongran Sun
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Liquan Wang
- Department of Obstetrics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Kefeng Ding
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Ben Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
45
|
Qiu J, Xu J, Zhang K, Gu W, Nie L, Wang G, Luo Y. Refining Cancer Management Using Integrated Liquid Biopsy. Am J Cancer Res 2020; 10:2374-2384. [PMID: 32089746 PMCID: PMC7019147 DOI: 10.7150/thno.40677] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 10/13/2019] [Indexed: 12/17/2022] Open
Abstract
Liquid biopsy has emerged in the last ten years as an appealing noninvasive strategy to support early cancer diagnosis and follow-up interventions. However, conventional liquid biopsy strategies involving specified biomarkers have encountered unexpected inconsistencies stemming from the use of different analytical methodologies. Recent reports have repeatedly demonstrated that integrated detection of multiple liquid biopsy biomarkers can significantly improve diagnostic performance by eliminating the influence of intratumoral heterogeneity. Herein, we review the progress in the field of liquid biopsy and propose a novel integrated liquid biopsy framework consisting of three categories: elementary, intermediate, and advanced integration. We also summarize the merits of the integration strategy and propose a roadmap toward refining cancer diagnosis, metastasis surveillance, and prognostication.
Collapse
|
46
|
Affiliation(s)
- Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66044, United States
- Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66044, United States
- Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Ian M. Freed
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66044, United States
- Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66044, United States
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66044, United States
- Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66044, United States
- Department of Mechanical Engineering, The University of Kansas, Lawrence, Kansas 66044, United States
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66044, United States
| |
Collapse
|
47
|
Liu D, Hong Y, Li Y, Hu C, Yip TC, Yu WK, Zhu Y, Fong CC, Wang W, Au SK, Wang S, Yang M. Targeted destruction of cancer stem cells using multifunctional magnetic nanoparticles that enable combined hyperthermia and chemotherapy. Theranostics 2020; 10:1181-1196. [PMID: 31938059 PMCID: PMC6956796 DOI: 10.7150/thno.38989] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 10/14/2019] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cells (CSCs) have been implicated in cancer recurrence and therapy resistance. Therefore, a CSC-targeted therapy that disrupts the maintenance and survival of CSCs may offer an effective approach in killing tumor cells in primary tumors and preventing the metastasis caused by CSCs. Nanoparticles (NPs)-based thermotherapy and/or chemotherapy are promising therapeutic methods for cancer treatment. Methods: A silica-based multifunctional NP system was present, which encapsulated a chemotherapeutic agent and magnetic cores and coated with a specific antibody against the lung CSCs. The efficacy of this novel therapeutic strategy was systematically studied both in vitro and in vivo by simultaneous activating the combined thermotherapy and chemotherapy via CSC-targeted NPs. Results: These NPs were systematically administered and activated for targeted chemotherapy and thermotherapy by using an externally applied alternating magnetic field (AMF). The antibody-modified NPs targeted to lung CSCs with enhanced cellular uptake in vitro and extended accumulation in tumor in vivo. Up to 98% of lung CSCs was killed in vitro with 30-min application of AMF, due to the combined effects of hyperthermia and chemotherapeutic drug treatment. In in vivo models, this combined therapy significantly suppressed tumor growth and metastasis in lung CSC xenograft-bearing mice, with minimal side effects and adverse effects. Conclusion: With good biocompatibility and targeting capability, the nanodrug delivery system may offer a promising clinical platform for the combined thermotherapy and chemotherapy. This work demonstrated the feasibility of developing multifunctional nanomedicine targeting CSCs for effective cancer treatment.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
- College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, China
| | - Yingcai Hong
- Department of Thoracic Surgery, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen 510000, China
| | - Yaping Li
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Chong Hu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Tak-Chun Yip
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Wai-Kin Yu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| | - Yu Zhu
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Chi-Chun Fong
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Department of Thoracic Surgery, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen 510000, China
| | - Weimao Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Siu-Kie Au
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Shubin Wang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Mengsu Yang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
48
|
Castillo-Torres KY, McLamore ES, Arnold DP. A High-Throughput Microfluidic Magnetic Separation (µFMS) Platform for Water Quality Monitoring. MICROMACHINES 2019; 11:E16. [PMID: 31877902 PMCID: PMC7019623 DOI: 10.3390/mi11010016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/11/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022]
Abstract
The long-term aim of this work is to develop a biosensing system that rapidly detects bacterial targets of interest, such as Escherichia coli, in drinking and recreational water quality monitoring. For these applications, a standard sample size is 100 mL, which is quite large for magnetic separation microfluidic analysis platforms that typically function with <20 µL/s throughput. Here, we report the use of 1.5-µm-diameter magnetic microdisc to selectively tag target bacteria, and a high-throughput microfluidic device that can potentially isolate the magnetically tagged bacteria from 100 mL water samples in less than 15 min. Simulations and experiments show ~90% capture efficiencies of magnetic particles at flow rates up to 120 µL/s. Also, the platform enables the magnetic microdiscs/bacteria conjugates to be directly imaged, providing a path for quantitative assay.
Collapse
Affiliation(s)
- Keisha Y. Castillo-Torres
- Interdisciplinary Microsystems Group, Department of Electrical and Computer Engineering; University of Florida, Gainesville, FL 32611, USA;
| | - Eric S. McLamore
- Institute of Food and Agricultural Sciences, Department of Agricultural and Biological Engineering; University of Florida, Gainesville, FL 32611, USA;
| | - David P. Arnold
- Interdisciplinary Microsystems Group, Department of Electrical and Computer Engineering; University of Florida, Gainesville, FL 32611, USA;
| |
Collapse
|
49
|
Maia FR, Reis RL, Oliveira JM. Finding the perfect match between nanoparticles and microfluidics to respond to cancer challenges. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 24:102139. [PMID: 31843662 DOI: 10.1016/j.nano.2019.102139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 01/24/2023]
Abstract
The clinical translation of new cancer theranostic has been delayed by inherent cancer's heterogeneity. Additionally, this delay has been enhanced by the lack of an appropriate in vitro model, capable to produce accurate data. Nanoparticles and microfluidic devices have been used to obtain new and more efficient strategies to tackle cancer challenges. On one hand, nanoparticles-based therapeutics can be modified to target specific cells, and/or molecules, and/or modified with drugs, releasing them over time. On the other hand, microfluidic devices allow the exhibition of physiologically complex systems, incorporation of controlled flow, and control of the chemical environment. Herein, we review the use of nanoparticles and microfluidic devices to address different cancer challenges, such as detection of CTCs and biomarkers, point-of-care devices for early diagnosis and improvement of therapies. The future perspectives of cancer challenges are also addressed herein.
Collapse
Affiliation(s)
- F Raquel Maia
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal; ICVS/3B's PT Government Associate Lab, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Barco, Guimarães, Portugal.
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal; ICVS/3B's PT Government Associate Lab, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Barco, Guimarães, Portugal
| | - Joaquim M Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, Portugal; ICVS/3B's PT Government Associate Lab, Braga, Guimarães, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Barco, Guimarães, Portugal
| |
Collapse
|
50
|
Buscail E, Alix-Panabières C, Quincy P, Cauvin T, Chauvet A, Degrandi O, Caumont C, Verdon S, Lamrissi I, Moranvillier I, Buscail C, Marty M, Laurent C, Vendrely V, Moreau-Gaudry F, Bedel A, Dabernat S, Chiche L. High Clinical Value of Liquid Biopsy to Detect Circulating Tumor Cells and Tumor Exosomes in Pancreatic Ductal Adenocarcinoma Patients Eligible for Up-Front Surgery. Cancers (Basel) 2019; 11:cancers11111656. [PMID: 31717747 PMCID: PMC6895804 DOI: 10.3390/cancers11111656] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 02/08/2023] Open
Abstract
PURPOSE Expediting the diagnosis of pancreatic ductal adenocarcinoma (PDAC) would benefit care management, especially for the start of treatments requiring histological evidence. This study evaluated the combined diagnostic performance of circulating biomarkers obtained by peripheral and portal blood liquid biopsy in patients with resectable PDAC. EXPERIMENTAL DESIGN Liquid biopsies were performed in a prospective translational clinical trial (PANC-CTC #NCT03032913) including 22 patients with resectable PDAC and 28 noncancer controls from February to November 2017. Circulating tumor cells (CTCs) were detected using the CellSearch® method or after RosetteSep® enrichment combined with CRISPR/Cas9-improved KRAS mutant alleles quantification by droplet digital PCR. CD63 bead-coupled Glypican-1 (GPC1)-positive exosomes were quantified by flow cytometry. RESULTS Liquid biopsies were positive in 7/22 (32%), 13/22 (59%), and 14/22 (64%) patients with CellSearch® or RosetteSep®-based CTC detection or GPC1-positive exosomes, respectively, in peripheral and/or portal blood. Liquid biopsy performance was improved in portal blood only with CellSearch®, reaching 45% of PDAC identification (5/11) versus 10% (2/22) in peripheral blood. Importantly, combining CTC and GPC1-positive-exosome detection displayed 100% of sensitivity and 80% of specificity, with a negative predictive value of 100%. High levels of GPC1+-exosomes and/or CTC presence were significantly correlated with progression-free survival and with overall survival when CTC clusters were found. CONCLUSION This study is the first to evaluate combined CTC and exosome detection to diagnose resectable pancreatic cancers. Liquid biopsy combining several biomarkers could provide a rapid, reliable, noninvasive decision-making tool in early, potentially curable pancreatic cancer. Moreover, the prognostic value could select patients eligible for neoadjuvant treatment before surgery. This exploratory study deserves further validation.
Collapse
Affiliation(s)
- Etienne Buscail
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Centre of Montpellier, EA2415 Montpellier, France;
| | - Pascaline Quincy
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Thomas Cauvin
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Alexandre Chauvet
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Olivier Degrandi
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Charline Caumont
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Séverine Verdon
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
| | - Isabelle Lamrissi
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Isabelle Moranvillier
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Camille Buscail
- Nutritional Epidemiology Research Team (EREN), Paris 13 University, U1153 INSERM, U1125 Institut national de la recherche agronomique (INRA), Conservatoire national des arts et métiers (CNAM), Paris Cité Epidemiology and Statistics Research Center (CRESS), 93017 Bobigny, France;
| | - Marion Marty
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
| | - Christophe Laurent
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Véronique Vendrely
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - François Moreau-Gaudry
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Aurélie Bedel
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| | - Sandrine Dabernat
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
- Correspondence: ; Tel.: +33-(0)5-5757-1374; Fax: +33-(0)5-5757-1374
| | - Laurence Chiche
- U1035 Institut National de la Santé et de la Recherche Médicale (INSERM), 33000 Bordeaux, France; (E.B.); (P.Q.); (T.C.); (A.C.); (O.D.); (I.L.); (I.M.); (C.L.); (V.V.); (F.M.-G.); (A.B.); (L.C.)
- Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000 Bordeaux, France; (C.C.); (S.V.); (M.M.)
- Université de Bordeaux, 33076 Bordeaux, France
| |
Collapse
|