1
|
Guo X, Hu F, Yong Z, Zhao S, Wan Y, Wang B, Peng N. Magnetic Nanoparticle-Based Microfluidic Platform for Automated Enrichment of High-Purity Extracellular Vesicles. Anal Chem 2024; 96:7212-7219. [PMID: 38660946 DOI: 10.1021/acs.analchem.4c00795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Extracellular vesicles (EVs) are available in various biological fluids and have highly heterogeneous sizes, origins, contents, and functions. Rapid enrichment of high-purity EVs remains crucial for enhancing research on EVs in tumors. In this work, we present a magnetic nanoparticle-based microfluidic platform (ExoCPR) for on-chip isolation, purification, and mild recovery of EVs from cell culture supernatant and plasma within 29 min. The ExoCPR chip integrates bubble-driven micromixers and immiscible filtration assisted by surface tension (IFAST) technology. The bubble-driven micromixer enhances the mixing between immunomagnetic beads and EVs, eliminating the need for manual pipetting or off-chip oscillatory incubation. The high-purity EVs were obtained after passing through the immiscible phase interface where hydrophilic or hydrophobic impurities nonspecifically bound to SIMI were removed. The ExoCPR chip had a capture efficiency of 75.8% and a release efficiency of 62.7% for model EVs. We also demonstrated the powerful performance of the ExoCPR in isolating EVs from biological samples (>90% purity). This chip was further employed in clinical plasma samples and showed that the number of GPC3-positive EVs isolated from hepatocellular carcinoma patients was significantly higher than that of healthy individuals. This ExoCPR chip may provide a promising tool for EV-based liquid biopsy and other fundamental research.
Collapse
Affiliation(s)
- Xiaoniu Guo
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Fei Hu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Zhang Yong
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Shuhao Zhao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Yong Wan
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Bingqing Wang
- Xi'an Key Laboratory of Biomedical Testing and High-End Equipment, Xi'an 710049, Shaanxi, China
| | - Niancai Peng
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
- Xi'an Key Laboratory of Biomedical Testing and High-End Equipment, Xi'an 710049, Shaanxi, China
| |
Collapse
|
2
|
Rodriguez-Mateos P, Ngamsom B, Iles A, Pamme N. Microscale immiscible phase magnetic processing for bioanalytical applications. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
3
|
Rodems TS, Heninger E, Stahlfeld CN, Gilsdorf CS, Carlson KN, Kircher MR, Singh A, Krueger TEG, Beebe DJ, Jarrard DF, McNeel DG, Haffner MC, Lang JM. Reversible epigenetic alterations regulate class I HLA loss in prostate cancer. Commun Biol 2022; 5:897. [PMID: 36050516 PMCID: PMC9437063 DOI: 10.1038/s42003-022-03843-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 08/15/2022] [Indexed: 11/09/2022] Open
Abstract
Downregulation of HLA class I (HLA-I) impairs immune recognition and surveillance in prostate cancer and may underlie the ineffectiveness of checkpoint blockade. However, the molecular mechanisms regulating HLA-I loss in prostate cancer have not been fully explored. Here, we conducted a comprehensive analysis of HLA-I genomic, epigenomic and gene expression alterations in primary and metastatic human prostate cancer. Loss of HLA-I gene expression was associated with repressive chromatin states including DNA methylation, histone H3 tri-methylation at lysine 27, and reduced chromatin accessibility. Pharmacological DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibition decreased DNA methylation and increased H3 lysine 27 acetylation and resulted in re-expression of HLA-I on the surface of tumor cells. Re-expression of HLA-I on LNCaP cells by DNMT and HDAC inhibition increased activation of co-cultured prostate specific membrane antigen (PSMA)27-38-specific CD8+ T-cells. HLA-I expression is epigenetically regulated by functionally reversible DNA methylation and chromatin modifications in human prostate cancer. Methylated HLA-I was detected in HLA-Ilow circulating tumor cells (CTCs), which may serve as a minimally invasive biomarker for identifying patients who would benefit from epigenetic targeted therapies.
Collapse
Affiliation(s)
- Tamara S Rodems
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Erika Heninger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Charlotte N Stahlfeld
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Cole S Gilsdorf
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Kristin N Carlson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Madison R Kircher
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Anupama Singh
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Timothy E G Krueger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - David J Beebe
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Pathology, University of Wisconsin, Madison, 3170 UW Medical Foundation Centennial Building, 1685 Highland Ave., Madison, WI, 53705, USA
| | - David F Jarrard
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Urology, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Douglas G McNeel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Michael C Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, N., Seattle, WA, 98109, USA.,Department of Pathology, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA.,Department of Pathology, Johns Hopkins School of Medicine, 600N Wolfe St., Baltimore, MD, 21287, USA
| | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
4
|
Rodems TS, Juang DS, Stahlfeld CN, Gilsdorf CS, Krueger TEG, Heninger E, Zhao SG, Sperger JM, Beebe DJ, Haffner MC, Lang JM. SEEMLIS: a flexible semi-automated method for enrichment of methylated DNA from low-input samples. Clin Epigenetics 2022; 14:37. [PMID: 35272673 PMCID: PMC8908705 DOI: 10.1186/s13148-022-01252-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/18/2022] [Indexed: 01/02/2023] Open
Abstract
Background DNA methylation alterations have emerged as hallmarks of cancer and have been proposed as screening, prognostic, and predictive biomarkers. Traditional approaches for methylation analysis have relied on bisulfite conversion of DNA, which can damage DNA and is not suitable for targeted gene analysis in low-input samples. Here, we have adapted methyl-CpG-binding domain protein 2 (MBD2)-based DNA enrichment for use on a semi-automated exclusion-based sample preparation (ESP) platform for robust and scalable enrichment of methylated DNA from low-input samples, called SEEMLIS. Results We show that combining methylation-sensitive enzyme digestion with ESP-based MBD2 enrichment allows for single gene analysis with high sensitivity for GSTP1 in highly impure, heterogenous samples. We also show that ESP-based MBD2 enrichment coupled with targeted pre-amplification allows for analysis of multiple genes with sensitivities approaching the single cell level in pure samples for GSTP1 and RASSF1 and sensitivity down to 14 cells for these genes in highly impure samples. Finally, we demonstrate the potential clinical utility of SEEMLIS by successful detection of methylated gene signatures in circulating tumor cells (CTCs) from patients with prostate cancer with varying CTC number and sample purity. Conclusions SEEMLIS is a robust assay for targeted DNA methylation analysis in low-input samples, with flexibility at multiple steps. We demonstrate the feasibility of this assay to analyze DNA methylation in prostate cancer cells using CTCs from patients with prostate cancer as a real-world example of a low-input analyte of clinical importance. In summary, this novel assay provides a platform for determining methylation signatures in rare cell populations with broad implications for research as well as clinical applications. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01252-4.
Collapse
Affiliation(s)
- Tamara S Rodems
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Duane S Juang
- Department of Pathology, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA
| | - Charlotte N Stahlfeld
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Cole S Gilsdorf
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Tim E G Krueger
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Erika Heninger
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Jamie M Sperger
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - David J Beebe
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA.,Department of Pathology, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Michael C Haffner
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, N., Seattle, WA, 98109, USA.,Department of Pathology, University of Washington, 1959 NE Pacific St., Seattle, WA, 98195, USA.,Department of Pathology, Johns Hopkins School of Medicine, 600 N Wolfe St., Baltimore, MD, 21287, USA
| | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,7151 WI Institutes for Medical Research, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
5
|
Sperger JM, Feng FY, Armstrong AJ, Zhao SG, Lang JM. Reply to M. K. Bos et al. J Clin Oncol 2021; 40:520-522. [PMID: 34878806 DOI: 10.1200/jco.21.02238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jamie M Sperger
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| | - Felix Y Feng
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| | - Andrew J Armstrong
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| | - Shuang G Zhao
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| | - Joshua M Lang
- Jamie M. Sperger, PhD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI; Felix Y. Feng, MD, Helen Diller Family Comprehensive Cancer Center, Division of Hematology and Oncology, Department of Medicine, UCSF, Department of Radiation Oncology, Department of Urology, University of California San Francisco, San Francisco, CA; Andrew J. Armstrong, MD, Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, Durham, NC; Shuang G. Zhao, MD, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Human Oncology, University of Wisconsin, Madison, WI, William S. Middleton Memorial Veterans Hospital, Madison, WI; and Joshua M. Lang, MD, MS, Carbone Cancer Center and Department of Medicine, University of Wisconsin, Madison, WI, Department of Medicine, University of Wisconsin, Madison, WI
| |
Collapse
|
6
|
Robust and easy-to-use microchip electrophoresis within sub-millimeter channels for fast and highly efficient separation. Talanta 2021; 235:122747. [PMID: 34517615 DOI: 10.1016/j.talanta.2021.122747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 11/20/2022]
Abstract
Microchip capillary electrophoresis (MCE) is a powerful technique for rapid separation; however, its acceptance in routine laboratories is still limited. Compromises caused by the efforts for solving different problems, such as reducing its cost of fabrication and ensuring high separation efficiency, undermine the competitiveness of this technology compared to other separation techniques. Contrary to the conventional pursuit of narrow microchannels, this study investigated the suitability of microchips with channels at the sub-millimeter level, targeting the simplification of the overall operation, cost reduction, and robustness improvement. To this effect, we considered the influence of pressurized flow and Joule heating on the separation. The suppression of pressurized flow with viscous solutions was confirmed through a combination of simulations and experimental results, indicating that the buffer viscosity was enough for successful separation. We fabricated channels of 200 μm × 230 μm using computer numerical controlled (CNC) machining and obtained theoretical plate numbers of 4.8 × 105 m-1 and 5.3 × 105 m-1 for fluorescein isothiocyanate (FITC) labeled small molecules and DNA fragments, respectively, with a buffer viscosity of 168 mPa s (0.5 % hydroxypropyl methylcellulose, HPMC). These values are comparable with that of narrow-bore microchips. Furthermore, we did not observe any deleterious effects with low-conductivity buffers. We investigated the rapid and highly sensitive detection of mycoplasma contamination and the real samples of circulating cell-free DNA (cfDNA), which gave a limit of detection (LOD) as low as 2.3 ng mL-1. Owing to the significant reduction in cost, ease of operation, and fast separation capabilities demonstrated in this work, MCE can be a viable alternative to the usual slab gel electrophoresis running in most biological laboratories.
Collapse
|
7
|
Juang DS, Juang TD, Dudley DM, Newman CM, Accola MA, Rehrauer WM, Friedrich TC, O'Connor DH, Beebe DJ. Oil immersed lossless total analysis system for integrated RNA extraction and detection of SARS-CoV-2. Nat Commun 2021; 12:4317. [PMID: 34262053 PMCID: PMC8280165 DOI: 10.1038/s41467-021-24463-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 06/10/2021] [Indexed: 12/03/2022] Open
Abstract
The COVID-19 pandemic exposed difficulties in scaling current quantitative PCR (qPCR)-based diagnostic methodologies for large-scale infectious disease testing. Bottlenecks include lengthy multi-step processes for nucleic acid extraction followed by qPCR readouts, which require costly instrumentation and infrastructure, as well as reagent and plastic consumable shortages stemming from supply chain constraints. Here we report an Oil Immersed Lossless Total Analysis System (OIL-TAS), which integrates RNA extraction and detection onto a single device that is simple, rapid, cost effective, and requires minimal supplies and infrastructure to perform. We validated the performance of OIL-TAS using contrived SARS-CoV-2 viral particle samples and clinical nasopharyngeal swab samples. OIL-TAS showed a 93% positive predictive agreement (n = 57) and 100% negative predictive agreement (n = 10) with clinical SARS-CoV-2 qPCR assays in testing clinical samples, highlighting its potential to be a faster, cheaper, and easier-to-deploy alternative for infectious disease testing. Bottlenecks in qPCR-based COVID-19 diagnostics include the lengthy multistep process and reagent shortages. Here the authors report OIL-TAS which integrates RNA extraction and detection into a single device.
Collapse
Affiliation(s)
- Duane S Juang
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Terry D Juang
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Dawn M Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Christina M Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Molly A Accola
- UW Health Clinical Laboratories, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - William M Rehrauer
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA.,UW Health Clinical Laboratories, University of Wisconsin Hospital and Clinics, Madison, WI, USA
| | - Thomas C Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA. .,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
8
|
Labib M, Kelley SO. Circulating tumor cell profiling for precision oncology. Mol Oncol 2021; 15:1622-1646. [PMID: 33448107 PMCID: PMC8169448 DOI: 10.1002/1878-0261.12901] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/19/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Analysis of circulating tumor cells (CTCs) collected from patient's blood offers a broad range of opportunities in the field of precision oncology. With new advances in profiling technology, it is now possible to demonstrate an association between the molecular profiles of CTCs and tumor response to therapy. In this Review, we discuss mechanisms of tumor resistance to therapy and their link to phenotypic and genotypic properties of CTCs. We summarize key technologies used to isolate and analyze CTCs and discuss recent clinical studies that examined CTCs for genomic and proteomic predictors of responsiveness to therapy. We also point out current limitations that still hamper the implementation of CTCs into clinical practice. We finally reflect on how these shortcomings can be addressed with the likely contribution of multiparametric approaches and advanced data analytics.
Collapse
Affiliation(s)
- Mahmoud Labib
- Department of Pharmaceutical SciencesUniversity of TorontoCanada
| | - Shana O. Kelley
- Department of Pharmaceutical SciencesUniversity of TorontoCanada
- Institute for Biomaterials and Biomedical EngineeringUniversity of TorontoCanada
- Department of BiochemistryUniversity of TorontoCanada
- Department of ChemistryUniversity of TorontoCanada
| |
Collapse
|
9
|
Sharma S, Bhatia V. Magnetic nanoparticles in microfluidics-based diagnostics: an appraisal. Nanomedicine (Lond) 2021; 16:1329-1342. [PMID: 34027677 DOI: 10.2217/nnm-2021-0007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The use of magnetic nanoparticles (MNPs) in microfluidics based diagnostics is a classic case of micro-, nano- and bio-technology coming together to design extremely controllable, reproducible, and scalable nano and micro 'on-chip bio sensing systems.' In this review, applications of MNPs in microfluidics ranging from molecular diagnostics and immunodiagnostics to clinical uses have been examined. In addition, microfluidic mixing and capture of analytes using MNPs, and MNPs as carriers in microfluidic devices has been investigated. Finally, the challenges and future directions of this upcoming field have been summarized. The use of MNP-based microfluidic devices, will help in developing decentralized or 'point of care' testing globally, contributing to affordable healthcare, particularly, for middle- and low-income developing countries.
Collapse
Affiliation(s)
- Smriti Sharma
- Department of Chemistry, Miranda House, University of Delhi, India
| | - Vinayak Bhatia
- ICARE Eye Hospital & Postgraduate Institute, Noida, U.P., India
| |
Collapse
|
10
|
Chen Y, Liu Y, Shi Y, Ping J, Wu J, Chen H. Magnetic particles for integrated nucleic acid purification, amplification and detection without pipetting. Trends Analyt Chem 2020; 127:115912. [PMID: 32382202 PMCID: PMC7202819 DOI: 10.1016/j.trac.2020.115912] [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] [Indexed: 12/18/2022]
Abstract
Nucleic acid amplification based detection plays an important role in food safety, environmental monitoring and clinical diagnosis. However, traditional nucleic acid detection process involves transferring liquid from one tube to another by pipetting. It requires trained persons, equipped labs and consumes lots of time. The ideal nucleic acid detection is integrated, closed, simplified and automated. Magnetic particles actuated by magnetic fields can efficiently adsorb nucleic acids and promote integrated nucleic acid assays without pipetting driven by pumps and centrifuges. We will comprehensively review magnetic particles assisted integrated system for nucleic acid detection and hope it can inspire further related study.
Collapse
Key Words
- ATP, adenosine triphosphate
- DLS, dynamic light scattering
- FMR, ferromagnetic resonance
- GTC, guanidinium thiocyanate
- ICP-AES, inductively coupled plasma atomic emission spectroscopy
- IFAST, immiscible filtration assisted by surface tension
- Immiscible interface
- Integrated detection
- LAMP, loop-mediated isothermal amplification
- Magnetic particles
- Nucleic acid
- PCR, polymerase chain reaction
- PEG, polyethylene glycol
- POCT, point-of-care testing
- RPA, recombinase polymerase amplification
- SQUID, superconducting quantum interference device magnetometer
- TEM, transmission electron microscopy
- XRD, X-Ray diffraction
- qPCR, quantitative PCR
Collapse
Affiliation(s)
- Yanju Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Yang Liu
- Key Laboratory of Microbiol Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, 310012, China
| | - Ya Shi
- Key Laboratory of Microbiol Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, 310012, China
| | - Jianfeng Ping
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jian Wu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of on Site Processing Equipment for Agricultural Products, Ministry of Agriculture, Hangzhou, 310058, China
| | - Huan Chen
- Key Laboratory of Microbiol Technology and Bioinformatics of Zhejiang Province, Zhejiang Institute of Microbiology, Hangzhou, 310012, China
| |
Collapse
|
11
|
Tokar JJ, Stahlfeld CN, Sperger JM, Niles DJ, Beebe DJ, Lang JM, Warrick JW. Pairing Microwell Arrays with an Affordable, Semiautomated Single-Cell Aspirator for the Interrogation of Circulating Tumor Cell Heterogeneity. SLAS Technol 2020; 25:162-176. [PMID: 31983266 PMCID: PMC8879417 DOI: 10.1177/2472630319898146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Comprehensive analysis of tumor heterogeneity requires robust methods for the isolation and analysis of single cells from patient samples. An ideal approach would be fully compatible with downstream analytic methods, such as advanced genomic testing. These endpoints necessitate the use of live cells at high purity. A multitude of microfluidic circulating tumor cell (CTC) enrichment technologies exist, but many of those perform bulk sample enrichment and are not, on their own, capable of single-cell interrogation. To address this, we developed an affordable semiautomated single-cell aspirator (SASCA) to further enrich rare-cell populations from a specialized microwell array, per their phenotypic markers. Immobilization of cells within microwells, integrated with a real-time image processing software, facilitates the detection and precise isolation of targeted cells that have been optimally seeded into the microwells. Here, we demonstrate the platform capabilities through the aspiration of target cells from an impure background population, where we obtain purity levels of 90%-100% and demonstrate the enrichment of the target population with high-quality RNA extraction. A range of low cell numbers were aspirated using SASCA before undergoing whole transcriptome and genome analysis, exhibiting the ability to obtain endpoints from low-template inputs. Lastly, CTCs from patients with castration-resistant prostate cancer were isolated with this platform and the utility of this method was confirmed for rare-cell isolation. SASCA satisfies a need for an affordable option to isolate single cells or highly purified subpopulations of cells to probe complex mechanisms driving disease progression and resistance in patients with cancer.
Collapse
Affiliation(s)
- Jacob J Tokar
- Dept. of Biomedical Eng. – Univ. of Wisconsin, Madison - Madison, USA
| | | | - Jamie M Sperger
- Dept. of Medicine – Univ. of Wisconsin, Madison - Madison, USA
| | - David J Niles
- Dept. of Biomedical Eng. – Univ. of Wisconsin, Madison - Madison, USA
| | - David J Beebe
- Dept. of Biomedical Eng. – Univ. of Wisconsin, Madison - Madison, USA
- UW Carbone Cancer Center. – Univ. of Wisconsin, Madison - Madison, USA
| | - Joshua M Lang
- UW Carbone Cancer Center. – Univ. of Wisconsin, Madison - Madison, USA
- Dept. of Medicine – Univ. of Wisconsin, Madison - Madison, USA
| | - Jay W Warrick
- Dept. of Biomedical Eng. – Univ. of Wisconsin, Madison - Madison, USA
| |
Collapse
|
12
|
Mu T, Toyoda H, Kimura Y, Yamada M, Utoh R, Umeno D, Seki M. Laborless, Automated Microfluidic Tandem Cell Processor for Visualizing Intracellular Molecules of Mammalian Cells. Anal Chem 2020; 92:2580-2588. [PMID: 31822057 DOI: 10.1021/acs.analchem.9b04288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Visualization and quantification of intracellular molecules of mammalian cells are crucial steps in clinical diagnosis, drug development, and basic biological research. However, conventional methods rely mostly on labor-intensive, centrifugation-based manual operations for exchanging the cell carrier medium and have limited reproducibility and recovery efficiency. Here we present a microfluidic cell processor that can perform four-step exchange of carrier medium, simply by introducing a cell suspension and fluid reagents into the device. The reaction time period for each reaction step, including fixation, membrane permeabilization, and staining, was tunable in the range of 2 to 15 min by adjusting the volume of the reaction tube connecting the neighboring exchanger modules. We double-stained the cell nucleus and cytoskeleton (F-actin) using the presented device with an overall reaction period of ∼30 min, achieving a high recovery ratio and high staining efficiency. Additionally, intracellular cytokine (IL-2) was visualized for T cells to demonstrate the feasibility of the device as a pretreatment system for downstream flow-cytometric analysis. The presented approach would facilitate the development of laborless, automated microfluidic systems that integrate cell processing and analysis operations and would pave a new path to high-throughput biological experiments.
Collapse
Affiliation(s)
- Tinglin Mu
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering , Chiba University , 1-33 Yayoi-cho , Inage-ku , Chiba 263-8522 , Japan
| | - Hajime Toyoda
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering , Chiba University , 1-33 Yayoi-cho , Inage-ku , Chiba 263-8522 , Japan
| | - Yuki Kimura
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering , Chiba University , 1-33 Yayoi-cho , Inage-ku , Chiba 263-8522 , Japan
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering , Chiba University , 1-33 Yayoi-cho , Inage-ku , Chiba 263-8522 , Japan
| | - Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering , Chiba University , 1-33 Yayoi-cho , Inage-ku , Chiba 263-8522 , Japan
| | - Daisuke Umeno
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering , Chiba University , 1-33 Yayoi-cho , Inage-ku , Chiba 263-8522 , Japan
| | - Minoru Seki
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering , Chiba University , 1-33 Yayoi-cho , Inage-ku , Chiba 263-8522 , Japan
| |
Collapse
|
13
|
Zhong R, Hou L, Zhao Y, Wang T, Wang S, Wang M, Xu D, Sun Y. A 3D mixing-based portable magnetic device for fully automatic immunofluorescence staining of γ-H2AX in UVC-irradiated CD4 + cells. RSC Adv 2020; 10:29311-29319. [PMID: 35521108 PMCID: PMC9055984 DOI: 10.1039/d0ra03925j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/02/2020] [Indexed: 11/22/2022] Open
Abstract
Immunofluorescence (IF) is a common method used in cell biology. The conventional protocol for IF staining is time and labor-intensive, operator dependent and reagent-consuming. Magnetic Bead (MB)-based microdevices are frequently utilized in cellular assays, but integration of simple and efficient mixing with downstream multi-step manipulation of MBs for automatic IF staining is still challenging. We herein present a portable, inexpensive and integratable device for MB-based automatic IF staining. First, a front-end cell capture step is performed using a 3D-mixing module, which is built upon a novel mechanism named ec-2MagRotors and generates periodically changing 3D magnetic fields. A 5-fold enhancement of cell capture efficiency was attained even with a low bead-to-cell concentration ratio (5 : 1), when conducting magnetic 3D mixing. Second, a 1D-moving module is employed downstream to automatically manipulate MB–cell complexes for IF staining. Further, a simplified protocol for staining of γ-H2AX, a biomarker widely used in evaluation of cell radiation damage, is presented for proof-of-principle study of the magnetic device. Using UVC-irradiated CD4+ cells as samples, our device achieved fully automatic γ-H2AX staining within 40 minutes at room temperature and showed a linear dose–response relationship. The developed portable magnetic device is automatic, efficient, cost-effective and simple-to-use, holding great potential for applications in different IF assays. A 3D mixing-based portable magnetic device to perform on-chip efficient cell capture and automatic intracellular immunofluorescence (IF) staining is presented.![]()
Collapse
Affiliation(s)
- Runtao Zhong
- Institute of Environmental Systems Biology
- Dalian Maritime University
- Dalian 116026
- China
| | - Liangsheng Hou
- College of Marine Engineering
- Dalian Maritime University, Dalian
- Dalian 116026
- China
| | - Yingbo Zhao
- Institute of Environmental Systems Biology
- Dalian Maritime University
- Dalian 116026
- China
| | - Tianle Wang
- Institute of Environmental Systems Biology
- Dalian Maritime University
- Dalian 116026
- China
| | - Shaohua Wang
- Institute of Environmental Systems Biology
- Dalian Maritime University
- Dalian 116026
- China
| | - Mengyu Wang
- Institute of Environmental Systems Biology
- Dalian Maritime University
- Dalian 116026
- China
| | - Dan Xu
- Institute of Environmental Systems Biology
- Dalian Maritime University
- Dalian 116026
- China
| | - Yeqing Sun
- Institute of Environmental Systems Biology
- Dalian Maritime University
- Dalian 116026
- China
| |
Collapse
|
14
|
Li W, Li R, Huang B, Wang Z, Sun Y, Wei X, Heng C, Liu W, Yu M, Guo SS, Zhao XZ. TiO 2 nanopillar arrays coated with gelatin film for efficient capture and undamaged release of circulating tumor cells. NANOTECHNOLOGY 2019; 30:335101. [PMID: 30965310 DOI: 10.1088/1361-6528/ab176c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Circulating tumor cells (CTCs) are important for the detection and treatment of cancer. Nevertheless, a low density of circulating tumor cells makes the capture and release of CTCs an obstacle. In this work, TiO2 nanopillar arrays coated with gelatin film were synthesized for efficient capture and undamaged release of circulating tumor cells. The scanning electron microscope and atomic force microscope images demonstrate that the substrate has a certain roughness. The interaction between the cell membrane and the nanostructure substrate contributes to the efficient capture of CTC (capture efficiency up to 94.98%). The gelatin layer has excellent biocompatibility and can be rapidly digested by matrix metalloproteinase (MMP9), which realizes the non-destructive release of CTCs (0.1 mg ml-1, 5 min, nearly 100% release efficiency, activity 100%). Therefore, by our strategy, the CTCs can be efficiently captured and released undamaged, which is important for subsequent analysis.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Iliescu FS, Poenar DP, Yu F, Ni M, Chan KH, Cima I, Taylor HK, Cima I, Iliescu C. Recent advances in microfluidic methods in cancer liquid biopsy. BIOMICROFLUIDICS 2019; 13:041503. [PMID: 31431816 PMCID: PMC6697033 DOI: 10.1063/1.5087690] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/24/2019] [Indexed: 05/04/2023]
Abstract
Early cancer detection, its monitoring, and therapeutical prediction are highly valuable, though extremely challenging targets in oncology. Significant progress has been made recently, resulting in a group of devices and techniques that are now capable of successfully detecting, interpreting, and monitoring cancer biomarkers in body fluids. Precise information about malignancies can be obtained from liquid biopsies by isolating and analyzing circulating tumor cells (CTCs) or nucleic acids, tumor-derived vesicles or proteins, and metabolites. The current work provides a general overview of the latest on-chip technological developments for cancer liquid biopsy. Current challenges for their translation and their application in various clinical settings are discussed. Microfluidic solutions for each set of biomarkers are compared, and a global overview of the major trends and ongoing research challenges is given. A detailed analysis of the microfluidic isolation of CTCs with recent efforts that aimed at increasing purity and capture efficiency is provided as well. Although CTCs have been the focus of a vast microfluidic research effort as the key element for obtaining relevant information, important clinical insights can also be achieved from alternative biomarkers, such as classical protein biomarkers, exosomes, or circulating-free nucleic acids. Finally, while most work has been devoted to the analysis of blood-based biomarkers, we highlight the less explored potential of urine as an ideal source of molecular cancer biomarkers for point-of-care lab-on-chip devices.
Collapse
Affiliation(s)
- Florina S. Iliescu
- School of Applied Science, Republic Polytechnic, Singapore 738964, Singapore
| | - Daniel P. Poenar
- VALENS-Centre for Bio Devices and Signal Analysis, School of EEE, Nanyang Technological University, Singapore 639798, Singapore
| | - Fang Yu
- Singapore Institute of Manufacturing Technology, A*STAR, Singapore 138634, Singapore
| | - Ming Ni
- School of Biological Sciences and Engineering, Yachay Technological University, San Miguel de Urcuquí 100105, Ecuador
| | - Kiat Hwa Chan
- Division of Science, Yale-NUS College, Singapore 138527, Singapore
| | | | - Hayden K. Taylor
- Department of Mechanical Engineering, University of California, Berkeley, California 94720, USA
| | - Igor Cima
- DKFZ-Division of Translational Oncology/Neurooncology, German Cancer Consortium (DKTK), Heidelberg and University Hospital Essen, Essen 45147, Germany
| | | |
Collapse
|
16
|
Pirozzi I, Snider A, Kraus M, Schönbrunner ER, Tripathi A. Microfluidic Immiscible Phase Filtration System for the Isolation of Small Numbers of Cells from Whole Blood. Cytometry A 2019; 95:885-897. [PMID: 30852843 DOI: 10.1002/cyto.a.23736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 12/24/2018] [Accepted: 02/05/2019] [Indexed: 11/09/2022]
Abstract
Isolation of circulating tumor cells (CTCs) has generated clinical and academic interest due to the important role that CTCs play in cancer metastasis and diagnosis. Here, we present a PDMS and glass prototype of a microfluidic device for the immunomagnetic, immiscible phase filtration based capture, and isolation of MCF-7 breast cancer cells, from various sample matrices including PBS-based buffer, blood plasma, and unprocessed whole blood. Following optimization of surface energy of an oil-water interface, microfluidic geometry, and bead-binding kinematics, our microfluidic device achieved 95 ± 4% recovery of target cells from PBS-based buffer with 95% purity, 90 ± 3% recovery of target cells from blood plasma and recovery of ~70 ± 5% from unprocessed whole blood with purity >99% with 1 ml blood samples with 1,000 spiked target cells. From quantitative studies to assess the nonspecific carryover of contaminants from whole blood, we found that our system accomplishes a >175 fold depletion in platelets, >900 fold depletion in erythrocytes, and >1,700 fold depletion in leukocytes with respect to unprocessed whole blood, enabling us to avoid sample pre-processing. In addition, we found that ~95% of the isolated target cells were viable, making them suitable for subsequent molecular and cellular studies. We quantify and propose mechanisms for the carryover of platelet, erythrocyte, and leukocyte contamination in purified samples, rather than relying on sample pre-processing. These results validate the continued study of our platform for extraction of CTCs from patient samples and other rare cell isolation applications. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Ileana Pirozzi
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island
| | - Adam Snider
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island
| | - Morey Kraus
- PerkinElmer, 940 Winter St, Waltham, Massachusetts
| | | | - Anubhav Tripathi
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island
| |
Collapse
|
17
|
Hu F, Li J, Peng N, Li Z, Zhang Z, Zhao S, Duan M, Tian H, Li L, Zhang P. Rapid isolation of cfDNA from large-volume whole blood on a centrifugal microfluidic chip based on immiscible phase filtration. Analyst 2019; 144:4162-4174. [DOI: 10.1039/c9an00493a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The C-IFAST device enabled the rapid isolation of cfDNA, from 4 ml whole blood to 50 μl elution, within 15 min.
Collapse
Affiliation(s)
- Fei Hu
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Juan Li
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Niancai Peng
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Zheng Li
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Zengming Zhang
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Shuhao Zhao
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Mingyue Duan
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Hui Tian
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Lei Li
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| | - Peng Zhang
- State Key Laboratory for Manufacturing Systems Engineering
- Xi'an Jiaotong University
- Xi'an
- P. R. China
| |
Collapse
|
18
|
Yang Y, Zeng Y. Microfluidic communicating vessel chip for expedited and automated immunomagnetic assays. LAB ON A CHIP 2018; 18:3830-3839. [PMID: 30394473 PMCID: PMC6279511 DOI: 10.1039/c8lc00927a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Rapid, sensitive analysis of protein biomarkers is of tremendous biological and clinical significance. Immunoassays are workhorse tools for protein analysis and have been under continuous investigation to develop new methods and to improve the analytical performance. Herein we report a pneumatically gated microfluidic communicating vessel (μCOVE) chip for rapid and sensitive immunomagnetic ELISA. A distinct feature of our device is that it employs the communicating vessel principle as a simple means to generate a fast transient hydrodynamic flow to enable effective flow washing without the need for excessive incubation, which greatly simplifies and expedites the assay workflow, compared to conventional microfluidic flow-based immunoassays. Stationary multi-phase microfluidic techniques have been developed for fast bead washing. However, they have some limitations, such as the need for careful control of interfacial properties, large bead quantity required for reliable interphase bead transport, and relatively high bead loss during surface tension-gated traverse. Our single-phase μCOVE chip can overcome such limitations and facilitate the manipulation of magnetic beads to streamline the assay workflow. We showed that the μCOVE device affords highly sensitive quantification of the CEA and EGFR proteins with a LOD down to the sub-picogram per mL level. Direct detection of the EGFR in the crude A431 cell lysate was also demonstrated to further validate the ability of our device for rapid and quantitative analysis of complex biological samples. Overall, our work presents a unique platform that combines the merits of the stationary multi-phase systems and the flow-based microfluidics. This novel immunoassay microsystem has promising potential for a broad range of biological and clinical applications, owing to its simplicity and high performance.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA.
| | | |
Collapse
|
19
|
Pezzi HM, Guckenberger DJ, Schehr JL, Rothbauer J, Stahlfeld C, Singh A, Horn S, Schultz ZD, Bade RM, Sperger JM, Berry SM, Lang JM, Beebe DJ. Versatile exclusion-based sample preparation platform for integrated rare cell isolation and analyte extraction. LAB ON A CHIP 2018; 18:3446-3458. [PMID: 30334061 PMCID: PMC6402328 DOI: 10.1039/c8lc00620b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Rare cell populations provide a patient-centric tool to monitor disease treatment, response, and resistance. However, understanding rare cells is a complex problem, which requires cell isolation/purification and downstream molecular interrogation - processes challenged by non-target populations, which vary patient-to-patient and change with disease. As such, cell isolation platforms must be amenable to a range of sample types while maintaining high efficiency and purity. The multiplexed technology for automated extraction (mTAE) is a versatile magnetic bead-based isolation platform that facilitates positive, negative, and combinatorial selection with integrated protein staining and nucleic acid isolation. mTAE is validated by isolating circulating tumor cells (CTCs) - a model rare cell population - from breast and prostate cancer patient samples. Negative selection yielded high efficiency capture of CTCs while positive selection yielded higher purity with an average of only 95 contaminant cells captured per milliliter of processed whole blood. With combinatorial selection, an overall increase in capture efficiency was observed, highlighting the potential significance of integrating multiple capture approaches on a single platform. Following capture (and staining), on platform nucleic acid extraction enabled the detection of androgen receptor-related transcripts from CTCs isolated from prostate cancer patients. The flexibility (e.g. negative, positive, combinatorial selection) and capabilities (e.g. isolation, protein staining, and nucleic acid extraction) of mTAE enable users to freely interrogate specific cell populations, a capability required to understand the potential of emerging rare cell populations and readily adapt to the heterogeneity presented across clinical samples.
Collapse
Affiliation(s)
- Hannah M Pezzi
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Burinaru TA, Avram M, Avram A, Mărculescu C, Ţîncu B, Ţucureanu V, Matei A, Militaru M. Detection of Circulating Tumor Cells Using Microfluidics. ACS COMBINATORIAL SCIENCE 2018; 20:107-126. [PMID: 29363937 DOI: 10.1021/acscombsci.7b00146] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Metastasis is the main cause of death in cancer patients worldwide. During metastasis, cancer cells detach from the primary tumor and invade distant tissue. The cells that undergo this process are called circulating tumor cells (CTCs). Studies show that the number of CTCs in the peripheral blood can predict progression-free survival and overall survival and can be informative concerning the efficacy of treatment. Research is now concentrated on developing devices that can detect CTCs in the blood of cancer patients with improved sensitivity and specificity that can lead to improved clinical evaluation. This review focuses on devices that detect and capture CTCs using different cell properties (surface markers, size, deformability, electrical properties, etc.). We also discuss the process of tumor cell dissemination, the biology of CTCs, epithelial-mesenchymal transition (EMT), and several challenges and clinical applications of CTC detection.
Collapse
Affiliation(s)
- Tiberiu A. Burinaru
- National Institute for R&D in Microtechnologies, IMT-Bucharest, Bucharest, Romania, 077190
| | - Marioara Avram
- National Institute for R&D in Microtechnologies, IMT-Bucharest, Bucharest, Romania, 077190
| | - Andrei Avram
- National Institute for R&D in Microtechnologies, IMT-Bucharest, Bucharest, Romania, 077190
| | - Cătălin Mărculescu
- National Institute for R&D in Microtechnologies, IMT-Bucharest, Bucharest, Romania, 077190
| | - Bianca Ţîncu
- National Institute for R&D in Microtechnologies, IMT-Bucharest, Bucharest, Romania, 077190
| | - Vasilica Ţucureanu
- National Institute for R&D in Microtechnologies, IMT-Bucharest, Bucharest, Romania, 077190
| | - Alina Matei
- National Institute for R&D in Microtechnologies, IMT-Bucharest, Bucharest, Romania, 077190
| | - Manuella Militaru
- University of Agronomic
Sciences and Veterinary Medicine, Bucharest, Romania, 050097
| |
Collapse
|
21
|
Heninger E, Krueger TEG, Thiede SM, Sperger JM, Byers BL, Kircher MR, Kosoff D, Yang B, Jarrard DF, McNeel DG, Lang JM. Inducible expression of cancer-testis antigens in human prostate cancer. Oncotarget 2018; 7:84359-84374. [PMID: 27769045 PMCID: PMC5341296 DOI: 10.18632/oncotarget.12711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 10/11/2016] [Indexed: 12/23/2022] Open
Abstract
Immune tolerance to self-antigens can limit robust anti-tumor immune responses in the use of tumor vaccines. Expression of novel tumor associated antigens can improve immune recognition and lysis of tumor cells. The cancer-testis antigen (CTA) family of proteins has been hypothesized to be an ideal class of antigens due to tumor-restricted expression, a subset of which have been found to induce antibody responses in patients with prostate disease. We demonstrate that CTA expression is highly inducible in five different Prostate Cancer (PC) cell lines using a hypomethylating agent 5-Aza-2′-deoxycytidine (5AZA) and/or a histone deacetylase inhibitor LBH589. These CTAs include NY-ESO1, multiple members of the MAGE and SSX families and NY-SAR35. A subset of CTAs is synergistically induced by the combination of 5AZA and LBH589. We developed an ex vivo organ culture using human PC biopsies for ex vivo drug treatments to evaluate these agents in clinical samples. These assays found significant induction of SSX2 in 9/9 distinct patient samples and NY-SAR35 in 7/9 samples. Further, we identify expression of SSX2 in circulating tumor cells (CTC) from patients with advanced PC. These results indicate that epigenetic modifying agents can induce expression of a broad range of neoantigens in human PC and may serve as a useful adjunctive therapy with novel tumor vaccines and checkpoint inhibitors.
Collapse
Affiliation(s)
- Erika Heninger
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Timothy E G Krueger
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Stephanie M Thiede
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Jamie M Sperger
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Brianna L Byers
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Madison R Kircher
- University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Bing Yang
- Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - David F Jarrard
- Department of Urology, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin, Madison, Madison, WI 53705, USA.,University of Wisconsin Carbone Cancer Center, Madison, Madison, WI 53705, USA
| |
Collapse
|
22
|
Jan YJ, Chen JF, Zhu Y, Lu YT, Chen SH, Chung H, Smalley M, Huang YW, Dong J, Chen LC, Yu HH, Tomlinson JS, Hou S, Agopian VG, Posadas EM, Tseng HR. NanoVelcro rare-cell assays for detection and characterization of circulating tumor cells. Adv Drug Deliv Rev 2018; 125:78-93. [PMID: 29551650 PMCID: PMC5993593 DOI: 10.1016/j.addr.2018.03.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
Circulating tumor cells (CTCs) are cancer cells shredded from either a primary tumor or a metastatic site and circulate in the blood as the potential cellular origin of metastasis. By detecting and analyzing CTCs, we will be able to noninvasively monitor disease progression in individual cancer patients and obtain insightful information for assessing disease status, thus realizing the concept of "tumor liquid biopsy". However, it is technically challenging to identify CTCs in patient blood samples because of the extremely low abundance of CTCs among a large number of hematologic cells. In order to address this challenge, our research team at UCLA pioneered a unique concept of "NanoVelcro" cell-affinity substrates, in which CTC capture agent-coated nanostructured substrates were utilized to immobilize CTCs with remarkable efficiency. Four generations of NanoVelcro CTC assays have been developed over the past decade for a variety of clinical utilities. The 1st-gen NanoVelcro Chips, composed of a silicon nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer, were created for CTC enumeration. The 2nd-gen NanoVelcro Chips (i.e., NanoVelcro-LMD), based on polymer nanosubstrates, were developed for single-CTC isolation in conjunction with the use of the laser microdissection (LMD) technique. By grafting thermoresponsive polymer brushes onto SiNS, the 3rd-gen Thermoresponsive NanoVelcro Chips have demonstrated the capture and release of CTCs at 37 and 4 °C respectively, thereby allowing for rapid CTC purification while maintaining cell viability and molecular integrity. Fabricated with boronic acid-grafted conducting polymer-based nanomaterial on chip surface, the 4th-gen NanoVelcro Chips (Sweet chip) were able to purify CTCs with well-preserved RNA transcripts, which could be used for downstream analysis of several cancer specific RNA biomarkers. In this review article, we will summarize the development of the four generations of NanoVelcro CTC assays, and the clinical applications of each generation of devices.
Collapse
Affiliation(s)
- Yu Jen Jan
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jie-Fu Chen
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yazhen Zhu
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yi-Tsung Lu
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Szu Hao Chen
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Howard Chung
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matthew Smalley
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; CytoLumina Technologies Corp., Los Angeles, CA, USA
| | - Yen-Wen Huang
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; CytoLumina Technologies Corp., Los Angeles, CA, USA
| | - Jiantong Dong
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA
| | - Li-Ching Chen
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan
| | - Hsiao-Hua Yu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - James S Tomlinson
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA; Center for Pancreatic Disease, University of California, Los Angeles, Los Angeles, CA, USA; Department of Surgery, Greater Los Angeles Veteran's Affairs Administration, Los Angeles, CA, USA
| | - Shuang Hou
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vatche G Agopian
- Department of Surgery, University of California, Los Angeles, Los Angeles, CA, USA; Liver Transplantation and Hepatobiliary Surgery, University of California, Los Angeles, Los Angeles, CA, USA
| | - Edwin M Posadas
- Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Serra M, Ferraro D, Pereiro I, Viovy JL, Descroix S. The power of solid supports in multiphase and droplet-based microfluidics: towards clinical applications. LAB ON A CHIP 2017; 17:3979-3999. [PMID: 28948991 DOI: 10.1039/c7lc00582b] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Multiphase and droplet microfluidic systems are growing in relevance in bioanalytical-related fields, especially due to the increased sensitivity, faster reaction times and lower sample/reagent consumption of many of its derived bioassays. Often applied to homogeneous (liquid/liquid) reactions, innovative strategies for the implementation of heterogeneous (typically solid/liquid) processes have recently been proposed. These involve, for example, the extraction and purification of target analytes from complex matrices or the implementation of multi-step protocols requiring efficient washing steps. To achieve this, solid supports such as functionalized particles (micro or nanometric) presenting different physical properties (e.g. magnetic, optical or others) are used for the binding of specific entities. The manipulation of such supports with different microfluidic principles has both led to the miniaturization of existing biomedical protocols and the development of completely new strategies for diagnostics and research. In this review, multiphase and droplet-based microfluidic systems using solid suspensions are presented and discussed with a particular focus on: i) working principles and technological developments of the manipulation strategies and ii) applications, critically discussing the level of maturity of these systems, which can range from initial proofs of concept to real clinical validations.
Collapse
Affiliation(s)
- M Serra
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France.
| | | | | | | | | |
Collapse
|
24
|
Zhou Z, Chen D, Wang X, Jiang J. Milling Positive Master for Polydimethylsiloxane Microfluidic Devices: The Microfabrication and Roughness Issues. MICROMACHINES 2017; 8:E287. [PMID: 30400477 PMCID: PMC6190291 DOI: 10.3390/mi8100287] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/12/2017] [Accepted: 09/18/2017] [Indexed: 11/17/2022]
Abstract
We provide a facile and low-cost method (F-L) to fabricate a two-dimensional positive master using a milling technique for polydimethylsiloxane (PDMS)-based microchannel molding. This method comprises the following steps: (1) a positive microscale master of the geometry is milled on to an acrylic block; (2) pre-cured PDMS is used to mold the microscale positive master; (3) the PDMS plate is peeled off from the master and punctured with a blunt needle; and (4) the PDMS plate is O₂ plasma bonded to a glass slide. Using this technique, we can fabricate microchannels with very simple protocols quickly and inexpensively. This method also avoids breakage of the end mill (ϕ = 0.4 mm) of the computerized numerical control (CNC) system when fabricating the narrow channels (width < 50 µm). The prominent surface roughness of the milled bottom-layer could be overcomed by pre-cured PDMS with size trade-off in design. Finally, emulsion formation successfully demonstrates the validity of the proposed fabrication protocol. This work represents an important step toward the use of a milling technique for PDMS-based microfabrication.
Collapse
Affiliation(s)
- Zhizhi Zhou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Dong Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Xiang Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Jiahuan Jiang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College of Chongqing University, Chongqing 400044, China.
| |
Collapse
|
25
|
van der Toom EE, Verdone JE, Jun C, Petrisor D, Lim S, de la Rosette JJMCH, de Reijke TM, Gorin MA, Pienta KJ, Stoianovici D. A surface tension magnetophoretic device for rare cell isolation and characterization. Med Oncol 2017; 34:22. [PMID: 28058627 DOI: 10.1007/s12032-016-0877-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 12/27/2016] [Indexed: 12/18/2022]
Abstract
The cancer community continues to search for an efficient and cost-effective technique to isolate and characterize circulating cells (CTCs) as a 'real-time liquid biopsy'. Existing methods to isolate and analyze CTCs require various transfer, wash, and staining steps that can be time consuming, expensive, and led to the loss of rare cells. To overcome the limitations of existing CTC isolation strategies, we have developed an inexpensive 'lab on a chip' device for the enrichment, staining, and analysis of rare cell populations. This device utilizes immunomagnetic positive selection of antibody-bound cells, isolation of cells through an immiscible interface, and filtration. The isolated cells can then be stained utilizing immunofluorescence or used for other downstream detection methods. We describe the construction and initial preclinical testing of the device. Initial tests suggest that the device may be well suited for the isolation of CTCs and could allow the monitoring of cancer progression and the response to therapy over time.
Collapse
Affiliation(s)
- Emma E van der Toom
- Department of Urology, The James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA.,Department of Urology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - James E Verdone
- Department of Urology, The James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Changhan Jun
- Department of Urology, The James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Doru Petrisor
- Department of Urology, The James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Sunghwan Lim
- Department of Urology, The James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | | | - Theo M de Reijke
- Department of Urology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael A Gorin
- Department of Urology, The James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Kenneth J Pienta
- Department of Urology, The James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Dan Stoianovici
- Department of Urology, The James Buchanan Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
26
|
Abstract
Circulating tumour cells (CTCs) have been studied as biomarkers of a number of solid malignancies. Potential clinical applications for CTC analysis include early cancer detection, disease staging, monitoring for recurrence, prognostication, and to aid in the selection of therapy. In the field of urologic oncology, CTCs have been most widely studied as prognostic biomarkers of castration-resistant prostate cancer. Additionally, emerging data support a role for CTCs to help identify which patients are most likely to respond to novel androgen-pathway targeted therapies, such as abiraterone and enzalutamide. CTCs have also been studied as predictive biomarkers of bladder cancer, in particular as a means to identify patients whose disease has been clinically understaged. Less is known regarding CTCs in kidney cancer; this has been attributed to the fact that a minority of renal tumours express EpCAM, the epithelial cell surface protein commonly used by CTC assays for positive cell selection. However, alternative approaches using markers specific for kidney cancer are being explored.
Collapse
|
27
|
Guckenberger DJ, Pezzi HM, Regier MC, Berry SM, Fawcett K, Barrett K, Beebe DJ. Magnetic System for Automated Manipulation of Paramagnetic Particles. Anal Chem 2016; 88:9902-9907. [PMID: 27598856 DOI: 10.1021/acs.analchem.6b02257] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The simple, rapid magnetic manipulation of paramagnetic particles (PMPs) paired with the wide range of available surface chemistries has strongly positioned PMPs in the field of analyte isolation. One recent technology, sliding lid for immobilized droplet extractions (SLIDE), presents a simple, rapid alternative to traditional PMP isolation protocols. Rather than remove fluid from PMP-bound analyte, SLIDE directly removes the PMPs from the fluid. SLIDE collects the PMPs on a hydrophobic, removable surface, which allows PMPs to be captured from one well and then transferred and released into a second well. Despite several key advantages, SLIDE remains limited by its passive magnetic manipulation that only allows for a one-time capture-and-release of PMPs, preventing wash steps and limiting purity. Furthermore, the strategy employed by SLIDE constrains the position of the wells, thereby limiting throughput and integration into automated systems. Here, we introduce a new, mechanically and operationally simplistic magnetic manipulation system for integration with the SLIDE technology to overcome the previously stated limitations. This magnetic system is compatible with nearly any plate design, can be integrated into automated workflows, enables high-throughput formats, simplifies mechanical requirements, and is amenable to a range of analytes. Using this magnetic system, PMPs can be collected, released, and resuspended throughout multiple wells regardless of proximity. We demonstrate this system's capabilities to isolate whole cells, mRNA, and DNA, demonstrating up to a 28-fold improvement of purity via the multiwash protocols enabled by this magnetic technology.
Collapse
Affiliation(s)
- David J Guckenberger
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison , 1111 Highland Avenue, Madison, Wisconsin 53705, United States.,Salus Discovery, LLC , 110 East Main Street, Madison, Wisconsin 53703, United States
| | - Hannah M Pezzi
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison , 1111 Highland Avenue, Madison, Wisconsin 53705, United States.,Salus Discovery, LLC , 110 East Main Street, Madison, Wisconsin 53703, United States
| | - Mary C Regier
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison , 1111 Highland Avenue, Madison, Wisconsin 53705, United States.,Gilson, Inc. , 3000 Parmenter Street, Middleton, Wisconsin 53562, United States
| | - Scott M Berry
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison , 1111 Highland Avenue, Madison, Wisconsin 53705, United States.,Salus Discovery, LLC , 110 East Main Street, Madison, Wisconsin 53703, United States
| | - Kevin Fawcett
- Gilson, Inc. , 3000 Parmenter Street, Middleton, Wisconsin 53562, United States
| | - Kevin Barrett
- Gilson, Inc. , 3000 Parmenter Street, Middleton, Wisconsin 53562, United States
| | - David J Beebe
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison , 1111 Highland Avenue, Madison, Wisconsin 53705, United States.,Salus Discovery, LLC , 110 East Main Street, Madison, Wisconsin 53703, United States
| |
Collapse
|
28
|
Sperger JM, Strotman LN, Welsh A, Casavant BP, Chalmers Z, Horn S, Heninger E, Thiede SM, Tokar J, Gibbs BK, Guckenberger DJ, Carmichael L, Dehm SM, Stephens PJ, Beebe DJ, Berry SM, Lang JM. Integrated Analysis of Multiple Biomarkers from Circulating Tumor Cells Enabled by Exclusion-Based Analyte Isolation. Clin Cancer Res 2016; 23:746-756. [PMID: 27401243 DOI: 10.1158/1078-0432.ccr-16-1021] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 05/26/2016] [Accepted: 06/25/2016] [Indexed: 12/28/2022]
Abstract
PURPOSE There is a critical clinical need for new predictive and pharmacodynamic biomarkers that evaluate pathway activity in patients treated with targeted therapies. A microscale platform known as VERSA (versatile exclusion-based rare sample analysis) was developed to integrate readouts across protein, mRNA, and DNA in circulating tumor cells (CTC) for a comprehensive analysis of the androgen receptor (AR) signaling pathway. EXPERIMENTAL DESIGN Utilizing exclusion-based sample preparation principles, a handheld chip was developed to perform CTC capture, enumeration, quantification, and subcellular localization of proteins and extraction of mRNA and DNA. This technology was validated across integrated endpoints in cell lines and a cohort of patients with castrate-resistant prostate cancer (CRPC) treated with AR-targeted therapies and chemotherapies. RESULTS The VERSA was validated in cell lines to analyze AR protein expression, nuclear localization, and gene expression targets. When applied to a cohort of patients, radiographic progression was predicted by the presence of multiple AR splice variants and activity in the canonical AR signaling pathway. AR protein expression and nuclear localization identified phenotypic heterogeneity. Next-generation sequencing with the FoundationOne panel detected copy number changes and point mutations. Longitudinal analysis of CTCs identified acquisition of multiple AR variants during targeted treatments and chemotherapy. CONCLUSIONS Complex mechanisms of resistance to AR-targeted therapies, across RNA, DNA, and protein endpoints, exist in patients with CRPC and can be quantified in CTCs. Interrogation of the AR signaling pathway revealed distinct patterns relevant to tumor progression and can serve as pharmacodynamic biomarkers for targeted therapies. Clin Cancer Res; 1-11. ©2016 AACR.
Collapse
Affiliation(s)
- Jamie M Sperger
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lindsay N Strotman
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | | | - Benjamin P Casavant
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | | | - Sacha Horn
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Erika Heninger
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Stephanie M Thiede
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jacob Tokar
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Benjamin K Gibbs
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - David J Guckenberger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lakeesha Carmichael
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Scott M Dehm
- Masonic Cancer Center and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | | | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Scott M Berry
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin. .,Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
29
|
Berry SM, Pezzi HM, LaVanway AJ, Guckenberger D, Anderson M, Beebe DJ. AirJump: Using Interfaces to Instantly Perform Simultaneous Extractions. ACS APPLIED MATERIALS & INTERFACES 2016; 8:15040-5. [PMID: 27249333 PMCID: PMC5058634 DOI: 10.1021/acsami.6b02555] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Analyte isolation is an important process that spans a range of biomedical disciplines, including diagnostics, research, and forensics. While downstream analytical techniques have advanced in terms of both capability and throughput, analyte isolation technology has lagged behind, increasingly becoming the bottleneck in these processes. Thus, there exists a need for simple, fast, and easy to integrate analyte separation protocols to alleviate this bottleneck. Recently, a new class of technologies has emerged that leverages the movement of paramagnetic particle (PMP)-bound analytes through phase barriers to achieve a high efficiency separation in a single or a few steps. Specifically, the passage of a PMP/analyte aggregate through a phase interface (aqueous/air in this case) acts to efficiently "exclude" unbound (contaminant) material from PMP-bound analytes with higher efficiency than traditional washing-based solid-phase extraction (SPE) protocols (i.e., bind, wash several times, elute). Here, we describe for the first time a new type of "exclusion-based" sample preparation, which we term "AirJump". Upon realizing that much of the contaminant carryover stems from interactions with the sample vessel surface (e.g., pipetting residue, wetting), we aim to eliminate the influence of that factor. Thus, AirJump isolates PMP-bound analyte by "jumping" analyte directly out of a free liquid/air interface. Through careful characterization, we have demonstrated the validity of AirJump isolation through comparison to traditional washing-based isolations. Additionally, we have confirmed the suitability of AirJump in three important independent biological isolations, including protein immunoprecipitation, viral RNA isolation, and cell culture gene expression analysis. Taken together, these data sets demonstrate that AirJump performs efficiently, with high analyte yield, high purity, no cross contamination, rapid time-to-isolation, and excellent reproducibility.
Collapse
|
30
|
Chen JF, Zhu Y, Lu YT, Hodara E, Hou S, Agopian VG, Tomlinson JS, Posadas EM, Tseng HR. Clinical Applications of NanoVelcro Rare-Cell Assays for Detection and Characterization of Circulating Tumor Cells. Theranostics 2016; 6:1425-39. [PMID: 27375790 PMCID: PMC4924510 DOI: 10.7150/thno.15359] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/06/2016] [Indexed: 12/22/2022] Open
Abstract
Liquid biopsy of tumor through isolation of circulating tumor cells (CTCs) allows non-invasive, repetitive, and systemic sampling of disease. Although detecting and enumerating CTCs is of prognostic significance in metastatic cancer, it is conceivable that performing molecular and functional characterization on CTCs will reveal unprecedented insight into the pathogenic mechanisms driving lethal disease. Nanomaterial-embedded cancer diagnostic platforms, i.e., NanoVelcro CTC Assays represent a unique rare-cell sorting method that enables detection isolation, and characterization of CTCs in peripheral blood, providing an opportunity to noninvasively monitor disease progression in individual cancer patients. Over the past decade, a series of NanoVelcro CTC Assays has been demonstrated for exploring the full potential of CTCs as a clinical biomarker, including CTC enumeration, phenotyping, genotyping and expression profiling. In this review article, the authors will briefly introduce the development of three generations of NanoVelcro CTC Assays, and highlight the clinical applications of each generation for various types of solid cancers, including prostate cancer, pancreatic cancer, lung cancer, and melanoma.
Collapse
Affiliation(s)
- Jie-Fu Chen
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yazhen Zhu
- 2. Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California, USA;; 3. Department of Pathology, Guangdong Provincial Hospital of TCM, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi-Tsung Lu
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Elisabeth Hodara
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shuang Hou
- 3. Department of Pathology, Guangdong Provincial Hospital of TCM, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Vatche G Agopian
- 4. Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA;; 5. Liver Transplantation and Hepatobiliary Surgery, University of California, Los Angeles, Los Angeles, California, USA
| | - James S Tomlinson
- 4. Department of Surgery, University of California, Los Angeles, Los Angeles, California, USA;; 6. Center for Pancreatic Disease, University of California, Los Angeles, Los Angeles, California, USA;; 7. Department of Surgery Greater Los Angeles Veteran's Affairs Administration, Los Angeles, California, USA
| | - Edwin M Posadas
- 1. Urologic Oncology Program and Uro-Oncology Research Laboratories, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hsian-Rong Tseng
- 2. Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
31
|
Mosley O, Melling L, Tarn MD, Kemp C, Esfahani MMN, Pamme N, Shaw KJ. Sample introduction interface for on-chip nucleic acid-based analysis of Helicobacter pylori from stool samples. LAB ON A CHIP 2016; 16:2108-15. [PMID: 27164181 DOI: 10.1039/c6lc00228e] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Despite recent advances in microfluidic-based integrated diagnostic systems, the sample introduction interface, especially with regards to large volume samples, has often been neglected. We present a sample introduction interface that allows direct on-chip processing of crude stool samples for the detection of Helicobacter pylori (H. pylori). The principle of IFAST (immiscible filtration assisted by surface tension) was adapted to include a large volume sample chamber with a septum-based interface for stool sample introduction. Solid chaotropic salt and dry superparamagnetic particles (PMPs) could be stored on-chip and reconstituted upon sample addition, simplifying the process of release of DNA from H. pylori cells and its binding to the PMPs. Finally, the PMPs were pulled via a magnet through a washing chamber containing an immiscible oil solution and into an elution chamber where the DNA was released into aqueous media for subsequent analysis. The entire process required only 7 min while enabling a 40-fold reduction in working volume from crude biological samples. The combination of a real-world interface and rapid DNA extraction offers the potential for the methodology to be used in point-of-care (POC) devices.
Collapse
Affiliation(s)
- O Mosley
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK.
| | - L Melling
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK.
| | - M D Tarn
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - C Kemp
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - M M N Esfahani
- School of Engineering, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - N Pamme
- Department of Chemistry, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - K J Shaw
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK.
| |
Collapse
|
32
|
Abstract
Isolation and analysis of cancer cells from body fluids have significant implications in diagnosis and therapeutic treatment of cancers. Circulating tumor cells (CTCs) are cancer cells circulating in the peripheral blood or spreading iatrogenically into blood vessels, which is an early step in the cascade of events leading to cancer metastasis. Therefore, CTCs can be used for diagnosing for therapeutic treatment, prognosing a given anticancer intervention, and estimating the risk of metastatic relapse. However, isolation of CTCs is a significant technological challenge due to their rarity and low recovery rate using traditional purification techniques. Recently microfluidic devices represent a promising platform for isolating cancer cells with high efficiency in processing complex cellular fluids, with simplicity, sensitivity, and throughput. This review summarizes recent methods of CTC isolation and analysis, as well as their applications in clinical studies.
Collapse
Affiliation(s)
- J Zhang
- University of Florida, Gainesville, FL, United States
| | - K Chen
- University of Florida, Gainesville, FL, United States
| | - Z H Fan
- University of Florida, Gainesville, FL, United States.
| |
Collapse
|
33
|
Scherr TF, Ryskoski HB, Doyle AB, Haselton FR. A two-magnet strategy for improved mixing and capture from biofluids. BIOMICROFLUIDICS 2016; 10:024118. [PMID: 27158286 PMCID: PMC4833749 DOI: 10.1063/1.4946014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/30/2016] [Indexed: 05/25/2023]
Abstract
Magnetic beads are a popular method for concentrating biomolecules from solution and have been more recently used in multistep pre-arrayed microfluidic cartridges. Typical processing strategies rely on a single magnet, resulting in a tight cluster of beads and requiring long incubation times to achieve high capture efficiencies, especially in highly viscous patient samples. This report describes a two-magnet strategy to improve the interaction of the bead surface with the surrounding fluid inside of a pre-arrayed, self-contained assay-in-a-tube. In the two-magnet system, target biomarker capture occurs at a rate three times faster than the single-magnet system. In clinically relevant biomatrices, we find a 2.5-fold improvement in biomarker capture at lower sample viscosities with the two-magnet system. In addition, we observe a 20% increase in the amount of protein captured at high viscosity for the two-magnet configuration relative to the single magnet approach. The two-magnet approach offers a means to achieve higher biomolecule extraction yields and shorter assay times in magnetic capture assays and in self-contained processor designs.
Collapse
Affiliation(s)
- Thomas F Scherr
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37235, USA
| | - Hayley B Ryskoski
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37235, USA
| | - Andrew B Doyle
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37235, USA
| | - Frederick R Haselton
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee 37235, USA
| |
Collapse
|
34
|
|
35
|
Li J, Gregory SG, Garcia-Blanco MA, Armstrong AJ. Using circulating tumor cells to inform on prostate cancer biology and clinical utility. Crit Rev Clin Lab Sci 2015; 52:191-210. [PMID: 26079252 DOI: 10.3109/10408363.2015.1023430] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Substantial advances in the molecular biology of prostate cancer have led to the approval of multiple new systemic agents to treat men with metastatic castration-resistant prostate cancer (mCRPC). These treatments encompass androgen receptor directed therapies, immunotherapies, bone targeting radiopharmaceuticals and cytotoxic chemotherapies. There is, however, great heterogeneity in the degree of patient benefit with these agents, thus fueling the need to develop predictive biomarkers that are able to rationally guide therapy. Circulating tumor cells (CTCs) have the potential to provide an assessment of tumor-specific biomarkers through a non-invasive, repeatable "liquid biopsy" of a patient's cancer at a given point in time. CTCs have been extensively studied in men with mCRPC, where CTC enumeration using the Cellsearch® method has been validated and FDA approved to be used in conjunction with other clinical parameters as a prognostic biomarker in metastatic prostate cancer. In addition to enumeration, more sophisticated molecular profiling of CTCs is now feasible and may provide more clinical utility as it may reflect tumor evolution within an individual particularly under the pressure of systemic therapies. Here, we review technologies used to detect and characterize CTCs, and the potential biological and clinical utility of CTC molecular profiling in men with metastatic prostate cancer.
Collapse
Affiliation(s)
- Jing Li
- a Duke Cancer Institute, Duke University Medical Center , Durham , NC , USA
| | | | | | | |
Collapse
|
36
|
Guckenberger DJ, de Groot TE, Wan AMD, Beebe DJ, Young EWK. Micromilling: a method for ultra-rapid prototyping of plastic microfluidic devices. LAB ON A CHIP 2015; 15:2364-78. [PMID: 25906246 PMCID: PMC4439323 DOI: 10.1039/c5lc00234f] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
This tutorial review offers protocols, tips, insight, and considerations for practitioners interested in using micromilling to create microfluidic devices. The objective is to provide a potential user with information to guide them on whether micromilling would fill a specific need within their overall fabrication strategy. Comparisons are made between micromilling and other common fabrication methods for plastics in terms of technical capabilities and cost. The main discussion focuses on "how-to" aspects of micromilling, to enable a user to select proper equipment and tools, and obtain usable microfluidic parts with minimal start-up time and effort. The supplementary information provides more extensive discussion on CNC mill setup, alignment, and programming. We aim to reach an audience with minimal prior experience in milling, but with strong interests in fabrication of microfluidic devices.
Collapse
Affiliation(s)
- David J Guckenberger
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | |
Collapse
|
37
|
Lin M, Chen JF, Lu YT, Zhang Y, Song J, Hou S, Ke Z, Tseng HR. Nanostructure embedded microchips for detection, isolation, and characterization of circulating tumor cells. Acc Chem Res 2014; 47:2941-50. [PMID: 25111636 PMCID: PMC4204926 DOI: 10.1021/ar5001617] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Circulating
tumor cells (CTCs) are cancer cells that break away
from either a primary tumor or a metastatic site and circulate in
the peripheral blood as the cellular origin of metastasis. With their
role as a “tumor liquid biopsy”, CTCs provide convenient
access to all disease sites, including that of the primary tumor and
the site of fatal metastases. It is conceivable that detecting and
analyzing CTCs will provide insightful information in assessing the
disease status without the flaws and limitations encountered in performing
conventional tumor biopsies. However, identifying CTCs in patient
blood samples is technically challenging due to the extremely low
abundance of CTCs among a large number of hematologic cells. To address
this unmet need, there have been significant research endeavors, especially
in the fields of chemistry, materials science, and bioengineering,
devoted to developing CTC detection, isolation, and characterization
technologies. Inspired by the nanoscale interactions observed
in the tissue microenvironment,
our research team at UCLA pioneered a unique concept of “NanoVelcro”
cell-affinity substrates, in which CTC capture agent-coated nanostructured
substrates were utilized to immobilize CTCs with high efficiency.
The working mechanism of NanoVelcro cell-affinity substrates mimics
that of Velcro: when the two fabric strips of a Velcro fastener are
pressed together, tangling between the hairy surfaces on two strips
leads to strong binding. Through continuous evolution, three generations
(gens) of NanoVelcro CTC chips have been established to achieve different
clinical utilities. The first-gen NanoVelcro chip, composed of a silicon
nanowire substrate (SiNS) and an overlaid microfluidic chaotic mixer,
was created for CTC enumeration. Side-by-side analytical validation
studies using clinical blood samples suggested that the sensitivity
of first-gen NanoVelcro chip outperforms that of FDA-approved CellSearch.
In conjunction with the use of the laser microdissection (LMD) technique,
second-gen NanoVelcro chips (i.e., NanoVelcro-LMD), based on polymer
nanosubstrates, were developed for single-CTC isolation. The individually
isolated CTCs can be subjected to single-CTC genotyping (e.g., Sanger
sequencing and next-generation sequencing, NGS) to verify the CTC’s
role as tumor liquid biopsy. Created by grafting of thermoresponsive
polymer brushes onto SiNS, third-gen NanoVelcro chips (i.e., Thermoresponsive
NanoVelcro) have demonstrated the capture and release of CTCs at 37
and 4 °C, respectively. The temperature-dependent conformational
changes of polymer brushes can effectively alter the accessibility
of the capture agent on SiNS, allowing for rapid CTC purification
with desired viability and molecular integrity. This Account
summarizes the continuous evolution of NanoVelcro
CTC assays from the emergence of the original idea all the way to
their applications in cancer research. We envision that NanoVelcro
CTC assays will lead the way for powerful and cost-efficient diagnostic
platforms for researchers to better understand underlying disease
mechanisms and for physicians to monitor real-time disease progression.
Collapse
Affiliation(s)
- Millicent Lin
- Department
of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080 Guangdong, People’s Republic of China
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Jie-Fu Chen
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Yi-Tsung Lu
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Yang Zhang
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Jinzhao Song
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Shuang Hou
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| | - Zunfu Ke
- Department
of Pathology, The First Affiliated hospital of Sun Yat-sen University, Guangzhou, 510080 Guangdong, People’s Republic of China
| | - Hsian-Rong Tseng
- Department
of Molecular and Medical Pharmacology, Crump Institute for Molecular
Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095-1770, United States
| |
Collapse
|
38
|
Dong C, Wang H, Zhang Z, Zhang T, Liu B. Carboxybetaine methacrylate oligomer modified nylon for circulating tumor cells capture. J Colloid Interface Sci 2014; 432:135-43. [DOI: 10.1016/j.jcis.2014.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/30/2014] [Accepted: 07/01/2014] [Indexed: 02/04/2023]
|
39
|
Phurimsak C, Yildirim E, Tarn MD, Trietsch SJ, Hankemeier T, Pamme N, Vulto P. Phaseguide assisted liquid lamination for magnetic particle-based assays. LAB ON A CHIP 2014; 14:2334-2343. [PMID: 24832933 DOI: 10.1039/c4lc00139g] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
We have developed a magnetic particle-based assay platform in which functionalised magnetic particles are transferred sequentially through laminated volumes of reagents and washing buffers. Lamination of aqueous liquids is achieved via the use of phaseguide technology; microstructures that control the advancing air-liquid interface of solutions as they enter a microfluidic chamber. This allows manual filling of the device, eliminating the need for external pumping systems, and preparation of the system requires only a few minutes. Here, we apply the platform to two on-chip strategies: (i) a one-step streptavidin-biotin binding assay, and (ii) a two-step C-reactive protein immunoassay. With these, we demonstrate how condensing multiple reaction and washing processes into a single step significantly reduces procedural times, with both assay procedures requiring less than 8 seconds.
Collapse
Affiliation(s)
- Chayakom Phurimsak
- Department of Chemistry, The University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | | | | | | | | | | | | |
Collapse
|
40
|
Casavant B, Guckenberger DJ, Beebe DJ, Berry SM. Efficient sample preparation from complex biological samples using a sliding lid for immobilized droplet extractions. Anal Chem 2014; 86:6355-62. [PMID: 24927449 PMCID: PMC4079323 DOI: 10.1021/ac500574t] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 05/20/2014] [Indexed: 01/25/2023]
Abstract
Sample preparation is a major bottleneck in many biological processes. Paramagnetic particles (PMPs) are a ubiquitous method for isolating analytes of interest from biological samples and are used for their ability to thoroughly sample a solution and be easily collected with a magnet. There are three main methods by which PMPs are used for sample preparation: (1) removal of fluid from the analyte-bound PMPs, (2) removal of analyte-bound PMPs from the solution, and (3) removal of the substrate (with immobilized analyte-bound PMPs). In this paper, we explore the third and least studied method for PMP-based sample preparation using a platform termed Sliding Lid for Immobilized Droplet Extractions (SLIDE). SLIDE leverages principles of surface tension and patterned hydrophobicity to create a simple-to-operate platform for sample isolation (cells, DNA, RNA, protein) and preparation (cell staining) without the need for time-intensive wash steps, use of immiscible fluids, or precise pinning geometries. Compared to other standard isolation protocols using PMPs, SLIDE is able to perform rapid sample preparation with low (0.6%) carryover of contaminants from the original sample. The natural recirculation occurring within the pinned droplets of SLIDE make possible the performance of multistep cell staining protocols within the SLIDE by simply resting the lid over the various sample droplets. SLIDE demonstrates a simple easy to use platform for sample preparation on a range of complex biological samples.
Collapse
Affiliation(s)
| | | | - David J. Beebe
- Department of Biomedical
Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Scott M. Berry
- Department of Biomedical
Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin, Madison, Wisconsin 53705, United States
| |
Collapse
|
41
|
van Reenen A, de Jong AM, den Toonder JMJ, Prins MWJ. Integrated lab-on-chip biosensing systems based on magnetic particle actuation--a comprehensive review. LAB ON A CHIP 2014; 14:1966-86. [PMID: 24806093 DOI: 10.1039/c3lc51454d] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The demand for easy to use and cost effective medical technologies inspires scientists to develop innovative lab-on-chip technologies for point-of-care in vitro diagnostic testing. To fulfill medical needs, the tests should be rapid, sensitive, quantitative, and miniaturizable, and need to integrate all steps from sample-in to result-out. Here, we review the use of magnetic particles actuated by magnetic fields to perform the different process steps that are required for integrated lab-on-chip diagnostic assays. We discuss the use of magnetic particles to mix fluids, to capture specific analytes, to concentrate analytes, to transfer analytes from one solution to another, to label analytes, to perform stringency and washing steps, and to probe biophysical properties of the analytes, distinguishing methodologies with fluid flow and without fluid flow (stationary microfluidics). Our review focuses on efforts to combine and integrate different magnetically actuated assay steps, with the vision that it will become possible in the future to realize integrated lab-on-chip biosensing assays in which all assay process steps are controlled and optimized by magnetic forces.
Collapse
Affiliation(s)
- Alexander van Reenen
- Department of Applied Physics, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | | | | | | |
Collapse
|
42
|
Yu ZTF, Yong KMA, Fu J. Microfluidic blood cell sorting: now and beyond. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:1687-703. [PMID: 24515899 PMCID: PMC4013196 DOI: 10.1002/smll.201302907] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 12/16/2013] [Indexed: 05/15/2023]
Abstract
Blood plays an important role in homeostatic regulation with each of its cellular components having important therapeutic and diagnostic uses. Therefore, separation and sorting of blood cells hasa been of a great interest to clinicians and researchers. However, while conventional methods of processing blood have been successful in generating relatively pure fractions, they are time consuming, labor intensive, and are not optimal for processing small volume blood samples. In recent years, microfluidics has garnered great interest from clinicians and researchers as a powerful technology for separating blood into different cell fractions. As microfluidics involves fluid manipulation at the microscale level, it has the potential for achieving high-resolution separation and sorting of blood cells down to a single-cell level, with an added benefit of integrating physical and biological methods for blood cell separation and analysis on the same single chip platform. This paper will first review the conventional methods of processing and sorting blood cells, followed by a discussion on how microfluidics is emerging as an efficient tool to rapidly change the field of blood cell sorting for blood-based therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Zeta Tak For Yu
- Integrated Biosystems and Biomechanics Laboratory, University of
Michigan, Ann Arbor, Michigan, USA
- Department of Mechanical Engineering, University of Michigan, Ann
Arbor, Michigan, USA
| | - Koh Meng Aw Yong
- Integrated Biosystems and Biomechanics Laboratory, University of
Michigan, Ann Arbor, Michigan, USA
- Department of Mechanical Engineering, University of Michigan, Ann
Arbor, Michigan, USA
| | - Jianping Fu
- Integrated Biosystems and Biomechanics Laboratory, University of
Michigan, Ann Arbor, Michigan, USA
- Department of Mechanical Engineering, University of Michigan, Ann
Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann
Arbor, Michigan, USA
| |
Collapse
|
43
|
Abstract
The availability of new therapeutic options for the treatment of metastatic castration-resistant prostate cancer (mCRPC) has heightened the importance of monitoring and assessing treatment response. Accordingly, there is an unmet clinical need for reliable biomarkers that can be used to guide therapy. Circulating tumour cells (CTCs) are rare cells that are shed from primary and metastatic tumour deposits into the peripheral circulation, and represent a means of performing noninvasive tumour sampling. Indeed, enumeration of CTCs before and after therapy has shown that CTC burden correlates with prognosis in patients with mCRPC. Moreover, studies have demonstrated the potential of molecular analysis of CTCs in monitoring and predicting response to therapy in patients. This Review describes the challenges associated with monitoring treatment response in mCRPC, and the advancements in CTC-analysis technologies applied to such assessments and, ultimately, guiding prostate cancer treatment.
Collapse
|
44
|
Krejcova L, Nejdl L, Rodrigo MAM, Zurek M, Matousek M, Hynek D, Zitka O, Kopel P, Adam V, Kizek R. 3D printed chip for electrochemical detection of influenza virus labeled with CdS quantum dots. Biosens Bioelectron 2014; 54:421-7. [PMID: 24296063 DOI: 10.1016/j.bios.2013.10.031] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 10/10/2013] [Accepted: 10/21/2013] [Indexed: 12/12/2022]
Abstract
In this study, we report a new three-dimensional (3D), bead-based microfluidic chip developed for rapid, sensitive and specific detection of influenza hemagglutinin. The principle of microfluidic chip is based on implementation of two-step procedure that includes isolation based on paramagnetic beads and electrochemical detection. As a platform for isolation process, streptavidin-modified MPs, which were conjugated via biotinylated glycan (through streptavidin-biotin affinity) followed by linkage of hemagglutinin to glycan, were used. Vaccine hemagglutinin (HA vaxi) was labeled with CdS quantum dots (QDs) at first. Detection of the isolation product by voltammetry was the end point of the procedure. The suggested and developed method can be used also for detection of other specific substances that are important for control, diagnosis or therapy of infectious diseases.
Collapse
Affiliation(s)
- Ludmila Krejcova
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic
| | - Lukas Nejdl
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic
| | - Miguel Angel Merlos Rodrigo
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, 61600 Brno, Czech Republic
| | - Michal Zurek
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic
| | - Miroslav Matousek
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic
| | - David Hynek
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, 61600 Brno, Czech Republic
| | - Ondrej Zitka
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, 61600 Brno, Czech Republic
| | - Pavel Kopel
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, 61600 Brno, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, 61600 Brno, Czech Republic
| | - Rene Kizek
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic; Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, 61600 Brno, Czech Republic.
| |
Collapse
|
45
|
Dudani JS, Go DE, Gossett DR, Tan AP, Di Carlo D. Mediating Millisecond Reaction Time around Particles and Cells. Anal Chem 2014; 86:1502-10. [DOI: 10.1021/ac402920m] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Jaideep S. Dudani
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
| | - Derek E. Go
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
| | - Daniel R. Gossett
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
- California NanoSystems Institute, Los Angeles, California 90095, United States
| | - Andrew P. Tan
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
| | - Dino Di Carlo
- Department
of Bioengineering, University of California Los Angeles, 420 Westwood Plaza, 5121
Engineering V, Box 951600, Los Angeles, California 90095, United States
- California NanoSystems Institute, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, Los
Angeles, California 90095, United States
| |
Collapse
|
46
|
Guckenberger DJ, Thomas PC, Rothbauer J, LaVanway AJ, Anderson M, Gilson D, Fawcett K, Berto T, Barrett K, Beebe DJ, Berry SM. A Combined Fabrication and Instrumentation Platform for Sample Preparation. ACTA ACUST UNITED AC 2014; 19:267-74. [PMID: 24401822 DOI: 10.1177/2211068213518312] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Indexed: 11/17/2022]
Abstract
While potentially powerful, access to molecular diagnostics is substantially limited in the developing world. Here we present an approach to reduced cost molecular diagnostic instrumentation that has the potential to empower developing world communities by reducing costs through streamlining the sample preparation process. In addition, this instrument is capable of producing its own consumable devices on demand, reducing reliance on assay suppliers. Furthermore, this instrument is designed with an "open" architecture, allowing users to visually observe the assay process and make modifications as necessary (as opposed to traditional "black box" systems). This open environment enables integration of microfluidic fabrication and viral RNA purification onto an easy-to-use modular system via the use of interchangeable trays. Here we employ this system to develop a protocol to fabricate microfluidic devices and then use these devices to isolate viral RNA from serum for the measurement of human immunodeficiency virus (HIV) viral load. Results obtained from this method show significantly reduced error compared with similar nonautomated sample preparation processes.
Collapse
Affiliation(s)
- David J Guckenberger
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Peter C Thomas
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Jacob Rothbauer
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Alex J LaVanway
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Meghan Anderson
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | | - David J Beebe
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Scott M Berry
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
47
|
King JD, Casavant BP, Lang JM. Rapid translation of circulating tumor cell biomarkers into clinical practice: technology development, clinical needs and regulatory requirements. LAB ON A CHIP 2014; 14:24-31. [PMID: 24190548 DOI: 10.1039/c3lc50741f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The great hope in circulating tumor cell (CTC) research lies in the use of these rare cells as an accessible "fluid biopsy" that would permit frequent, minimally invasive sampling of tumor cells for similar molecular assays that are performed on traditional biopsies. Given the rarity of CTCs in peripheral circulation, microscale methods show great promise and superiority to capture and analyze these cells from patients with solid tumors. Novel technologies that produce validated CTC biomarkers may finally provide medical oncologists the tools needed to provide precise, personalized medical care for patients with advanced cancer. However, few CTC technologies demonstrate both experimental and clinical evidence of an accurate, reliable and reproducible assay that also meets the regulatory requirements to enter routine clinical practice. Many opportunities exist to incorporate clinical needs and regulatory benchmarks into technology development to more quickly garner FDA approval to direct decisions on patient care. This review will address: 1) device development tailored to address predictive, prognostic and/or therapeutic needs across the multitude of malignancies and disease stages; 2) validation benchmarks for clinical assay development; 3) early establishment of standard operating procedures for sample acquisition and analysis; 4) demonstration of clinical utility; 5) clinical qualification of a novel biomarker; and 6) integration of a newly validated and qualified technology into routine clinical practice. Early understanding and incorporation of these regulatory requirements into assay development can simplify and speed the integration of these novel technologies into patient care. Meeting these benchmarks will lead to the true personalization of cancer therapies, directing initial and subsequent treatments for each individual based on initial tumor characteristics while monitoring for emerging mechanisms of resistance in these continually evolving tumors.
Collapse
Affiliation(s)
- Jonathan D King
- Department of Medicine, Wisconsin Institutes for Medical Research, University of Wisconsin, 1111 Highland Ave., Madison, WI 53705, USA.
| | | | | |
Collapse
|
48
|
Berry SM, Chin EN, Jackson SS, Strotman LN, Goel M, Thompson NE, Alexander CM, Miyamoto S, Burgess RR, Beebe DJ. Weak protein-protein interactions revealed by immiscible filtration assisted by surface tension. Anal Biochem 2013; 447:133-40. [PMID: 24215910 DOI: 10.1016/j.ab.2013.10.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/24/2013] [Accepted: 10/30/2013] [Indexed: 11/25/2022]
Abstract
Biological mechanisms are often mediated by transient interactions between multiple proteins. The isolation of intact protein complexes is essential to understanding biochemical processes and an important prerequisite for identifying new drug targets and biomarkers. However, low-affinity interactions are often difficult to detect. Here, we use a newly described method called immiscible filtration assisted by surface tension (IFAST) to isolate proteins under defined binding conditions. This method, which gives a near-instantaneous isolation, enables significantly higher recovery of transient complexes compared to current wash-based protocols, which require reequilibration at each of several wash steps, resulting in protein loss. The method moves proteins, or protein complexes, captured on a solid phase through one or more immiscible-phase barriers that efficiently exclude the passage of nonspecific material in a single operation. We use a previously described polyol-responsive monoclonal antibody to investigate the potential of this new method to study protein binding. In addition, difficult-to-isolate complexes involving the biologically and clinically important Wnt signaling pathway were isolated. We anticipate that this simple, rapid method to isolate intact, transient complexes will enable the discoveries of new signaling pathways, biomarkers, and drug targets.
Collapse
Affiliation(s)
- Scott M Berry
- Department of Biomedical Engineering, University of Wisconsin at Madison, Madison, WI 53705, USA.
| | - Emily N Chin
- Department of Oncology, University of Wisconsin at Madison, Madison, WI 53705, USA
| | - Shawn S Jackson
- Department of Oncology, University of Wisconsin at Madison, Madison, WI 53705, USA
| | - Lindsay N Strotman
- Department of Biomedical Engineering, University of Wisconsin at Madison, Madison, WI 53705, USA
| | - Mohit Goel
- Department of Biotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Nancy E Thompson
- Department of Oncology, University of Wisconsin at Madison, Madison, WI 53705, USA
| | - Caroline M Alexander
- Department of Oncology, University of Wisconsin at Madison, Madison, WI 53705, USA
| | - Shigeki Miyamoto
- Department of Oncology, University of Wisconsin at Madison, Madison, WI 53705, USA
| | - Richard R Burgess
- Department of Oncology, University of Wisconsin at Madison, Madison, WI 53705, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin at Madison, Madison, WI 53705, USA
| |
Collapse
|
49
|
Strotman L, O'Connell R, Casavant BP, Berry SM, Sperger JM, Lang JM, Beebe DJ. Selective nucleic acid removal via exclusion (SNARE): capturing mRNA and DNA from a single sample. Anal Chem 2013; 85:9764-70. [PMID: 24016179 PMCID: PMC3897163 DOI: 10.1021/ac402162r] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The path from gene (DNA) to gene product (RNA or protein) is the foundation of genotype giving rise to phenotype. Comparison of genomic analyses (DNA) with paired transcriptomic studies (mRNA) is critical to evaluating the pathogenic processes that give rise to human disease. The ability to analyze both DNA and mRNA from the same sample is not only important for biologic interrogation but also to minimize variance (e.g., sample loss) unrelated to the biology. Existing methods for RNA and DNA purification from a single sample are typically time-consuming and labor intensive or require large sample sizes to split for separate RNA and DNA extraction procedures. Thus, there is a need for more efficient and cost-effective methods to purify both RNA and DNA from a single sample. To address this need, we have developed a technique, termed SNARE (Selective Nucleic Acid Removal via Exclusion), that uses pinned oil interfaces to simultaneous purify mRNA and DNA from a single sample. A unique advantage of SNARE is the elimination of dilutive wash and centrifugation processes that are fundamental to conventional methods where sample is typically discarded. This minimizes loss and maximizes recovery by allowing nondilutive reinterrogation of the sample. We demonstrate that SNARE is more sensitive than commercially available kits, robustly and repeatably achieving mRNA and DNA purification from extremely low numbers of cells for downstream analyses. In addition to sensitivity, SNARE is fast, easy to use, and cost-effective and requires no laboratory infrastructure or hazardous chemicals. We demonstrate the clinical utility of the SNARE with prostate cancer circulating tumor cells to demonstrate its ability to perform both genomic and transcriptomic interrogation on rare cell populations that would be difficult to achieve with any current method.
Collapse
Affiliation(s)
- Lindsay Strotman
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison , 1111 Highland Avenue, Madison, Wisconsin53706, United States
| | | | | | | | | | | | | |
Collapse
|
50
|
Wang L, Asghar W, Demirci U, Wan Y. Nanostructured substrates for isolation of circulating tumor cells. NANO TODAY 2013; 8:347-387. [PMID: 24944563 PMCID: PMC4059613 DOI: 10.1016/j.nantod.2013.07.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Circulating tumor cells (CTCs) originate from the primary tumor mass and enter into the peripheral bloodstream. CTCs hold the key to understanding the biology of metastasis and also play a vital role in cancer diagnosis, prognosis, disease monitoring, and personalized therapy. However, CTCs are rare in blood and hard to isolate. Additionally, the viability of CTCs can easily be compromised under high shear stress while releasing them from a surface. The heterogeneity of CTCs in biomarker expression makes their isolation quite challenging; the isolation efficiency and specificity of current approaches need to be improved. Nanostructured substrates have emerged as a promising biosensing platform since they provide better isolation sensitivity at the cost of specificity for CTC isolation. This review discusses major challenges faced by CTC isolation techniques and focuses on nanostructured substrates as a platform for CTC isolation.
Collapse
Affiliation(s)
- Lixue Wang
- Department of Oncology, The Second Affiliated Hospital of Southeast University, Southeast University, Nanjing, Jiangsu 210003, PR China
| | - Waseem Asghar
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratories, Center for Biomedical Engineering, Renal Division and Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratories, Center for Biomedical Engineering, Renal Division and Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-Massachusetts Institute of Technology (MIT), Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Yuan Wan
- Department of Oncology, The Second Affiliated Hospital of Southeast University, Southeast University, Nanjing, Jiangsu 210003, PR China
- Ian Wark Research Institute, University of South Australia, Mawson Lakes, Adelaide, SA 5095, Australia
| |
Collapse
|