1
|
Witek MA, Larkey NE, Bartakova A, Hupert ML, Mog S, Cronin JK, Vun J, August KJ, Soper SA. Microfluidic Affinity Selection of B-Lineage Cells from Peripheral Blood for Minimal Residual Disease Monitoring in Pediatric B-Type Acute Lymphoblastic Leukemia Patients. Int J Mol Sci 2024; 25:10619. [PMID: 39408948 PMCID: PMC11477226 DOI: 10.3390/ijms251910619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Assessment of minimal residual disease (MRD) is the most powerful predictor of outcome in B-type acute lymphoblastic leukemia (B-ALL). MRD, defined as the presence of leukemic cells in the blood or bone marrow, is used for the evaluation of therapy efficacy. We report on a microfluidic-based MRD (MF-MRD) assay that allows for frequent evaluation of blood for the presence of circulating leukemia cells (CLCs). The microfluidic chip affinity selects B-lineage cells, including CLCs using anti-CD19 antibodies poised on the wall of the microfluidic chip. Affinity-selected cells are released from the capture surface and can be subjected to immunophenotyping to enumerate the CLCs, perform fluorescence in situ hybridization (FISH), and/or molecular analysis of the CLCs' mRNA/gDNA. During longitudinal testing of 20 patients throughout induction and consolidation therapy, the MF-MRD performed 116 tests, while only 41 were completed with multiparameter flow cytometry (MFC-MRD) using a bone marrow aspirate, as standard-of-care. Overall, 57% MF-MRD tests were MRD(+) as defined by CLC numbers exceeding a threshold of 5 × 10-4%, which was determined to be the limit of quantitation. Above a threshold of 0.01%, MFC-MRD was positive in 34% of patients. The MF offered the advantage of the opportunity for efficiently processing small volumes of blood (2 mL), which is important in the care of pediatric patients, especially infants. The minimally invasive means of blood collection are of high value when treating patients whose MRD is typically tested using an invasive bone marrow biopsy. MF-MRD detection can be useful for stratification of patients into risk groups and monitoring of patient well-being after completion of treatment for early recognition of potential impending disease recurrence.
Collapse
Affiliation(s)
- Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA;
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
| | - Nicholas E. Larkey
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Alena Bartakova
- Biofluidica Inc., San Diego, CA 92121, USA; (A.B.); (M.L.H.)
| | | | - Shalee Mog
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
| | - Jami K. Cronin
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Judy Vun
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Keith J. August
- Division of Hematology/Oncology/Bone Marrow Transplant, Children’s Mercy Kansas City, Kansas City, MO 64108, USA; (J.K.C.); (J.V.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA;
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (N.E.L.); (S.M.)
- Department of Cancer Biology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
- Biofluidica Inc., San Diego, CA 92121, USA; (A.B.); (M.L.H.)
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
2
|
Childers K, Freed IM, Hupert ML, Shaw B, Larsen N, Herring P, Norton JH, Shiri F, Vun J, August KJ, Witek MA, Soper SA. Novel thermoplastic microvalves based on an elastomeric cyclic olefin copolymer. LAB ON A CHIP 2024; 24:4422-4439. [PMID: 39171671 PMCID: PMC11339931 DOI: 10.1039/d4lc00501e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Microfluidic systems combine multiple processing steps and components to perform complex assays in an autonomous fashion. To enable the integration of several bio-analytical processing steps into a single system, valving is used as a component that directs fluids and controls introduction of sample and reagents. While elastomer polydimethylsiloxane has been the material of choice for valving, it does not scale well to accommodate disposable integrated systems where inexpensive and fast production is needed. As an alternative to polydimethylsiloxane, we introduce a membrane made of thermoplastic elastomeric cyclic olefin copolymer (eCOC), that displays unique attributes for the fabrication of reliable valving. The eCOC membrane can be extruded or injection molded to allow for high scale production of inexpensive valves. Normally hydrophobic, eCOC can be activated with UV/ozone to produce a stable hydrophilic monolayer. Valves are assembled following in situ UV/ozone activation of eCOC membrane and thermoplastic valve seat and bonded by lamination at room temperature. eCOC formed strong bonding with polycarbonate (PC) and polyethylene terephthalate glycol (PETG) able to hold high fluidic pressures of 75 kPa and 350 kPa, respectively. We characterized the eCOC valves with mechanical and pneumatic actuation and found the valves could be reproducibly actuated >50 times without failure. Finally, an integrated system with eCOC valves was employed to detect minimal residual disease (MRD) from a blood sample of a pediatric acute lymphoblastic leukemia (ALL) patient. The two module integrated system evaluated MRD by affinity-selecting CD19(+) cells and enumerating leukemia cells via immunophenotyping with ALL-specific markers.
Collapse
Affiliation(s)
- Katie Childers
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA.
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
| | - Ian M Freed
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | | | - Benjamin Shaw
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
- Department of Chemical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| | - Noah Larsen
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
- Department of Engineering Physics, The University of Kansas, Lawrence, KS 66045, USA
| | - Paul Herring
- Department of Plastics Engineering Technology, Pittsburg State University, Pittsburg, KS 66762, USA
| | - Jeanne H Norton
- Department of Plastics Engineering Technology, Pittsburg State University, Pittsburg, KS 66762, USA
| | - Farhad Shiri
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Judy Vun
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Keith J August
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Małgorzata A Witek
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
| | - Steven A Soper
- Bioengineering Program, The University of Kansas, Lawrence, KS 66045, USA.
- Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66045, USA
- Department of Chemistry, The University of Kansas, Lawrence, KS 66045, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
- KU Cancer Center, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
3
|
Gao S, Li X, Hu Z, Wang Z, Hao X. Dual targeting negative enrichment strategy for highly sensitive and purity detection of CTCs. Front Chem 2024; 12:1400988. [PMID: 38831912 PMCID: PMC11144890 DOI: 10.3389/fchem.2024.1400988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/15/2024] [Indexed: 06/05/2024] Open
Abstract
Circulating tumor cells (CTCs) have significant clinical value in early tumor detection, dynamic monitoring and immunotherapy. CTC detection stands out as a leading non-invasive approach for tumor diagnostics and therapeutics. However, the high heterogeneity of CTCs and the occurrence of epithelial-mesenchymal transition (EMT) during metastasis pose challenges to methods relying on EpCAM-positive enrichment. To address these limitations, a method based on negative enrichment of CTCs using specific leukocyte targets has been developed. In this study, aiming to overcome the low purity associated with immunomagnetic beads targeting solely the leukocyte common antigen CD45, we introduced CD66b-modified immunomagnetic beads. CD66b, a specific target for neutrophils with abundant residues, was chosen as a complementary approach. The process involved initial collection of nucleated cells from whole blood samples using density gradient centrifugation. Subsequently, magnetically labeled leukocytes were removed by magnetic field, enabling the capture of CTCs with higher sensitivity and purity while retaining their activity. Finally, we selected 20 clinical blood samples from patients with various cancers to validate the effectiveness of this strategy, providing a new generalized tool for the clinical detection of CTCs.
Collapse
Affiliation(s)
- Siying Gao
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Xuejie Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Zhiyuan Hu
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- School of Nanoscience and Technology, SinoDanish College, University of Chinese Academy of Sciences, Beijing, China
| | - Zihua Wang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xiaopeng Hao
- Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
4
|
Deng Q, Jiang B, Yan H, Wu J, Cao Z. Circulating tumor cells in gastric cancer: developments and clinical applications. Clin Exp Med 2023; 23:4385-4399. [PMID: 37548815 DOI: 10.1007/s10238-023-01158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023]
Abstract
Circulating tumor cells (CTCs), which are shed from primary tumor or metastatic sites into the bloodstream and subsequently seed into distant tissues, are considered as the precursors of metastases. Gastric cancer (GC) is a highly heterogeneous malignant tumor. With regard to the diagnosis of GC, secondary pathological biopsy is difficult, while invasive examination is harmful to patients. In recent years, CTCs have made great progress in tumor diagnosis, prognosis prediction, efficacy detection and treatment guidance, but the research on the role of CTCs in GC remains limited. The following sections review the landmark studies demonstrating the technical approaches of CTCs monitoring in the field of GC. Moreover, we highlight the clinical application of CTCs numbers and phenotypes in monitoring the therapeutic efficacy and judging patient prognosis by sequential blood analyses.
Collapse
Affiliation(s)
- Qian Deng
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Bo Jiang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Haijiao Yan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
| | - Jun Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Zhenzhen Cao
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| |
Collapse
|
5
|
Yeh PY, Chen JY, Shen MY, Che TF, Lim SC, Wang J, Tsai WS, Frank CW, Huang CJ, Chang YC. Liposome-tethered supported lipid bilayer platform for capture and release of heterogeneous populations of circulating tumor cells. J Mater Chem B 2023; 11:8159-8169. [PMID: 37313622 DOI: 10.1039/d3tb00547j] [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: 06/15/2023]
Abstract
Because of scarcity, vulnerability, and heterogeneity in the population of circulating tumor cells (CTCs), the CTC isolation system relying on immunoaffinity interaction exhibits inconsistent efficiencies for all types of cancers and even CTCs with different phenotypes in individuals. Moreover, releasing viable CTCs from an isolation system is of importance for molecular analysis and drug screening in precision medicine, which remains a challenge for current systems. In this work, a new CTC isolation microfluidic platform was developed and contains a coating of the antibody-conjugated liposome-tethered-supported lipid bilayer in a developed chaotic-mixing microfluidic system, referred to as the "LIPO-SLB" platform. The biocompatible, soft, laterally fluidic, and antifouling properties of the LIPO-SLB platform offer high CTC capture efficiency, viability, and selectivity. We successfully demonstrated the capability of the LIPO-SLB platform to recapitulate different cancer cell lines with different antigen expression levels. In addition, the captured CTCs in the LIPO-SLB platform can be detached by air foam to destabilize the physically assembled bilayer structures due to a large water/air interfacial area and strong surface tension. More importantly, the LIPO-SLB platform was constructed and used for the verification of clinical samples from 161 patients with different primary cancer types. The mean values of both single CTCs and CTC clusters correlated well with the cancer stages. Moreover, a considerable number of CTCs were isolated from patients' blood samples in the early/localized stages. The clinical validation demonstrated the enormous potential of the universal LIPO-SLB platform as a tool for prognostic and predictive purposes in precision medicine.
Collapse
Affiliation(s)
- Po-Ying Yeh
- Genomics Research Center, Academia Sinica, 128, Sec 2, Academic Rd., Nankang, Taipei 115, Taiwan.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Jia-Yang Chen
- Genomics Research Center, Academia Sinica, 128, Sec 2, Academic Rd., Nankang, Taipei 115, Taiwan.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Mo-Yuan Shen
- Genomics Research Center, Academia Sinica, 128, Sec 2, Academic Rd., Nankang, Taipei 115, Taiwan.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Ting-Fang Che
- Genomics Research Center, Academia Sinica, 128, Sec 2, Academic Rd., Nankang, Taipei 115, Taiwan.
| | - Syer Choon Lim
- Genomics Research Center, Academia Sinica, 128, Sec 2, Academic Rd., Nankang, Taipei 115, Taiwan.
| | - Jocelyn Wang
- The College, The University of Chicago, Chicago, IL 60637, USA
| | - Wen-Sy Tsai
- Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Science, Chang Gung University, Linkou, Taoyuan, Taiwan
| | - Curtis W Frank
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Chun-Jen Huang
- Department of Chemical & Materials Engineering, and NCU-Covestro Research Center, National Central University, Jhong-Li, Taoyuan 320, Taiwan.
- R&D Center for Membrane Technology, Chung Yuan Christian University, 200 Chung Pei Rd., Chung-Li City 32023, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, 128, Sec 2, Academic Rd., Nankang, Taipei 115, Taiwan.
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
6
|
Macaraniag C, Luan Q, Zhou J, Papautsky I. Microfluidic techniques for isolation, formation, and characterization of circulating tumor cells and clusters. APL Bioeng 2022; 6:031501. [PMID: 35856010 PMCID: PMC9288269 DOI: 10.1063/5.0093806] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cell (CTC) clusters that are shed from the primary tumor into the bloodstream are associated with a poor prognosis, elevated metastatic potential, higher proliferation rate, and distinct molecular features compared to single CTCs. Studying CTC clusters may give us information on the differences in the genetic profiles, somatic mutations, and epigenetic changes in circulating cells compared to the primary tumor and metastatic sites. Microfluidic systems offer the means of studying CTC clusters through the ability to efficiently isolate these rare cells from the whole blood of patients in a liquid biopsy. Microfluidics can also be used to develop in vitro models of CTC clusters and make possible their characterization and analysis. Ultimately, microfluidic systems can offer the means to gather insight on the complexities of the metastatic process, the biology of cancer, and the potential for developing novel or personalized therapies. In this review, we aim to discuss the advantages and challenges of the existing microfluidic systems for working with CTC clusters. We hope that an improved understanding of the role microfluidics can play in isolation, formation, and characterization of CTC clusters, which can lead to increased sophistication of microfluidic platforms in cancer research.
Collapse
Affiliation(s)
- Celine Macaraniag
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Qiyue Luan
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Jian Zhou
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Ian Papautsky
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
7
|
Chelakkot C, Yang H, Shin YK. Relevance of Circulating Tumor Cells as Predictive Markers for Cancer Incidence and Relapse. Pharmaceuticals (Basel) 2022; 15:75. [PMID: 35056131 PMCID: PMC8781286 DOI: 10.3390/ph15010075] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023] Open
Abstract
Shedding of cancer cells from the primary site or undetectable bone marrow region into the circulatory system, resulting in clinically overt metastasis or dissemination, is the hallmark of unfavorable invasive cancers. The shed cells remain in circulation until they extravasate to form a secondary metastatic lesion or undergo anoikis. The circulating tumor cells (CTCs) found as single cells or clusters carry a plethora of information, are acknowledged as potential biomarkers for predicting cancer prognosis and cancer progression, and are supposed to play key roles in determining tailored therapies for advanced diseases. With the advent of novel technologies that allow the precise isolation of CTCs, more and more clinical trials are focusing on the prognostic and predictive potential of CTCs. In this review, we summarize the role of CTCs as a predictive marker for cancer incidence, relapse, and response to therapy.
Collapse
Affiliation(s)
- Chaithanya Chelakkot
- Bio-MAX/N-Bio, Bio-MAX Institute, Seoul National University, Seoul 08226, Korea
- Genobio Corp., Seoul 08394, Korea
| | - Hobin Yang
- Research Institute of Pharmaceutical Science, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08226, Korea
| | - Young Kee Shin
- Bio-MAX/N-Bio, Bio-MAX Institute, Seoul National University, Seoul 08226, Korea
- Research Institute of Pharmaceutical Science, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 08226, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08226, Korea
| |
Collapse
|
8
|
Negative enrichment of circulating tumor cells from unmanipulated whole blood with a 3D printed device. Sci Rep 2021; 11:20583. [PMID: 34663896 PMCID: PMC8523721 DOI: 10.1038/s41598-021-99951-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/23/2021] [Indexed: 01/07/2023] Open
Abstract
Reliable and routine isolation of circulating tumor cells (CTCs) from peripheral blood would allow effective monitoring of the disease and guide the development of personalized treatments. Negative enrichment of CTCs by depleting normal blood cells ensures against a biased selection of a subpopulation and allows the assay to be applied on different tumor types. Here, we report an additively manufactured microfluidic device that can negatively enrich viable CTCs from clinically-relevant volumes of unmanipulated whole blood samples. Our device depletes nucleated blood cells based on their surface antigens and the smaller anucleated cells based on their size. Enriched CTCs are made available off the device in suspension making our technique compatible with standard immunocytochemical, molecular and functional assays. Our device could achieve a ~ 2.34-log depletion by capturing > 99.5% of white blood cells from 10 mL of whole blood while recovering > 90% of spiked tumor cells. Furthermore, we demonstrated the capability of the device to isolate CTCs from blood samples collected from patients (n = 15) with prostate and pancreatic cancers in a pilot study. A universal CTC assay that can differentiate tumor cells from normal blood cells with the specificity of clinically established membrane antigens yet require no label has the potential to enable routine blood-based tumor biopsies at the point-of-care.
Collapse
|
9
|
Real-Time Detection of Tumor Cells during Capture on a Filter Element Significantly Enhancing Detection Rate. BIOSENSORS-BASEL 2021; 11:bios11090312. [PMID: 34562902 PMCID: PMC8472380 DOI: 10.3390/bios11090312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/23/2021] [Accepted: 08/29/2021] [Indexed: 12/21/2022]
Abstract
Circulating tumor cells (CTCs) that enter the bloodstream play an important role in the formation of metastases. The prognostic significance of CTCs as biomarkers obtained from liquid biopsies is intensively investigated and requires accurate methods for quantification. The purpose of this study was the capture of CTCs on an optically accessible surface for real-time quantification. A filtration device was fabricated from a transparent material so that capturing of cells could be observed microscopically. Blood samples were spiked with stained tumor cells and the sample was filtrated using a porous structure with pore sizes of 7.4 µm. The possible removal of lysed erythrocytes and the retention of CTCs were assessed. The filtration process was observed in real-time using fluorescence microscopy, whereby arriving cells were counted in order to determine the number of CTCs present in the blood. Through optimization of the microfluidic channel design, the cell retention rate could be increased by 13% (from 76% ± 7% to 89% ± 5%). Providing the possibility for real-time detection significantly improved quantification efficiency even for the smallest cells evaluated. While end-point evaluation resulted in a detection rate of 63% ± 3% of the spiked cells, real-time evaluation led to an increase of 21% to 84% ± 4%. The established protocol provides an advantageous and efficient method for integration of fully automated sample preparation and CTC quantification into a lab-on-a-chip system.
Collapse
|
10
|
Li S, Li Y, Yao J, Chen B, Song J, Xue Q, Yang X. Label-free classification of dead and live colonic adenocarcinoma cells based on 2D light scattering and deep learning analysis. Cytometry A 2021; 99:1134-1142. [PMID: 34145728 DOI: 10.1002/cyto.a.24475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 01/20/2023]
Abstract
The measurement of cell viability plays an essential role in the area of cell biology. At present, the common methods for cell viability assay mainly on the responses of cells to different dyes. However, the additional steps of cell staining will consequently cause time-consuming and laborious efforts. Furthermore, the process of cell staining is invasive and may cause internal structure damage of cells, restricting their reuse in subsequent experiments. In this work, we proposed a label-free method to classify live and dead colonic adenocarcinoma cells by 2D light scattering combined with the deep learning algorithm. The deep convolutional network of YOLO-v3 was used to identify and classify light scattering images of live and dead HT29 cells. This method achieved an excellent sensitivity (93.6%), specificity (94.4%), and accuracy (94%). The results showed that the combination of 2D light scattering images and deep neural network may provide a new label-free method for cellular analysis.
Collapse
Affiliation(s)
- Shuaiyi Li
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Ya Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianning Yao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bing Chen
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiayou Song
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Qi Xue
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| | - Xiaonan Yang
- School of Information Engineering, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Circulating Tumor Cells from Enumeration to Analysis: Current Challenges and Future Opportunities. Cancers (Basel) 2021; 13:cancers13112723. [PMID: 34072844 PMCID: PMC8198976 DOI: 10.3390/cancers13112723] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 01/19/2023] Open
Abstract
Simple Summary With estimated numbers of 1–10 per mL of blood, circulating tumor cells (CTCs) are extremely rare compared to white (a few million) or red (billions) blood cells. Given their critical role in metastasis, CTCs have enormous potential as a biomarker for cancer diagnosis, prognosis, and monitoring of treatment response. There are now efforts to characterize CTCs more precisely through molecular and functional analysis, expanding the CTC effort from one of diagnosis and prognosis to now include the use of CTCs to specifically target cancers and discover therapeutic solutions, establishing CTCs as critical in precision medicine. This article summarizes current knowledge about CTC isolation technologies and discusses the translational benefits of different types of downstream analysis approaches, including single-CTC analysis, ex vivo expansion of CTCs, and characterization of CTC-associated cells. Abstract Circulating tumor cells (CTCs) have been recognized as a major contributor to distant metastasis. Their unique role as metastatic seeds renders them a potential marker in the circulation for early cancer diagnosis and prognosis as well as monitoring of therapeutic response. In the past decade, researchers mainly focused on the development of isolation techniques for improving the recovery rate and purity of CTCs. These developed techniques have significantly increased the detection sensitivity and enumeration accuracy of CTCs. Currently, significant efforts have been made toward comprehensive molecular characterization, ex vivo expansion of CTCs, and understanding the interactions between CTCs and their associated cells (e.g., immune cells and stromal cells) in the circulation. In this review, we briefly summarize existing CTC isolation technologies and specifically focus on advances in downstream analysis of CTCs and their potential applications in precision medicine. We also discuss the current challenges and future opportunities in their clinical utilization.
Collapse
|
12
|
Pahattuge TN, Freed IM, Hupert ML, Vaidyanathan S, Childers K, Witek MA, Weerakoon-Ratnayake K, Park D, Kasi A, Al-Kasspooles MF, Murphy MC, Soper SA. System Modularity Chip for Analysis of Rare Targets (SMART-Chip): Liquid Biopsy Samples. ACS Sens 2021; 6:1831-1839. [PMID: 33938745 DOI: 10.1021/acssensors.0c02728] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Liquid biopsies are becoming popular for managing a variety of diseases due to the minimally invasive nature of their acquisition, thus potentially providing better outcomes for patients. Circulating tumor cells (CTCs) are among the many different biomarkers secured from a liquid biopsy, and a number of efficient platforms for their isolation and enrichment from blood have been reported. However, many of these platforms require manual sample handling, which can generate difficulties when translating CTC assays into the clinic due to potential sample loss, contamination, and the need for highly specialized operators. We report a system modularity chip for the analysis of rare targets (SMART-Chip) composed of three task-specific modules that can fully automate processing of CTCs. The modules were used for affinity selection of the CTCs from peripheral blood with subsequent photorelease, simultaneous counting, and viability determinations of the CTCs and staining/imaging of the CTCs for immunophenotyping. The modules were interconnected to a fluidic motherboard populated with valves, interconnects, pneumatic control channels, and a fluidic network. The SMART-Chip components were made from thermoplastics via microreplication, which lowers the cost of production making it amenable to clinical implementation. The utility of the SMART-Chip was demonstrated by processing blood samples secured from colorectal cancer (CRC) and pancreatic ductal adenocarcinoma (PDAC) patients. We were able to affinity-select EpCAM expressing CTCs with high purity (0-3 white blood cells/mL of blood), enumerate the selected cells, determine their viability, and immunophenotype the cells. The assay could be completed in <4 h, while manual processing required >8 h.
Collapse
Affiliation(s)
- Thilanga N. Pahattuge
- Department of Chemistry, University of Kansas, 1567 Irving Hill Road, Lawrence, Kansas 66045, United States
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
| | - Ian M. Freed
- Department of Chemistry, University of Kansas, 1567 Irving Hill Road, Lawrence, Kansas 66045, United States
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
| | - Mateusz L. Hupert
- BioFluidica, Inc., 1567 Irving Hill Road, Lawrence, Kansas 66045, United States
| | - Swarnagowri Vaidyanathan
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
- Department of BioEngineering, University of Kansas, 1530 West 15th Street, Lawrence, Kansas 66045, United States
| | - Katie Childers
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
- Department of BioEngineering, University of Kansas, 1530 West 15th Street, Lawrence, Kansas 66045, United States
| | - Malgorzata A. Witek
- Department of Chemistry, University of Kansas, 1567 Irving Hill Road, Lawrence, Kansas 66045, United States
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
| | - Kumuditha Weerakoon-Ratnayake
- Department of Chemistry, University of Kansas, 1567 Irving Hill Road, Lawrence, Kansas 66045, United States
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
| | - Daniel Park
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
- Department of Mechanical & Industrial Engineering, Louisiana State University, 3261 Patrick F. Taylor Hall, Baton Rouge, Louisiana 70803, United States
| | - Anup Kasi
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Mazin F Al-Kasspooles
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Michael C. Murphy
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
- Department of Mechanical & Industrial Engineering, Louisiana State University, 3261 Patrick F. Taylor Hall, Baton Rouge, Louisiana 70803, United States
| | - Steven A. Soper
- Department of Chemistry, University of Kansas, 1567 Irving Hill Road, Lawrence, Kansas 66045, United States
- Center of BioModular Multi-scale Systems for Precision Medicine, University of Kansas, Lawrence, Kansas 66045, United States
- BioFluidica, Inc., 1567 Irving Hill Road, Lawrence, Kansas 66045, United States
- Department of BioEngineering, University of Kansas, 1530 West 15th Street, Lawrence, Kansas 66045, United States
- Department of Mechanical Engineering, University of Kansas, 3138 Learned Hall, 1530 West 15th Street, Lawrence, Kansas 66045, United States
| |
Collapse
|
13
|
Pang TCY, Po JW, Becker TM, Goldstein D, Pirola RC, Wilson JS, Apte MV. Circulating tumour cells in pancreatic cancer: A systematic review and meta-analysis of clinicopathological implications. Pancreatology 2021; 21:103-114. [PMID: 33309014 DOI: 10.1016/j.pan.2020.11.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The detection and quantification of circulating tumour cells (CTCs) in pancreatic cancer (PC) has the potential to provide prognostic information. The aim of this review was to provide an overview of the literature surrounding CTCs in PC. METHODS A systematic literature review on CTCs in PC between 2005-2020 was performed. Data based on peripheral vein samples were used to determine the positivity rate of CTCs, their prognostic significance and their relative numbers compared to portal vein (PV) samples. RESULTS The overall CTC detection rate in forty-four articles was 65% (95%CI: 55-75%). Detection rate for CellSearch was 26% (95%CI: 14-38%), which was lower than for both filtration and microfluidic techniques. In nine studies with >50 patients, overall survival was worse with CTC positivity (HR 1.82; 95%CI: 1.61-2.05). Five of seven studies which described PV CTC collection provided patient-level data. PV CTC yield was 7.7-fold (95%CI 1.35-43.9) that of peripheral blood. CONCLUSIONS CTCs were detected in the peripheral circulation of most patients with PC and may be related to prognosis and disease stage. PV blood contains more CTCs than peripheral blood sampling. This review points to the maturation of techniques of CTC enrichment, and its evidence base for eventual clinical deployment.
Collapse
Affiliation(s)
- Tony C Y Pang
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia; Surgical Innovations Unit, Westmead Hospital, Westmead, Australia; Westmead Clinical School, University of Sydney, Westmead, Australia
| | - Joseph W Po
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, South Western Clinical School, University of New South Wales, School of Medicine, Western Sydney University, Australia; Surgical Innovations Unit, Westmead Hospital, Westmead, Australia; Westmead Clinical School, University of Sydney, Westmead, Australia
| | - Therese M Becker
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, South Western Clinical School, University of New South Wales, School of Medicine, Western Sydney University, Australia
| | - David Goldstein
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Romano C Pirola
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Jeremy S Wilson
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, University of New South Wales, Australia.
| |
Collapse
|
14
|
Sun S, Yang S, Hu X, Zheng C, Song H, Wang L, Shen Z, Wu ZS. Combination of Immunomagnetic Separation with Aptamer-Mediated Double Rolling Circle Amplification for Highly Sensitive Circulating Tumor Cell Detection. ACS Sens 2020; 5:3870-3878. [PMID: 33205648 DOI: 10.1021/acssensors.0c01082] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most frequently diagnosed cancer among women, and the circulating tumor cell (CTC)-meditated distant metastasis is the leading cause of death. Thus, the detection of CTCs is of great importance for the early diagnosis of breast cancer and the prevention of metastasis. In this study, using human breast carcinoma BT474 cells as the model CTCs, a powerful assay platform is demonstrated by fluorescence spectrometry for the highly sensitive CTC detection by combining the dual-recognizing elements receptor-binding antibody and aptamer-mediated separation with double rolling circle amplification reactions (d-RCA, including RCA1 and RCA2). The aptamer-inserted RCA1 product (RCA1-p) exhibits the considerably improved affinity towards target cells originating from the multivalent binding effect. The immunomagnetic separation removes nontarget cells coexisting in complex biological milieu, while the centrifugal separation of cells/DNAs mixture eliminates the excess probes, thereby circumventing the unwanted interferences. The fluorescence spectrometric results show that a 34-fold enhanced fluorescence signal is achieved upon BT474 cells, and the target cells can be quantitatively detected down to 9 cells/200 μL with the linear range of five orders of magnitude, indicating a significantly enhanced detection performance. Even if BT474 cells are spiked in the fresh whole blood, no obvious fluctuation in the fluorescence signal is detected, demonstrating that the newly developed d-RCA assay system is suitable for screening CTCs in complex environments and is expected to be a promising tool for estimating distant metastasis and predicting the recurrence of tumors.
Collapse
Affiliation(s)
- Shujuan Sun
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Shulin Yang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xuemei Hu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Cheng Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang 325000, P.R. China
| | - Huanxia Song
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Lisha Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhifa Shen
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, China, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| |
Collapse
|
15
|
Czaplicka M, Niciński K, Nowicka A, Szymborski T, Chmielewska I, Trzcińska-Danielewicz J, Girstun A, Kamińska A. Effect of Varying Expression of EpCAM on the Efficiency of CTCs Detection by SERS-Based Immunomagnetic Optofluidic Device. Cancers (Basel) 2020; 12:cancers12113315. [PMID: 33182636 PMCID: PMC7697545 DOI: 10.3390/cancers12113315] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In this work we present a magnetically supported SERS-based immunoassay based on solid SERS-active support for the detection of circulating tumor cells. The SERS response in our optofluidic device was correlated with the level of EpCAM expression. The level of EpCAM cell expression in four cell lines with relatively high (human metastatic prostate adenocarcinoma cells (LNCaP)), medium (human metastatic prostate adenocarcinoma cells (LNCaP)), weak (human metastatic prostate adenocarcinoma cells (LNCaP)), and no EpCAM expressions (cervical cancer cells (HeLa) has been estimated using Western Blot method supported by immunochemistry and correlated with responses of immunomagnetic SERS-based analysis. The capture efficiency of developed assay was investigated in metastatic lung cancer patients. The assay demonstrates the capability to detect circulating tumor cells from blood samples over a broad linear range (from 1 to 100 cells/mL) reflecting clinically relevant amount of CTCs depending on the stage of metastasis, age, applied therapy. Abstract The circulating tumor cells (CTCs) isolation and characterization has a great potential for non-invasive biopsy. In the present research, the surface–enhanced Raman spectroscopy (SERS)-based assay utilizing magnetic nanoparticles and solid SERS-active support integrated in the external field assisted microfluidic device was designed for efficient isolation of CTCs from blood samples. Magnetic nanospheres (Fe2O3) were coated with SERS-active metal and then modified with p-mercaptobenzoic acid (p-MBA) which works simultaneously as a Raman reporter and linker to an antiepithelial-cell-adhesion-molecule (anti-EpCAM) antibodies. The newly developed laser-induced SERS-active silicon substrate with a very strong enhancement factor (up to 108) and high stability and reproducibility provide the additional extra-enhancement in the sandwich plasmonic configuration of immune assay which finally leads to increase the efficiency of detection. The sensitive immune recognition of cancer cells is assisted by the introducing of the controllable external magnetic field into the microfluidic chip. Moreover, the integration of the SERS-active platform and p-MBA-labeled immuno-Ag@Fe2O3 nanostructures with microfluidic device offers less sample and analytes demand, precise operation, increase reproducibly of spectral responses, and enables miniaturization and portability of the presented approach. In this work, we have also investigated the effect of varying expression of the EpCAM established by the Western Blot method supported by immunochemistry on the efficiency of CTCs’ detection with the developed SERS method. We used four target cancer cell lines with relatively high (human metastatic prostate adenocarcinoma cells (LNCaP)), medium (human metastatic prostate adenocarcinoma cells (LNCaP)), weak (human metastatic prostate adenocarcinoma cells (LNCaP)), and no EpCAM expressions (cervical cancer cells (HeLa)) to estimate the limits of detection based on constructed calibration curves. Finally, blood samples from lung cancer patients were used to validate the efficiency of the developed method in clinical trials.
Collapse
Affiliation(s)
- Marta Czaplicka
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland; (M.C.); (K.N.); (A.N.); (T.S.)
| | - Krzysztof Niciński
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland; (M.C.); (K.N.); (A.N.); (T.S.)
| | - Ariadna Nowicka
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland; (M.C.); (K.N.); (A.N.); (T.S.)
| | - Tomasz Szymborski
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland; (M.C.); (K.N.); (A.N.); (T.S.)
| | - Izabela Chmielewska
- Department of Pneumology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland;
| | - Joanna Trzcińska-Danielewicz
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland; (J.T.-D.); (A.G.)
| | - Agnieszka Girstun
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland; (J.T.-D.); (A.G.)
| | - Agnieszka Kamińska
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland; (M.C.); (K.N.); (A.N.); (T.S.)
- Correspondence:
| |
Collapse
|
16
|
Zhao X, Park DSW, Soper SA, Murphy MC. Microfluidic gasketless interconnects sealed by superhydrophobic surfaces. JOURNAL OF MICROELECTROMECHANICAL SYSTEMS : A JOINT IEEE AND ASME PUBLICATION ON MICROSTRUCTURES, MICROACTUATORS, MICROSENSORS, AND MICROSYSTEMS 2020; 29:894-899. [PMID: 33746475 PMCID: PMC7978084 DOI: 10.1109/jmems.2020.3000325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Existing methods for sealing chip-to-chip (or module-to-motherboard) microfluidic interconnects commonly use additional interconnect components (O-rings, gaskets, and tubing), and manual handling expertise for assembly. Novel gasketless superhydrophobic fluidic interconnects (GSFIs) sealed by transparent superhydrophobic surfaces, forming liquid bridges between the fluidic ports for fluidic passages were demonstrated. Two test platforms were designed, fabricated, and evaluated, a multi-port chip system (ten interconnects) and a modules-on-a-motherboard system (four interconnects). System assembly in less than 3 sec was done by embedded magnets and pin-in-V-groove structures. Flow tests with deionized (DI) water, ethanol/water mixture, and plasma confirmed no leakage through the gasketless interconnects up to a maximum flow rate of 100 μL/min for the multi-port chip system. The modules-on-a-motherboard system showed no leakage of water at a flow rate of 20 μL/min and a pressure drop of 3.71 psi. Characterization of the leakage pressure as a function of the surface tension of the sample liquid in the multi-port chip system revealed that lower surface tension of the liquid led to lower static water contact angles on the superhydrophobic-coated substrate and lower leakage pressures. The high-density, rapidly assembled, gasketless interconnect technology will open up new avenues for chip-to-chip fluid transport in complex microfluidic modular systems.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Center for BioModular Multiscale Systems for Precision Medicine, Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Daniel S-W Park
- Center for BioModular Multiscale Systems for Precision Medicine, Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Steven A Soper
- Center for BioModular Multiscale Systems for Precision Medicine Departments of Chemistry and Mechanical Engineering, University of Kansas, Lawrence, KS, 66045, USA
| | - Michael C Murphy
- Center for BioModular Multiscale Systems for Precision Medicine, Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
17
|
Target-triggered "signal-off" electrochemical aptasensor assisted by Au nanoparticle-modified sensing platform for high-sensitivity determination of circulating tumor cells. Anal Bioanal Chem 2020; 412:8107-8115. [PMID: 32929571 DOI: 10.1007/s00216-020-02940-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/17/2022]
Abstract
In this study, we fabricated a high-sensitivity "signal-off" electrochemical aptasensing platform for quantifying circulating tumor cells (CTCs) based on target-triggered signal readout of methylene blue (MB). Au nanoparticles (AuNPs) were introduced to enlarge the specific surface area of the gold electrode (GE), which would immobilize homogeneous and more MB-aptamers. MB-modified and stem-loop-like aptamers were assigned as a recognition element with K562 cells. Thiolated complementary strands hybridized with MB-aptamers to form double-stranded DNA (dsDNA) conformation which were further self-assembled on the surface of AuNP-modified GE, leading to a marked current peak of MB signal. In the presence of K562 cells, the MB-aptamers preferred to recognize and bind with the cells, causing the disassembly of MB-aptamers from the GE surface. Therefore, the reduced value of MB signal was related to the number of K562 cells. With the proposed aptasensor, a dynamic linear range from 1 × 102 to 1 × 106 cells mL-1 was obtained with a detection limit of 23 cells mL-1. Moreover, the aptasensor showed good selectivity, stability, and reproducibility as well as potential use in the clinical setting. Meanwhile, characterization techniques such as field-emission scanning electron microscopy, energy-dispersive X-ray spectroscopy, atomic force microscopy, cyclic voltammetry, and electrochemical impedance spectroscopy were performed to analyze the evolution of the morphology and each fabricated step of the constructed aptasensor. Our proposed aptasensor could be designed as a universal platform for CTC determination by replacing tumor cell-specific aptamers, which is a promising strategy for basic research and clinical applications. Graphical abstract.
Collapse
|
18
|
Park DSW, Young BM, You BH, Singh V, Soper SA, Murphy MC. An integrated, optofluidic system with aligned optical waveguides, microlenses, and coupling prisms for fluorescence sensing. JOURNAL OF MICROELECTROMECHANICAL SYSTEMS : A JOINT IEEE AND ASME PUBLICATION ON MICROSTRUCTURES, MICROACTUATORS, MICROSENSORS, AND MICROSYSTEMS 2020; 29:600-609. [PMID: 39391841 PMCID: PMC11465942 DOI: 10.1109/jmems.2020.3004374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
An improved, laser-induced fluorescence-based micro-optical biosensor was designed and fabricated, with cyclic olefin copolymer (COC) optical waveguides, a poly(methyl methacrylate) (PMMA) fluidic substrate with an array of microlenses, and a COC coupling prism integrated with the waveguide substrate or cover plate. The double-sided hot embossed fluidic substrate had sampling zone microchannels on the bottom and microlenses on the top. Dissolved COC injected into polydimethylsiloxane (PDMS) lost molds embedded the waveguides in the PMMA cover plate and formed the integrated coupling prism. The embedded COC waveguide was flycut down to 50 μm. The cover plate and shallow, 1:20 aspect ratio, microchannels were thermal fusion bonded using a pressure-assisted boiling point control system, without sagging. The large COC prism coupled better to the waveguide. The highest intensity evanescent excitation of the waveguide was obtained near the critical angle. The maximum signal-to-noise ratio (SNR) was 119 and the lowest detection limit was 7.34 × 10-20 mol at a SNR of 2 for a 100 μm wide by 50 μm deep waveguide. The microlenses highly focused the fluorescent radiation in the sampling zone. The microfabricated waveguide enables rapid, low-cost detection of fluorescent samples with high SNR, a low detection limit, and high sampling efficiency.
Collapse
Affiliation(s)
- Daniel S-W Park
- Center for BioModular Multiscale Systems for Precision Medicine, Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Brandon M Young
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA and is now with the Center for BioModular Multiscale Systems for Precision Medicine, at the University of Kansas, Lawrence, KS 66045
| | - Byoung H You
- Department of Engineering Technology, Texas State University, San Marcos, TX 78666, USA
| | - Varshni Singh
- Center for Advanced Microstructures and Devices (CAMD), Louisiana State University, Baton Rouge, LA 70806, USA
| | - Steven A Soper
- Center for BioModular Multiscale Systems for Precision Medicine, Departments of Chemistry and Mechanical Engineering, University of Kansas, Lawrence, KS, 66045, USA
| | - Michael C Murphy
- Center for BioModular Multiscale Systems for Precision Medicine, Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
19
|
Habli Z, AlChamaa W, Saab R, Kadara H, Khraiche ML. Circulating Tumor Cell Detection Technologies and Clinical Utility: Challenges and Opportunities. Cancers (Basel) 2020; 12:cancers12071930. [PMID: 32708837 PMCID: PMC7409125 DOI: 10.3390/cancers12071930] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/03/2020] [Accepted: 06/17/2020] [Indexed: 12/16/2022] Open
Abstract
The potential clinical utility of circulating tumor cells (CTCs) in the diagnosis and management of cancer has drawn a lot of attention in the past 10 years. CTCs disseminate from tumors into the bloodstream and are believed to carry vital information about tumor onset, progression, and metastasis. In addition, CTCs reflect different biological aspects of the primary tumor they originate from, mainly in their genetic and protein expression. Moreover, emerging evidence indicates that CTC liquid biopsies can be extended beyond prognostication to pharmacodynamic and predictive biomarkers in cancer patient management. A key challenge in harnessing the clinical potential and utility of CTCs is enumerating and isolating these rare heterogeneous cells from a blood sample while allowing downstream CTC analysis. That being said, there have been serious doubts regarding the potential value of CTCs as clinical biomarkers for cancer due to the low number of promising outcomes in the published results. This review aims to present an overview of the current preclinical CTC detection technologies and the advantages and limitations of each sensing platform, while surveying and analyzing the published evidence of the clinical utility of CTCs.
Collapse
Affiliation(s)
- Zeina Habli
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
| | - Walid AlChamaa
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
| | - Raya Saab
- Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon;
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, 77030 TX, USA;
| | - Massoud L. Khraiche
- Neural Engineering and Nanobiosensors Group, Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; (Z.H.); (W.A.)
- Correspondence:
| |
Collapse
|
20
|
Kong C, Hu M, Weerakoon-Ratnayake KM, Witek MA, Dathathreya K, Hupert ML, Soper SA. Label-free counting of affinity-enriched circulating tumor cells (CTCs) using a thermoplastic micro-Coulter counter (μCC). Analyst 2020; 145:1677-1686. [PMID: 31867587 PMCID: PMC7350181 DOI: 10.1039/c9an01802f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Coulter counters are used for counting particles and biological cells. Most Coulter counters are designed to analyze a sample without the ability to pre-process the sample prior to counting. For the analysis of rare cells, such as circulating tumor cells (CTCs), it is not uncommon to require enrichment before counting due to the modest throughput of μCCs and the high abundance of interfering cells, such as blood cells. We report a microfluidic-based Coulter Counter (μCC) fabricated using simple, low-cost techniques for counting rare cells that can be interfaced to sample pre- and/or post-processing units. In the current work, a microfluidic device for the affinity-based enrichment of CTCs from whole blood into a relatively small volume of ∼10 μL was interfaced to the μCC to allow for exhaustive counting of single CTCs following release of the CTCs from the enrichment chip. When integrated to the CTC affinity enrichment chip, the μCC could count the CTCs without loss and the cells could be collected for downstream molecular profiling or culturing if required. The μCC sensor counting efficiency was >93% and inter-chip variability was ∼1%.
Collapse
Affiliation(s)
- Cong Kong
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA and Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA and Key Laboratory of East China Sea Fishery Resources Exploitation, Ministry of Agriculture and Rural Affairs, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai 200090, China
| | - Mengjia Hu
- BioFluidica, Inc., Lawrence, KS 66047, USA.
| | - Kumuditha M Weerakoon-Ratnayake
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA and Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA
| | - Malgorzata A Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA and Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA
| | - Kavya Dathathreya
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA and Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA
| | | | - Steven A Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA and Center of BioModular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA and BioFluidica, Inc., Lawrence, KS 66047, USA. and BioEngineering Program, The University of Kansas, Lawrence, KS 66047, USA and Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66047.
| |
Collapse
|
21
|
M. Weerakoon-Ratnayake K, Vaidyanathan S, Larkey N, Dathathreya K, Hu M, Jose J, Mog S, August K, K. Godwin A, L. Hupert M, A. Witek M, A. Soper S. Microfluidic Device for On-Chip Immunophenotyping and Cytogenetic Analysis of Rare Biological Cells. Cells 2020; 9:E519. [PMID: 32102446 PMCID: PMC7072755 DOI: 10.3390/cells9020519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 01/09/2023] Open
Abstract
The role of circulating plasma cells (CPCs) and circulating leukemic cells (CLCs) as biomarkers for several blood cancers, such as multiple myeloma and leukemia, respectively, have recently been reported. These markers can be attractive due to the minimally invasive nature of their acquisition through a blood draw (i.e., liquid biopsy), negating the need for painful bone marrow biopsies. CPCs or CLCs can be used for cellular/molecular analyses as well, such as immunophenotyping or fluorescence in situ hybridization (FISH). FISH, which is typically carried out on slides involving complex workflows, becomes problematic when operating on CLCs or CPCs due to their relatively modest numbers. Here, we present a microfluidic device for characterizing CPCs and CLCs using immunofluorescence or FISH that have been enriched from peripheral blood using a different microfluidic device. The microfluidic possessed an array of cross-channels (2-4 µm in depth and width) that interconnected a series of input and output fluidic channels. Placing a cover plate over the device formed microtraps, the size of which was defined by the width and depth of the cross-channels. This microfluidic chip allowed for automation of immunofluorescence and FISH, requiring the use of small volumes of reagents, such as antibodies and probes, as compared to slide-based immunophenotyping and FISH. In addition, the device could secure FISH results in <4 h compared to 2-3 days for conventional FISH.
Collapse
Affiliation(s)
- Kumuditha M. Weerakoon-Ratnayake
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Swarnagowri Vaidyanathan
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
- Bioengineering, The University of Kansas, Lawrence, KS 66045, USA
| | - Nicholas Larkey
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Kavya Dathathreya
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Mengjia Hu
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Jilsha Jose
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Shalee Mog
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Keith August
- Children’s Mercy Hospital, Kansas City, MO 64108, USA;
| | - Andrew K. Godwin
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Mateusz L. Hupert
- Biofluidica Inc., BioFluidica Research Laboratory, Lawrence, KS 66047, USA
| | - Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA; (K.M.W.-R.); (K.D.); (S.M.)
- Center of BioModular Multiscale Systems for Precision Medicine, Lawrence, KS 66045, USA; (S.V.); (N.L.); (M.H.); (J.J.)
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Biofluidica Inc., BioFluidica Research Laboratory, Lawrence, KS 66047, USA
- Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
22
|
McGrath JS, Honrado C, Moore JH, Adair SJ, Varhue WB, Salahi A, Farmehini V, Goudreau BJ, Nagdas S, Blais EM, Bauer TW, Swami NS. Electrophysiology-based stratification of pancreatic tumorigenicity by label-free single-cell impedance cytometry. Anal Chim Acta 2019; 1101:90-98. [PMID: 32029124 DOI: 10.1016/j.aca.2019.12.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/13/2019] [Accepted: 12/14/2019] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer lacking specific biomarkers that can be correlated to disease onset, promotion and progression. To assess whether tumor cell electrophysiology may serve as a marker for PDAC tumorigenicity, we use multi-frequency impedance cytometry at high throughput (∼350 cells/s) to measure the electrical phenotype of single PDAC tumor cells from xenografts, which are derived from primary pancreatic tumors versus those from liver metastases of different patients. A novel phase contrast metric based on variations in the high and low frequency impedance phase responses that is related to electrophysiology of the cell interior is found to be systematically altered as a function of tumorigenicity. PDAC cells of higher tumorigenicity exhibited lowered interior conductivity and enhanced permittivity, which is validated by the dielectrophoresis on the respective cell types. Using genetic analysis, we suggest the role of dysregulated Na+ transport and removal of Ca2+ ions from the cytoplasm on key oncogenic KRAS-driven processes that may be responsible for lowering of the interior cell conductivity. We envision that impedance cytometry can serve as a tool to quantify phenotypic heterogeneity for rapidly stratifying tumorigenicity. It can also aid in protocols for dielectrophoretic isolation of cells with a particular phenotype for prognostic studies on patient survival and to tailor therapy selection to specific patients.
Collapse
Affiliation(s)
- J S McGrath
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - C Honrado
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - J H Moore
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - S J Adair
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - W B Varhue
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - A Salahi
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - V Farmehini
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - B J Goudreau
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - S Nagdas
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - E M Blais
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - T W Bauer
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - N S Swami
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
23
|
On-chip label-free determination of cell survival rate. Biosens Bioelectron 2019; 148:111820. [PMID: 31706174 DOI: 10.1016/j.bios.2019.111820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 10/01/2019] [Accepted: 10/23/2019] [Indexed: 12/25/2022]
Abstract
Cell survival rate (CSR) is a very important parameter in biological and medical fields. Today, the routine method to determine this parameter is time-consuming; it also makes the labeled cells no longer useable for subsequent experiments. Here, we developed an on-chip label-free method for determining the CSR. For the method, a hypertonic stimulus was designed to create volume differences between living and dead cells, and then, the differences were characterized with measurements of impedance as the cells flowed through two electrodes. Based on the method, a microfluidic hypertonic stimulus-based impedance flow cytometry chip (HSIFC) was designed, and the localized function of the HSIFC was verified. Finally, the performance of the HSIFC was confirmed by measuring the different CSRs for the different types of cells. The results show that the HSIFC can accurately determine the CSR, and the accuracy is comparable to that of flow cytometry. This work paves the way for the label-free evaluation of CSR after various cell manipulations and treatments on the chip and promotes the versatility of lab-on-a-chip devices.
Collapse
|
24
|
Shen C, Liu S, Li X, Yang M. Electrochemical Detection of Circulating Tumor Cells Based on DNA Generated Electrochemical Current and Rolling Circle Amplification. Anal Chem 2019; 91:11614-11619. [PMID: 31452368 DOI: 10.1021/acs.analchem.9b01897] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Circulating tumor cells (CTCs) are important indicators for tumor diagnosis and tumor metastasis. However, the extremely low levels of CTCs in peripheral blood challenges the precise detection of CTCs. Herein, we report DNA generated electrochemical current combined with rolling circle amplification (RCA) as well as magnetic nanospheres for highly efficient magnetic capture and ultrasensitive detection of CTCs. The antiepithelial cell adhesion molecule (EpCAM) antibody-modified magnetic nanospheres were used to capture and enrich CTCs. The following binding of an aptamer onto the CTC surface and the subsequent RCA assembled a significant amount of DNA molecules onto the electrode. The reaction of the DNA molecules with molybdate can then form redox molybdophosphate and produce an electrochemical current. Using the breast cancer cell MCF-7 as a model, the sensor displays good performances toward detection of MCF-7 that was spiked into peripheral blood. The signal amplification strategy integrated with a magnetic nanosphere platform exhibits good performance in the efficient capture and detection of CTCs, which may find wide potential in cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Congcong Shen
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering , Central South University , Changsha 410083 , China.,School of Chemistry and Chemical Engineering , Henan Normal University , Xinxiang 453007 , China
| | - Shuping Liu
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering , Central South University , Changsha 410083 , China
| | - Xiaoqing Li
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering , Central South University , Changsha 410083 , China
| | - Minghui Yang
- Key Laboratory of Hunan Province for Water Environment and Agriculture Product Safety, College of Chemistry and Chemical Engineering , Central South University , Changsha 410083 , China
| |
Collapse
|
25
|
Lee J, Park SS, Lee YK, Norton JA, Jeffrey SS. Liquid biopsy in pancreatic ductal adenocarcinoma: current status of circulating tumor cells and circulating tumor DNA. Mol Oncol 2019; 13:1623-1650. [PMID: 31243883 PMCID: PMC6670020 DOI: 10.1002/1878-0261.12537] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/07/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
Reliable biomarkers are required to evaluate and manage pancreatic ductal adenocarcinoma. Circulating tumor cells and circulating tumor DNA are shed into blood and can be relatively easily obtained from minimally invasive liquid biopsies for serial assays and characterization, thereby providing a unique potential for early diagnosis, forecasting disease prognosis, and monitoring of therapeutic response. In this review, we provide an overview of current technologies used to detect circulating tumor cells and circulating tumor DNA and describe recent advances regarding the multiple clinical applications of liquid biopsy in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Jee‐Soo Lee
- Department of Laboratory MedicineHallym University Sacred Heart HospitalAnyangKorea
- Department of Laboratory MedicineSeoul National University College of MedicineSeoulKorea
| | - Sung Sup Park
- Department of Laboratory MedicineSeoul National University College of MedicineSeoulKorea
| | - Young Kyung Lee
- Department of Laboratory MedicineHallym University Sacred Heart HospitalAnyangKorea
- Department of Laboratory MedicineHallym University College of MedicineAnyangKorea
| | - Jeffrey A. Norton
- Department of SurgeryStanford University School of MedicineStanfordCAUSA
| | | |
Collapse
|
26
|
Tian F, Liu C, Lin L, Chen Q, Sun J. Microfluidic analysis of circulating tumor cells and tumor-derived extracellular vesicles. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.05.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
27
|
Sun Y, Wu G, Cheng KS, Chen A, Neoh KH, Chen S, Tang Z, Lee PF, Dai M, Han RPS. CTC phenotyping for a preoperative assessment of tumor metastasis and overall survival of pancreatic ductal adenocarcinoma patients. EBioMedicine 2019; 46:133-149. [PMID: 31375425 PMCID: PMC6712350 DOI: 10.1016/j.ebiom.2019.07.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 06/27/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The evaluation for surgical resectability of pancreatic ductal adenocarcinoma (PDAC) patients is not only imaging-based but highly subjective. An objective method is urgently needed. We report on the clinical value of a phenotypic circulating tumor cell (CTC)-based blood test for a preoperative prognostic assessment of tumor metastasis and overall survival (OS) of PDAC patients. METHODS Venous blood samples from 46 pathologically confirmed PDAC patients were collected prospectively before surgery and immunoassayed using a specially designed TU-chip™. Captured CTCs were differentiated into epithelial (E), mesenchymal and hybrid (H) phenotypes. A further 45 non-neoplastic healthy donors provided blood for cell line validation study and CTC false positive quantification. FINDINGS A validated multivariable model consisting of disjunctively combined CTC phenotypes: "H-CTC≥15.0 CTCs/2ml OR E-CTC≥11.0 CTCs/2ml" generated an optimal prediction of metastasis with a sensitivity of 1.000 (95% CI 0.889-1.000) and specificity of 0.886 (95% CI 0.765-0.972). The adjusted Kaplan-Meier median OS constructed using Cox proportional-hazard models and stratified for E-CTC < 11.0 CTCs/2 ml was 16.5 months and for E-CTC ≥ 11.0 CTCs/2 ml was 5.5 months (HR = 0.050, 95% CI 0.004-0.578, P = .016). These OS results were consistent with the outcome of the metastatic analysis. INTERPRETATION Our work suggested that H-CTC is a better predictor of metastasis and E-CTC is a significant independent predictor of OS. The CTC phenotyping model has the potential to be developed into a reliable and accurate blood test for metastatic and OS assessments of PDAC patients. FUND: National Natural Science Foundation of China; Zhejiang Province Science and Technology Program; China Scholarship Council.
Collapse
Affiliation(s)
- Yukun Sun
- College of Engineering, Peking University, Beijing 100871, China
| | - Guangdong Wu
- Dept of Hepatopancreatobiliary Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Institute for Precision Medicine, Tsinghua University, Beijing 102218, China; Dept. of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Kok Suen Cheng
- College of Engineering, Peking University, Beijing 100871, China
| | - Anqi Chen
- College of Engineering, Peking University, Beijing 100871, China
| | - Kuang Hong Neoh
- College of Engineering, Peking University, Beijing 100871, China
| | - Shuiyu Chen
- College of Engineering, Peking University, Beijing 100871, China
| | - Zhewen Tang
- College of Engineering, Peking University, Beijing 100871, China
| | - Poh Foong Lee
- Dept. of Mechanical & Materials Engineering, University Tunku Abdul Rahman, Bandar Sungai Long, Selangor, Malaysia
| | - Menghua Dai
- Dept. of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Ray P S Han
- College of Engineering, Peking University, Beijing 100871, China; Integrated Chinese & Western Medicine Oncology Research Center, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, China..
| |
Collapse
|
28
|
Yu D, Tang L, Dong Z, Loftis KA, Ding Z, Cheng J, Qin B, Yan J, Li W. Effective reduction of non-specific binding of blood cells in a microfluidic chip for isolation of rare cancer cells. Biomater Sci 2019; 6:2871-2880. [PMID: 30246818 DOI: 10.1039/c8bm00864g] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The high purity of target cells enriched from blood samples plays an important role in the clinical detection of diseases. However, non-specific binding of blood cells in the isolated cell samples can complicate downstream molecular and genetic analysis. In this work, we report a simple solution to non-specific binding of blood cells by modifying the surface of microchips with a multilayer nanofilm, with the outmost layer containing both PEG brushes for reducing blood cell adhesion and antibodies for enriching target cells. This layer-by-layer (LbL) polysaccharide nanofilm was modified with neutravindin and then conjugated with a mixture of biotinylated PEG molecules and biotinylated antibodies. Using EpCAM-expressing and HER2-expressing cancer cells in blood as model platforms, we were able to dramatically reduce the non-specific binding of blood cells to approximately 1 cell per mm2 without sacrificing the high capture efficiency of the microchip. To support the rational extension of this approach to other applications for cell isolation and blood cell resistance, we conducted extensive characterization on the nanofilm formation and degradation, antifouling with PEG brushes and introducing functional antibodies. This simple, yet effective, approach can be applied to a variety of microchip applications that require high purity of sample cells containing minimal contamination from blood cells.
Collapse
Affiliation(s)
- Dan Yu
- Department of Critical Care Medicine, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou, 450003, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Insights on CTC Biology and Clinical Impact Emerging from Advances in Capture Technology. Cells 2019; 8:cells8060553. [PMID: 31174404 PMCID: PMC6627072 DOI: 10.3390/cells8060553] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 01/01/2023] Open
Abstract
Circulating tumor cells (CTCs) and circulating tumor microemboli (CTM) have been shown to correlate negatively with patient survival. Actual CTC counts before and after treatment can be used to aid in the prognosis of patient outcomes. The presence of circulating tumor materials (CTMat) can advertise the presence of metastasis before clinical presentation, enabling the early detection of relapse. Importantly, emerging evidence is indicating that cancer treatments can actually increase the incidence of CTCs and metastasis in pre-clinical models. Subsequently, the study of CTCs, their biology and function are of vital importance. Emerging technologies for the capture of CTC/CTMs and CTMat are elucidating vitally important biological and functional information that can lead to important alterations in how therapies are administered. This paves the way for the development of a "liquid biopsy" where treatment decisions can be informed by information gleaned from tumor cells and tumor cell debris in the blood.
Collapse
|
30
|
Hu W, Soper SA, Jackson JM. Time-Delayed Integration-Spectral Flow Cytometer (TDI-SFC) for Low-Abundance-Cell Immunophenotyping. Anal Chem 2019; 91:4656-4664. [PMID: 30817129 PMCID: PMC6554645 DOI: 10.1021/acs.analchem.9b00021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We describe a unique flow cytometer (TDI-SFC) for the immunophenotyping of low-abundance cells, particularly when cell counts are sample-limited and operationally difficult for analysis by fluorescence microscopy (>100 cells) or multiparameter flow cytometry (MFC, <10 000 cells). TDI-SFC combines the high spectral resolution of spectral flow cytometry (SFC) with a CCD operated in time-delayed integration (TDI) for improved duty cycle and sensitivity. Cells were focused with a 1D-sheathing microfluidic device, and fluorescence emission generated from a 488 nm laser was collected by epi-illumination and dispersed along one axis of a CCD by a spectrograph. Along the other axis, the CCD's shift rate was clocked at a rate that closely matched the cells' velocity through the field of view. This TDI-SFC format allowed the CCD shutter to remain open during signal acquisition, providing a duty cycle ∼100% and assurance that ∼95% cells were interrogated. We used fluorescent beads to optimize synchronization of TDI clocking with the sheathed-cell velocity and to improve sensitivity via the excitation intensity, epi-illumination numerical aperture, and integration time. TDI achieved integrated signals of 106 counts at a signal-to-noise ratio (SNR) of 610 for beads corresponding to a load of 4 × 105 antibodies. We also evaluated multiplexing capabilities by spectral deconvolution and undertook a proof-of-concept application to immunophenotype low-abundance cells; the demonstration consisted of immunophenotyping a model cell line, in this case SUP-B15 cells representing B-cell acute lymphoblastic leukemia (B-ALL). The B-ALL cell line was stained against a leukemic marker (terminal deoxynucleotidyl transferase, TdT), and we successfully used spectral unmixing to discriminate TdT(+) cells from TdT(-) cells even at low cell counts (∼100 cells). The TDI-SFC could potentially be used in any application requiring the immunophenotyping of low-abundance cells, such as in monitoring measurable residual disease in acute leukemias following affinity enrichment of circulating leukemia cells from peripheral blood.
Collapse
Affiliation(s)
- Wenting Hu
- Department of Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multi-Scale Systems for Precision Medicine (CBM), University of Kansas, Lawrence, Kansas 66045, United States
| | - Steven A. Soper
- Department of Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multi-Scale Systems for Precision Medicine (CBM), University of Kansas, Lawrence, Kansas 66045, United States
- Department of Mechanical Engineering, University of Kansas, Lawrence, Kansas 66045, United States
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - J. Matt Jackson
- Department of Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
- Center of BioModular Multi-Scale Systems for Precision Medicine (CBM), University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
31
|
Cho H, Kim J, Song H, Sohn KY, Jeon M, Han KH. Microfluidic technologies for circulating tumor cell isolation. Analyst 2019; 143:2936-2970. [PMID: 29796523 DOI: 10.1039/c7an01979c] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metastasis is the main cause of tumor-related death, and the dispersal of tumor cells through the circulatory system is a critical step in the metastatic process. Early detection and analysis of circulating tumor cells (CTCs) is therefore important for early diagnosis, prognosis, and effective treatment of cancer, enabling favorable clinical outcomes in cancer patients. Accurate and reliable methods for isolating and detecting CTCs are necessary to obtain this clinical information. Over the past two decades, microfluidic technologies have demonstrated great potential for isolating and detecting CTCs from blood. The present paper reviews current advanced microfluidic technologies for isolating CTCs based on various biological and physical principles, and discusses their fundamental advantages and drawbacks for subsequent cellular and molecular assays. Owing to significant genetic heterogeneity among CTCs, microfluidic technologies for isolating individual CTCs have recently been developed. We discuss these single-cell isolation methods, as well as approaches to overcoming the limitations of current microfluidic CTC isolation technologies. Finally, we provide an overview of future innovative microfluidic platforms.
Collapse
Affiliation(s)
- Hyungseok Cho
- Department of Nanoscience and Engineering, Center for Nano Manufacturing, Inje University, Gimhae 621-749, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
32
|
Thege FI, Gruber CN, Cardle II, Cong SH, Lannin TB, Kirby BJ. anti-EGFR capture mitigates EMT- and chemoresistance-associated heterogeneity in a resistance-profiling CTC platform. Anal Biochem 2019; 577:26-33. [PMID: 30790546 DOI: 10.1016/j.ab.2019.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 01/18/2019] [Accepted: 02/06/2019] [Indexed: 12/19/2022]
Abstract
Capture and analysis of circulating tumor cells (CTCs) holds promise for diagnosing and guiding treatment of pancreatic cancer. To accurately monitor disease progression, capture platforms must be robust to processes that increase the phenotypic heterogeneity of CTCs. Most CTC-analysis technologies rely on the recognition of epithelial-specific markers for capture and identification, in particular the epithelial cell-adhesion molecule (EpCAM) and cytokeratin. As the epithelial-to-mesenchymal transition (EMT) and the acquisition of chemoresistance are both associated with loss of epithelial markers and characteristics, the effect of these processes on the expression of commonly used CTC markers, specifically EpCAM, EGFR and cytokeratin, requires further exploration. To determine this effect, we developed an in vitro model of EMT and acquired gemcitabine resistance in human pancreatic cancer cell lines. Using this model, we show that EMT-induction and acquired chemoresistance decrease EpCAM expression and microfluidic anti-EpCAM capture performance. Furthermore, we find that EGFR capture is more robust to these processes. By measuring the expression of known mediators of chemoresistance in captured cells using automated imaging and image processing, we demonstrate the ability to resistance-profile cells on-chip. We expect that this approach will allow for the development of improved non-invasive biomarkers of pancreatic cancer progression.
Collapse
Affiliation(s)
| | | | | | | | | | - Brian J Kirby
- Cornell University, Ithaca, USA; Weill Cornell Medicine, New York, USA.
| |
Collapse
|
33
|
Samandari M, Julia MG, Rice A, Chronopoulos A, Del Rio Hernandez AE. Liquid biopsies for management of pancreatic cancer. Transl Res 2018; 201:98-127. [PMID: 30118658 DOI: 10.1016/j.trsl.2018.07.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/17/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is one of the main causes of cancer-related deaths worldwide. It is asymptomatic at an early stage, and most diagnosis occurs when the disease is already at a late stage, by which time the tumor is nonresectable. In order to increase the overall survival of patients with pancreatic cancer, as well as to decrease the cancer burden, it is necessary to perform early diagnosis, prognosis stratifications and cancer monitoring using accurate, minimally invasive, and cost-effective methods. Liquid biopsies seek to detect tumor-associated biomarkers in a variety of extractable body fluids and can help to monitor treatment response and disease progression, and even predict patient outcome. In patients with pancreatic cancer, tumor-derived materials, primarily circulating tumor DNA, circulating tumor cells and exosomes, are being studied for inclusion in the management of the disease. This review focuses on describing the biology of these biomarkers, methods for their enrichment and detection, as well as their potential for clinical application. Moreover, we discuss the future direction of liquid biopsies and introduce how they can be exploited toward point of care personalized medicine for the management of pancreatic cancer.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - María Gil Julia
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Antonios Chronopoulos
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Armando E Del Rio Hernandez
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom.
| |
Collapse
|
34
|
Tang W, Jiang D, Li Z, Zhu L, Shi J, Yang J, Xiang N. Recent advances in microfluidic cell sorting techniques based on both physical and biochemical principles. Electrophoresis 2018; 40:930-954. [DOI: 10.1002/elps.201800361] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 09/28/2018] [Accepted: 09/30/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Wenlai Tang
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
- Nanjing Institute of Intelligent High-end Equipment Industry Co., Ltd.; P. R. China
| | - Di Jiang
- School of Mechanical and Electronic Engineering; Nanjing Forestry University; P. R. China
| | - Zongan Li
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
| | - Liya Zhu
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
| | - Jianping Shi
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
| | - Jiquan Yang
- School of Electrical and Automation Engineering; Jiangsu Key Laboratory of 3D Printing Equipment and Manufacturing; Nanjing Normal University; P. R. China
- Nanjing Institute of Intelligent High-end Equipment Industry Co., Ltd.; P. R. China
| | - Nan Xiang
- School of Mechanical Engineering; Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments; Southeast University; P. R. China
| |
Collapse
|
35
|
Wu LL, Tang M, Zhang ZL, Qi CB, Hu J, Ma XY, Pang DW. Chip-Assisted Single-Cell Biomarker Profiling of Heterogeneous Circulating Tumor Cells Using Multifunctional Nanospheres. Anal Chem 2018; 90:10518-10526. [DOI: 10.1021/acs.analchem.8b02585] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Ling-Ling Wu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Man Tang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Zhi-Ling Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Chu-Bo Qi
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Jiao Hu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Xu-Yan Ma
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| | - Dai-Wen Pang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Wuhan 430072, PR China
| |
Collapse
|
36
|
Ren D, Chui CO. Feasibility of Tracking Multiple Single-Cell Properties with Impedance Spectroscopy. ACS Sens 2018; 3:1005-1015. [PMID: 29737153 DOI: 10.1021/acssensors.8b00152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Electric cell-substrate impedance sensing (ECIS) has been instrumental in tracking collective behavior of confluent cell layers for decades. Toward probing cellular heterogeneity in a population, the single-cell version of ECIS has also been explored, yet its intrinsic capability and limitation remain unclear. In this work, we argue for the fundamental feasibility of impedance spectroscopy to track changes of multiple cellular properties using a noninvasive single-cell approach. While changing individual properties is experimentally prohibitive, we take a simulation approach instead and mimic the corresponding changes using a 3D computational model. From the resultant impedance spectra, we identify the spectroscopic signature characteristic to each property considered herein. Since multiple properties change concurrently in practice, the respective signatures often overlap spectroscopically and become hidden. We further attempt to deconvolve such spectra and reveal the underlying property changes. This work provides the theoretical foundation to inspire experimental validation and adoption of ECIS for multiproperty single-cell measurements.
Collapse
|
37
|
Kamande JW, Lindell MAM, Witek MA, Voorhees PM, Soper SA. Isolation of circulating plasma cells from blood of patients diagnosed with clonal plasma cell disorders using cell selection microfluidics. Integr Biol (Camb) 2018; 10:82-91. [PMID: 29372735 PMCID: PMC5877822 DOI: 10.1039/c7ib00183e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Blood samples from patients with plasma cell disorders were analysed for the presence of circulating plasma cells (CPCs) using a microfluidic device modified with monoclonal anti-CD138 antibodies. CPCs were immuno-phenotyped using a CD38/CD56/CD45 panel and identified in 78% of patients with monoclonal gammopathy of undetermined significance (MGUS), all patients with smouldering and symptomatic multiple myeloma (MM), and none in the controls. The burden of CPCs was higher in patients with symptomatic MM compared with MGUS and smouldering MM (p < 0.05). FISH analysis revealed the presence of chromosome 13 deletions in CPCs that correlated with bone marrow results. Point mutations in KRAS were identified, including different mutations from sub-clones derived from the same patient. The microfluidic assay represents a highly sensitive method for enumerating CPCs and allows for the cytogenetic and molecular characterization of CPCs.
Collapse
Affiliation(s)
- Joyce W Kamande
- Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
38
|
Court CM, Ankeny JS, Sho S, Winograd P, Hou S, Song M, Wainberg ZA, Girgis MD, Graeber TG, Agopian VG, Tseng HR, Tomlinson JS. Circulating Tumor Cells Predict Occult Metastatic Disease and Prognosis in Pancreatic Cancer. Ann Surg Oncol 2018; 25:1000-1008. [PMID: 29442211 DOI: 10.1245/s10434-017-6290-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Occult metastatic tumors, below imaging thresholds, are a limitation of staging systems that rely on cross-sectional imaging alone and are a cause of the routine understaging of pancreatic ductal adenocarcinomas (PDACs). We investigated circulating tumor cells (CTCs) as a preoperative predictor of occult metastatic disease and as a prognostic biomarker for PDAC patients. EXPERIMENTAL DESIGN A total of 126 patients (100 with cancer, 26 with benign disease) were enrolled in our study and CTCs were identified and enumerated from 4 mL of venous blood using the microfluidic NanoVelcro assay. CTC enumeration was correlated with clinicopathologic variables and outcomes following both surgical and systemic therapies. RESULTS CTCs were identified in 78% of PDAC patients and CTC counts correlated with increasing stage (ρ = 0.42, p < 0.001). Of the 53 patients taken for potentially curative surgery, 13 (24.5%) had occult metastatic disease intraoperatively. Patients with occult disease had significantly more CTCs than patients with local disease only (median 7 vs. 1 CTC, p < 0.0001). At a cut-off of three or more CTCs/4 mL, CTCs correctly identified patients with occult metastatic disease preoperatively (area under the receiver operating characteristic curve 0.82, 95% confidence interval (CI) 0.76-0.98, p < 0.0001). CTCs were a univariate predictor of recurrence-free survival following surgery [hazard ratio (HR) 2.36, 95% CI 1.17-4.78, p = 0.017], as well as an independent predictor of overall survival on multivariate analysis (HR 1.38, 95% CI 1.01-1.88, p = 0.040). CONCLUSIONS CTCs show promise as a prognostic biomarker for PDAC patients at all stages of disease being treated both medically and surgically. Furthermore, CTCs demonstrate potential as a preoperative biomarker for identifying patients at high risk of occult metastatic disease.
Collapse
Affiliation(s)
- Colin M Court
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA.,Department of Surgery, Veteran's Health Administration, Los Angeles, CA, USA.,Department of Molecular, Cellular, and Integrative Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jacob S Ankeny
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA.,Department of Surgery, Veteran's Health Administration, Los Angeles, CA, USA
| | - Shonan Sho
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA.,Department of Surgery, Veteran's Health Administration, Los Angeles, CA, USA
| | - Paul Winograd
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA.,Department of Surgery, Veteran's Health Administration, Los Angeles, CA, USA
| | - Shuang Hou
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA.,Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Min Song
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Zev A Wainberg
- Department of Hematology/Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Mark D Girgis
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Thomas G Graeber
- Department of Molecular, Cellular, and Integrative Physiology, University of California Los Angeles, Los Angeles, CA, USA.,Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Vatche G Agopian
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - James S Tomlinson
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA. .,Department of Surgery, Veteran's Health Administration, Los Angeles, CA, USA. .,California NanoSystems Institute (CNSI), University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
39
|
Rauta PR, Hallur PM, Chaubey A. Gold nanoparticle-based rapid detection and isolation of cells using ligand-receptor chemistry. Sci Rep 2018; 8:2893. [PMID: 29440656 PMCID: PMC5811494 DOI: 10.1038/s41598-018-21068-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/22/2018] [Indexed: 01/07/2023] Open
Abstract
Identification and isolation of low-frequency cells of interest from a heterogeneous cell mixture is an important aspect of many diagnostic applications (including enumeration of circulating tumor cells) and is integral to various assays in (cancer) biology. Current techniques typically require expensive instrumentation and are not amenable to high throughput. Here, we demonstrate a simple and effective platform for cell detection and isolation using gold nanoparticles (Au NPs) conjugated with hyaluronic acid (HA) i.e. Au-PEG-HA NPs. The proposed platform exploits ligand-receptor chemistry to detect/isolate cells with high specificity and efficiency. When the Au-PEG-HA NPs come in contact with cells that express CD44 (the receptor for HA), a clear colorimetric change occurs (along with an accompanying SPR peak shift from 521 nm to 559 nm) in the solution due to NPs-cell interaction. This clearly discernible, colorimetric change can be leveraged by point-of-care devices employed in diagnostic applications. Finally, we show that we can successfully isolate viable cells from a heterogeneous cell population (including from human blood samples) with high specificity, which can be used in further downstream applications. The developed NPs-based platform can be a convenient and cost-efficient alternative for diagnostic applications and for cell isolation or sorting in research laboratories.
Collapse
Affiliation(s)
- Pradipta Ranjan Rauta
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research, NH Health City, Bangalore, 560 099, India
| | - Pavan M Hallur
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research, NH Health City, Bangalore, 560 099, India
| | - Aditya Chaubey
- Anti-Cancer Technologies Program, Mazumdar Shaw Center for Translational Research, NH Health City, Bangalore, 560 099, India.
| |
Collapse
|
40
|
Lee TY, Han K, Barrett DO, Park S, Soper SA, Murphy MC. Accurate, predictable, repeatable micro-assembly technology for polymer, microfluidic modules. SENSORS AND ACTUATORS. B, CHEMICAL 2018. [PMID: 29531428 PMCID: PMC5844477 DOI: 10.1016/j.snb.2017.07.189] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
A method for the design, construction, and assembly of modular, polymer-based, microfluidic devices using simple micro-assembly technology was demonstrated to build an integrated fluidic system consisting of vertically stacked modules for carrying out multi-step molecular assays. As an example of the utility of the modular system, point mutation detection using the ligase detection reaction (LDR) following amplification by the polymerase chain reaction (PCR) was carried out. Fluid interconnects and standoffs ensured that temperatures in the vertically stacked reactors were within ± 0.2 C° at the center of the temperature zones and ± 1.1 C° overall. The vertical spacing between modules was confirmed using finite element models (ANSYS, Inc., Canonsburg, PA) to simulate the steady-state temperature distribution for the assembly. Passive alignment structures, including a hemispherical pin-in-hole, a hemispherical pin-in-slot, and a plate-plate lap joint, were developed using screw theory to enable accurate exactly constrained assembly of the microfluidic reactors, cover sheets, and fluid interconnects to facilitate the modular approach. The mean mismatch between the centers of adjacent through holes was 64 ± 7.7 μm, significantly reducing the dead volume necessary to accommodate manufacturing variation. The microfluidic components were easily assembled by hand and the assembly of several different configurations of microfluidic modules for executing the assay was evaluated. Temperatures were measured in the desired range in each reactor. The biochemical performance was comparable to that obtained with benchtop instruments, but took less than 45 min to execute, half the time.
Collapse
Affiliation(s)
- Tae Yoon Lee
- Department of Mechanical & Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Bio-Modular Multi-Scale Systems for Precision Medicine
- Department of Technology Education and Department of Biomedical Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kyudong Han
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Dwhyte O. Barrett
- Department of Mechanical & Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Bio-Modular Multi-Scale Systems for Precision Medicine
| | - Sunggook Park
- Department of Mechanical & Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Bio-Modular Multi-Scale Systems for Precision Medicine
| | - Steven A. Soper
- Department of Mechanical & Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Bio-Modular Multi-Scale Systems for Precision Medicine
- Department of Mechanical Engineering and Department of Chemistry, University of Kansas, Lawrence, KS, USA
| | - Michael C. Murphy
- Department of Mechanical & Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Bio-Modular Multi-Scale Systems for Precision Medicine
- Correspondence: Dr. Michael C. Murphy; , Tel: 1-225-578-5921, Fax: 1-225-578-5924
| |
Collapse
|
41
|
Peptide nanoparticles (PNPs) modified disposable platform for sensitive electrochemical cytosensing of DLD-1 cancer cells. Biosens Bioelectron 2017; 104:50-57. [PMID: 29306761 DOI: 10.1016/j.bios.2017.12.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/21/2017] [Accepted: 12/23/2017] [Indexed: 01/19/2023]
Abstract
A novel diphenylalaninamid (FFA) based peptide nanoparticles (PNPs) modified pencil graphite electrodes (PGEs) for construction of electrochemical cytosensor was demonstrated for the first time in this study. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) images revealed the spherical nanostructure of the synthesized FFA based PNPs while attenuated total reflectance-fourier transform infrared (ATR-FTIR) spectra provided information about the structure and conformation of proteins in their structure. Self-assembly of PNPs on PGE surface and adhesion of DLD-1 cancer cells on this surface was also characterized by electrochemical measurements. PNP/PGEs acted as a sensitive platform for simple and rapid quantification of low concentration of DLD-1 cancer cells in early diagnosis using the electrochemical impedance method (EIS). The offered cytosensor demonstrated outstanding performance for the detection of DLD-1 cells by the EIS method. The impedance of electronic transduction was associated with the amount of the immobilized cells ranging from 2 × 102 to 2.0 × 105 cellsmL-1 with a limit of detection of 100 cellsmL-1. The efficient performance of the cytosensor was attributed to the well-defined nanostructure and biocompability of PNPs on the substrate.
Collapse
|
42
|
Abstract
The majority of cancer-related deaths result from metastasis, the process by which cancer cells escape the primary tumor site and enter into the blood circulation in order to disseminate to secondary locations throughout the body. Tumor cells found within the circulation are referred to as circulating tumor cells (CTCs), and their detection and enumeration correlate with poor prognosis. The epithelial-to-mesenchymal transition (EMT) is a dynamic process that imparts epithelial cells with mesenchymal-like properties, thus facilitating tumor cell dissemination and contributing to metastasis. However, EMT also results in the downregulation of various epithelial proteins typically utilized by CTC technologies for enrichment and detection of these rare cells, resulting in reduced detection of some CTCs, potentially those with a more metastatic phenotype. In addition to the current clinical role of CTCs as a prognostic biomarker, they also have potential as a predictive biomarker via CTC characterization. However, CTC characterization is complicated by the unknown biological significance of CTCs possessing an EMT-like phenotype, and the ability to capture and understand this CTC subpopulation is an essential step in the utilization of CTCs for patient management. This chapter will review the process of EMT and its contribution to metastasis; discusses current and future clinical applications of CTCs; and describes both traditional and novel methods for CTC enrichment, detection, and characterization with a specific focus on CTCs with an EMT phenotype.
Collapse
|
43
|
Keane MG, Shah A, Pereira SP, Joshi D. Novel biomarkers and endoscopic techniques for diagnosing pancreaticobiliary malignancy. F1000Res 2017; 6:1643. [PMID: 28944047 PMCID: PMC5585877 DOI: 10.12688/f1000research.11371.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/06/2017] [Indexed: 12/12/2022] Open
Abstract
The UK incidence of pancreatic ductal adenocarcinoma is 9 per 100,000 population, and biliary tract cancer occurs at a rate of 1–2 per 100,000. The incidence of both cancers is increasing annually and these tumours continue to be diagnosed late and at an advanced stage, limiting options for curative treatment. Population-based screening programmes do not exist for these cancers, and diagnosis currently is dependent on symptom recognition, but often symptoms are not present until the disease is advanced. Recently, a number of promising blood and urine biomarkers have been described for pancreaticobiliary malignancy and are summarised in this review. Novel endoscopic techniques such as single-operator cholangioscopy and confocal endomicroscopy have been used in some centres to enhance standard endoscopic diagnostic techniques and are also evaluated in this review.
Collapse
Affiliation(s)
| | - Amar Shah
- Institute of Liver Studies, King's College Hospital, London, UK
| | - Stephen P Pereira
- UCL Institute for Liver and Digestive Health, Royal Free Campus, London, UK
| | - Deepak Joshi
- Institute of Liver Studies, King's College Hospital, London, UK
| |
Collapse
|
44
|
Gencturk E, Mutlu S, Ulgen KO. Advances in microfluidic devices made from thermoplastics used in cell biology and analyses. BIOMICROFLUIDICS 2017; 11:051502. [PMID: 29152025 PMCID: PMC5654984 DOI: 10.1063/1.4998604] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/11/2017] [Indexed: 05/10/2023]
Abstract
Silicon and glass were the main fabrication materials of microfluidic devices, however, plastics are on the rise in the past few years. Thermoplastic materials have recently been used to fabricate microfluidic platforms to perform experiments on cellular studies or environmental monitoring, with low cost disposable devices. This review describes the present state of the development and applications of microfluidic systems used in cell biology and analyses since the year 2000. Cultivation, separation/isolation, detection and analysis, and reaction studies are extensively discussed, considering only microorganisms (bacteria, yeast, fungi, zebra fish, etc.) and mammalian cell related studies in the microfluidic platforms. The advantages/disadvantages, fabrication methods, dimensions, and the purpose of creating the desired system are explained in detail. An important conclusion of this review is that these microfluidic platforms are still open for research and development, and solutions need to be found for each case separately.
Collapse
Affiliation(s)
- Elif Gencturk
- Department of Chemical Engineering, Biosystems Engineering Laboratory, Bogazici University, 34342 Istanbul, Turkey
| | - Senol Mutlu
- Department of Electrical and Electronics Engineering, BUMEMS Laboratory, Bogazici University, 34342 Istanbul, Turkey
| | - Kutlu O Ulgen
- Department of Chemical Engineering, Biosystems Engineering Laboratory, Bogazici University, 34342 Istanbul, Turkey
| |
Collapse
|
45
|
Witek MA, Aufforth RD, Wang H, Kamande JW, Jackson JM, Pullagurla SR, Hupert ML, Usary J, Wysham WZ, Hilliard D, Montgomery S, Bae-Jump V, Carey LA, Gehrig PA, Milowsky MI, Perou CM, Soper JT, Whang YE, Yeh JJ, Martin G, Soper SA. Discrete microfluidics for the isolation of circulating tumor cell subpopulations targeting fibroblast activation protein alpha and epithelial cell adhesion molecule. NPJ Precis Oncol 2017; 1. [PMID: 29657983 PMCID: PMC5871807 DOI: 10.1038/s41698-017-0028-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Circulating tumor cells consist of phenotypically distinct subpopulations that originate from the tumor microenvironment. We report a circulating tumor cell dual selection assay that uses discrete microfluidics to select circulating tumor cell subpopulations from a single blood sample; circulating tumor cells expressing the established marker epithelial cell adhesion molecule and a new marker, fibroblast activation protein alpha, were evaluated. Both circulating tumor cell subpopulations were detected in metastatic ovarian, colorectal, prostate, breast, and pancreatic cancer patients and 90% of the isolated circulating tumor cells did not co-express both antigens. Clinical sensitivities of 100% showed substantial improvement compared to epithelial cell adhesion molecule selection alone. Owing to high purity (>80%) of the selected circulating tumor cells, molecular analysis of both circulating tumor cell subpopulations was carried out in bulk, including next generation sequencing, mutation analysis, and gene expression. Results suggested fibroblast activation protein alpha and epithelial cell adhesion molecule circulating tumor cells are distinct subpopulations and the use of these in concert can provide information needed to navigate through cancer disease management challenges.
Collapse
Affiliation(s)
- Małgorzata A Witek
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA.,Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA.,Department of Biomedical Engineering, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rachel D Aufforth
- Department of Surgery, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Wang
- Department of Biomedical Engineering, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joyce W Kamande
- Department of Biomedical Engineering, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joshua M Jackson
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA.,Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA
| | - Swathi R Pullagurla
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA.,Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, KS 66047, USA
| | - Mateusz L Hupert
- Department of Biomedical Engineering, The University of North Carolina, Chapel Hill, NC 27599, USA.,BioFluidica, Inc., c/o Carolina Kick-Start, 321 Bondurant Hall, Chapel Hill NC27599, USA
| | - Jerry Usary
- Department of Genetics, The University of North Carolina, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Weiya Z Wysham
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, UNC-Chapel Hill, NC 27599, USA
| | - Dawud Hilliard
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Animal Histopathology Core, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Stephanie Montgomery
- Animal Histopathology Core, The University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Victoria Bae-Jump
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, UNC-Chapel Hill, NC 27599, USA
| | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Medicine, Division of Hematology and Oncology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Paola A Gehrig
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, UNC-Chapel Hill, NC 27599, USA
| | - Matthew I Milowsky
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - John T Soper
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, UNC-Chapel Hill, NC 27599, USA
| | - Young E Whang
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jen Jen Yeh
- Department of Surgery, The University of North Carolina, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Pharmacology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Steven A Soper
- BioEngineering Program, The University of Kansas, Lawrence, KS 66047, USA.,Department of Mechanical Engineering, The University of Kansas, Lawrence, KS 66047, USA.,Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
46
|
Jackson JM, Witek MA, Kamande JW, Soper SA. Materials and microfluidics: enabling the efficient isolation and analysis of circulating tumour cells. Chem Soc Rev 2017; 46:4245-4280. [PMID: 28632258 PMCID: PMC5576189 DOI: 10.1039/c7cs00016b] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We present a critical review of microfluidic technologies and material effects on the analyses of circulating tumour cells (CTCs) selected from the peripheral blood of cancer patients. CTCs are a minimally invasive source of clinical information that can be used to prognose patient outcome, monitor minimal residual disease, assess tumour resistance to therapeutic agents, and potentially screen individuals for the early diagnosis of cancer. The performance of CTC isolation technologies depends on microfluidic architectures, the underlying principles of isolation, and the choice of materials. We present a critical review of the fundamental principles used in these technologies and discuss their performance. We also give context to how CTC isolation technologies enable downstream analysis of selected CTCs in terms of detecting genetic mutations and gene expression that could be used to gain information that may affect patient outcome.
Collapse
|
47
|
Ankeny JS, Court CM, Hou S, Li Q, Song M, Wu D, Chen JF, Lee T, Lin M, Sho S, Rochefort MM, Girgis MD, Yao J, Wainberg ZA, Muthusamy VR, Watson RR, Donahue TR, Hines OJ, Reber HA, Graeber TG, Tseng HR, Tomlinson JS. Circulating tumour cells as a biomarker for diagnosis and staging in pancreatic cancer. Br J Cancer 2017; 114:1367-75. [PMID: 27300108 PMCID: PMC4984454 DOI: 10.1038/bjc.2016.121] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/18/2016] [Accepted: 04/08/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Current diagnosis and staging of pancreatic ductal adenocarcinoma (PDAC) has important limitations and better biomarkers are needed to guide initial therapy. We investigated the performance of circulating tumour cells (CTCs) as an adjunctive biomarker at the time of disease presentation. METHODS Venous blood (VB) was collected prospectively from 100 consecutive, pre-treatment patients with PDAC. Utilising the microfluidic NanoVelcro CTC chip, samples were evaluated for the presence and number of CTCs. KRAS mutation analysis was used to compare the CTCs with primary tumour tissue. CTC enumeration data was then evaluated as a diagnostic and staging biomarker in the setting of PDAC. RESULTS We found 100% concordance for KRAS mutation subtype between primary tumour and CTCs in all five patients tested. Evaluation of CTCs as a diagnostic revealed the presence of CTCs in 54/72 patients with confirmed PDAC (sensitivity=75.0%, specificity=96.4%, area under the curve (AUROC)=0.867, 95% CI=0.798-0.935, and P<0.001). Furthermore, a cut-off of ⩾3 CTCs in 4 ml VB was able to discriminate between local/regional and metastatic disease (AUROC=0.885; 95% CI=0.800-0.969; and P<0.001). CONCLUSION CTCs appear to function well as a biomarker for diagnosis and staging in PDAC.
Collapse
Affiliation(s)
- J S Ankeny
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,Veteran's Health Administration, Greater Los Angeles, Department of Surgery, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA
| | - C M Court
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,Veteran's Health Administration, Greater Los Angeles, Department of Surgery, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA
| | - S Hou
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - Q Li
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - M Song
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - D Wu
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - J F Chen
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - T Lee
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, CA 90095, USA
| | - M Lin
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - S Sho
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,Veteran's Health Administration, Greater Los Angeles, Department of Surgery, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA
| | - M M Rochefort
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| | - M D Girgis
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| | - J Yao
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - Z A Wainberg
- UCLA Center for Pancreatic Diseases, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,Department of Hematology/Oncology, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| | - V R Muthusamy
- UCLA Center for Pancreatic Diseases, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,Department of Gastroenterology, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| | - R R Watson
- UCLA Center for Pancreatic Diseases, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,Department of Gastroenterology, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| | - T R Donahue
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,UCLA Center for Pancreatic Diseases, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| | - O J Hines
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,UCLA Center for Pancreatic Diseases, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| | - H A Reber
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,UCLA Center for Pancreatic Diseases, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| | - T G Graeber
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - H R Tseng
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, California NanoSystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095-1770, USA
| | - J S Tomlinson
- Department of Surgery, University of California Los Angeles, 575 Westwood Plaza, Los Angeles, CA 90095, USA.,Veteran's Health Administration, Greater Los Angeles, Department of Surgery, 11301 Wilshire Boulevard, Los Angeles, CA 90073, USA.,UCLA Center for Pancreatic Diseases, 575 Westwood Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
48
|
Rawal S, Yang YP, Cote R, Agarwal A. Identification and Quantitation of Circulating Tumor Cells. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2017; 10:321-343. [PMID: 28301753 DOI: 10.1146/annurev-anchem-061516-045405] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Circulating tumor cells (CTCs) are shed from the primary tumor into the circulatory system and act as seeds that initiate cancer metastasis to distant sites. CTC enumeration has been shown to have a significant prognostic value as a surrogate marker in various cancers. The widespread clinical utility of CTC tests, however, is still limited due to the inherent rarity and heterogeneity of CTCs, which necessitate robust techniques for their efficient enrichment and detection. Significant recent advances have resulted in technologies with the ability to improve yield and purity of CTC enrichment as well as detection sensitivity. Current efforts are largely focused on the translation and standardization of assays to fully realize the clinical utility of CTCs. In this review, we aim to provide a comprehensive overview of CTC enrichment and detection techniques with an emphasis on novel approaches for rapid quantification of CTCs.
Collapse
Affiliation(s)
- Siddarth Rawal
- Department of Pathology, DJTMF Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida 33146
| | - Yu-Ping Yang
- Department of Pathology, DJTMF Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida 33146
- Department of Biochemistry and Molecular Biology, University of Miami, Coral Gables, Florida 33146
| | - Richard Cote
- Department of Pathology, DJTMF Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida 33146
- Department of Biochemistry and Molecular Biology, University of Miami, Coral Gables, Florida 33146
| | - Ashutosh Agarwal
- Department of Pathology, DJTMF Biomedical Nanotechnology Institute, University of Miami, Coral Gables, Florida 33146
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida 33146;
| |
Collapse
|
49
|
Pang TCY, Xu Z, Pothula S, Becker T, Goldstein D, Pirola RC, Wilson JS, Apte MV. Circulating pancreatic stellate (stromal) cells in pancreatic cancer—a fertile area for novel research. Carcinogenesis 2017; 38:588-591. [PMID: 28379317 DOI: 10.1093/carcin/bgx030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023] Open
|
50
|
Dong Z, Ahrens CC, Yu D, Ding Z, Lim H, Li W. Cell Isolation and Recovery Using Hollow Glass Microspheres Coated with Nanolayered Films for Applications in Resource-Limited Settings. ACS APPLIED MATERIALS & INTERFACES 2017; 9:15265-15273. [PMID: 28414907 DOI: 10.1021/acsami.7b02197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Established cell isolation and purification techniques such as fluorescence-activated cell sorting (FACS), isolation through magnetic micro/nanoparticles, and recovery via microfluidic devices have limited application as disposable technologies appropriate for point-of-care use in remote areas where lab equipment as well as electrical, magnetic, and optical sources are restricted. We report a simple yet effective method for cell isolation and recovery that requires neither specialized lab equipment nor any form of power source. Specifically, self-floating hollow glass microspheres were coated with an enzymatically degradable nanolayered film and conjugated with antibodies to allow both fast capture and release of subpopulations of cells from a cell mixture. Targeted cells were captured by the microspheres and allowed to float to the top of the hosting liquid, thereby isolating targeted cells. To minimize nonspecific adhesion of untargeted cells and to enhance the purity of the isolated cell population, an antifouling polymer brush layer was grafted onto the nanolayered film. Using the EpCAM-expressing cancer cell line PC-3 in blood as a model system, we have demonstrated the isolation and recovery of cancer cells without compromising cell viability or proliferative potential. The whole process takes less than 1 h. To support the rational extension of this platform technology, we introduce extensive characterization of the critical design parameters: film formation and degradation, grafting with a poly(ethylene glycol) (PEG) sheath, and introducing functional antibodies. Our approach is expected to overcome practical hurdles and provide viable targeted cells for downstream analyses in resource-limited settings.
Collapse
Affiliation(s)
- Ziye Dong
- Department of Chemical Engineering, Texas Tech University , Lubbock, Texas 79409, United States
| | - Caroline C Ahrens
- Department of Chemical Engineering, Texas Tech University , Lubbock, Texas 79409, United States
| | - Dan Yu
- Department of Critical Care Medicine, People's Hospital of Zhengzhou University Zhengzhou, China 450003
| | - Zhenya Ding
- Department of Chemical Engineering, Texas Tech University , Lubbock, Texas 79409, United States
| | - HyunTaek Lim
- Department of Chemical Engineering, Texas Tech University , Lubbock, Texas 79409, United States
| | - Wei Li
- Department of Chemical Engineering, Texas Tech University , Lubbock, Texas 79409, United States
| |
Collapse
|