GET THE APP

Development of a Microfluidic-Based Platform for Rapid Detection of Circulating Tumor Cells
..

Journal of Bioanalysis & Biomedicine

ISSN: 1948-593X

Open Access

Short Communication - (2023) Volume 15, Issue 4

Development of a Microfluidic-Based Platform for Rapid Detection of Circulating Tumor Cells

Jianwen Yang*
*Correspondence: Jianwen Yang, Department of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China, Email:
Department of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China

Received: 01-Aug-2023, Manuscript No. jbabm-23-112175; Editor assigned: 03-Aug-2023, Pre QC No. P-112175; Reviewed: 17-Aug-2023, QC No. Q-112175; Revised: 23-Aug-2023, Manuscript No. R-112175; Published: 31-Aug-2023 , DOI: 10.37421/1948-593X.2023.15.391
Citation: Yang, Jianwen. Development of a Microfluidic-Based Platform for Rapid Detection of Circulating Tumor Cells.� J Bioanal Biomed 15 (2023): 391.
Copyright: © 2023 Yang J. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Introduction

Circulating Tumor Cells (CTCs) have emerged as a valuable resource in the field of oncology. These rare cancer cells shed from primary tumors and enter the bloodstream, carrying vital information about tumor progression, metastasis, and treatment response. The detection and characterization of CTCs hold immense potential for early cancer diagnosis, prognosis, and the development of personalized treatment strategies. However, the effective isolation and analysis of CTCs from patient blood samples present considerable technical challenges.

Traditional methods for CTC detection often face limitations related to sensitivity, specificity, and the complexity of blood samples. To address these challenges, a novel microfluidic-based platform has been developed, aiming to streamline and enhance the detection of CTCs. Microfluidics, with its ability to manipulate fluids at the microscale, offers unique advantages for CTC isolation, allowing for precise control over sample processing and the integration of biomarker-specific capture strategies. This platform leverages advanced microfluidic design, selective capture techniques, and high-resolution imaging to enable the rapid and efficient detection of CTCs [1].

Description

The development of the microfluidic-based platform for CTC detection is centered around several key components and functionalities:

Microfluidic design: The platform's foundation lies in the intricate design of microfluidic channels, chambers, and surfaces. These components are engineered to enable the efficient manipulation and isolation of CTCs from complex blood samples. Microfluidic principles, including laminar flow, inertial focusing, and surface modifications, are harnessed to facilitate CTC capture and separation [2].

Biomarker-specific capture strategies: Specificity is paramount in CTC detection. The platform integrates biomarker-specific capture strategies, typically involving the immobilization of antibodies or aptamers targeting cancer-specific markers on microfluidic surfaces. This selective binding ensures the preferential capture of CTCs, even in the presence of a high background of normal blood cells [3].

High-resolution imaging: To characterize and identify CTCs, highresolution imaging techniques are incorporated into the platform. These techniques, such as fluorescence microscopy or other imaging modalities, provide detailed information about the captured CTCs, including their morphological features, phenotypic markers, and genetic profiles [4].

Clinical applications: The microfluidic-based platform holds significant promise for clinical applications. It has the potential to revolutionize cancer diagnostics by enabling the early detection of CTCs, which can serve as prognostic and predictive biomarkers. Moreover, it facilitates real-time monitoring of minimal residual disease, allowing clinicians to assess treatment response and disease progression. In the context of personalized medicine, the platform offers the opportunity to tailor treatment strategies based on individual CTC profiles, optimizing therapeutic outcomes [5].

Conclusion

In conclusion, the development of a microfluidic-based platform for the rapid detection of circulating tumor cells represents a significant advancement in the field of cancer diagnostics and personalized medicine. This innovative technology harnesses the precision and versatility of microfluidics to address the challenges associated with CTC isolation and analysis. By enabling the efficient capture, characterization, and enumeration of CTCs, this platform holds the potential to reshape the landscape of cancer management.

As we continue to refine the capabilities of microfluidic-based CTC detection, it is anticipated that this technology will become an indispensable tool in clinical oncology. Its ability to provide valuable insights into cancer dynamics, treatment response, and disease progression has the potential to improve patient outcomes, inform treatment decisions, and contribute to our understanding of cancer biology. In the era of precision medicine, the development of such innovative platforms brings us one step closer to tailored, patient-specific cancer therapies.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Nagrath, Sunitha, Lecia V. Sequist, Shyamala Maheswaran and Matthew R. Smith, et al. "Isolation of rare circulating tumour cells in cancer patients by microchip technology." Nat 450 (2007): 1235-1239.
  2. Google Scholar, Crossref, Indexed at

  3. Hu, Xuejia, Daoming Zhu, Ming Chen and Yi Yang, et al. "Precise and non-invasive circulating tumor cell isolation based on optical force using homologous erythrocyte binding." Lab Chip 19 (2019): 2549-2556.
  4. Google Scholar, Crossref, Indexed at

  5. Parker, Stephen G., Ying Yang, Simone Ciampi and J. Justin Gooding, et al. "A photoelectrochemical platform for the capture and release of rare single cells." Nat Commun 9 (2018): 2288.
  6. Google Scholar, Crossref, Indexed at

  7. Hu, Yuhao, Wanyi Tang, Pan Cheng and Hao He, et al. "Monitoring circulating tumor cells in vivo by a confocal microscopy system." Cytometry A 95 (2019): 657-663.
  8. Google Scholar, Crossref, Indexed at

  9. Chang, Chun-Li, Wanfeng Huang, Shadia I. Jalal and Cagri A. Savran, et al. "Circulating tumor cell detection using a parallel flow micro-aperture chip system." Lab Chip 15 (2015): 1677-1688.
  10. Google Scholar, Crossref

Google Scholar citation report
Citations: 3099

Journal of Bioanalysis & Biomedicine received 3099 citations as per Google Scholar report

Journal of Bioanalysis & Biomedicine peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward
https://www.olimpbase.org/1937/