1
|
Ou X, Chen P, Liu BF. Liquid Biopsy on Microfluidics: From Existing Endogenous to Emerging Exogenous Biomarkers Analysis. Anal Chem 2025; 97:8625-8640. [PMID: 40247704 DOI: 10.1021/acs.analchem.4c05407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Liquid biopsy is an appealing approach for early diagnosis and assessment of treatment efficacy in cancer. Typically, liquid biopsy involves the detection of endogenous biomarkers, including circulating tumor cells (CTCs), extracellular vesicles (EVs), circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA), and proteins. The levels of these endogenous biomarkers are higher in cancer patients compared to those in healthy individuals. However, the clinical application of liquid biopsy using endogenous biomarker analysis faces challenges due to its low abundance and poor stability in circulation. Recently, a promising strategy involving the engineering of exogenous probes has been developed to overcome these limitations. These exogenous probes are activated within the tumor microenvironment, generating distinct exogenous markers that can be easily distinguished from background biological signals. Alternatively, these exogenous probes can be labeled with intrinsic endogenous biomarkers in vivo and detected in vitro after metabolic processes. In this review, we primarily focus on microfluidic-based liquid biopsy techniques that allow for the transition from analyzing existing endogenous biomarkers to emerging exogenous ones. First, we introduce common endogenous biomarkers, as well as synthetic exogenous ones. Next, we discuss recent advancements in microfluidic-based liquid biopsy techniques for analyzing both existing endogenous and emerging exogenous biomarkers. Lastly, we provide insights into future directions for liquid biopsy on microfluidic systems.
Collapse
Affiliation(s)
- Xiaowen Ou
- Hubei Key Laboratory of Purification and Application of Plant Anti-Cancer Active Ingredients, Department of Chemistry and Life Science, Hubei University of Education, Wuhan, 430205, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
2
|
Bae J, Ryu Y, Choi J, Jeong MS, Lee CK, Hong CA, Ji S, Heo S, Kim S, Jo SM, Lee JJ. Exploring multivalency-driven sensitivity modulation for optimization and fine-tuning of avidity-based biosensors. Biosens Bioelectron 2025; 271:116989. [PMID: 39615224 DOI: 10.1016/j.bios.2024.116989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 01/06/2025]
Abstract
The development of multivalent nanoprobes has garnered considerable interest due to their enhanced sensitivity and precision in diagnosing and monitoring diverse diseases. Despite significant advances in nanobiotechnology, the optimal density of binding motifs to maximize the diagnostic efficacy of biosensors remains incompletely understood. Herein, we investigate the influence of multivalency in the functional performance of avidity-based biosensors. By post-translationally modifying the surface of M13 bacteriophages, we achieved multivalent displays of epidermal growth factor receptor-specific repebodies and enhanced green fluorescent proteins in a controlled configuration. Through an array of phage-based nanoprobes, our findings reveal an inverted U-shaped correlation between the degree of multivalency and biosensor sensitivity. Briefly, excessive multivalency negatively impacts analytical performance, likely due to irreversible target depletion, while fine-tuned multivalency significantly enhances antigen recognition and signal amplification. Additionally, the advantages of avidity-demonstrated as improved targeting specificity and sensing capability-are especially prominent in nanoprobes incorporating low-affinity repebodies. This work underscores the critical role of multivalency optimization in biosensor design, providing valuable insights for the development of highly sensitive and reliable diagnostic tools in clinical settings.
Collapse
Affiliation(s)
- Juhyeon Bae
- Department of Biochemistry, Kangwon National University, Chuncheon, 24341, South Korea
| | - Yiseul Ryu
- Institute of Life Sciences (ILS), Kangwon National University, Chuncheon, 24341, South Korea
| | - Junho Choi
- Department of Biochemistry, Kangwon National University, Chuncheon, 24341, South Korea
| | - Myeong Seon Jeong
- Center for Bio-imaging & Translational Research, Korea Basic Science Institute (KBSI), Cheongju, 28119, South Korea
| | - Cheol-Ki Lee
- Department of Biochemistry, Kangwon National University, Chuncheon, 24341, South Korea
| | - Cheol Am Hong
- Department of Chemistry, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Seoha Ji
- Department of Biochemistry, Kangwon National University, Chuncheon, 24341, South Korea
| | - Seungnyeong Heo
- Department of Biochemistry, Kangwon National University, Chuncheon, 24341, South Korea
| | - Seongjoon Kim
- Department of Biochemistry, Kangwon National University, Chuncheon, 24341, South Korea
| | - Seong-Min Jo
- Department of Biomaterial Science, Pusan National University, Miryang, 50463, South Korea
| | - Joong-Jae Lee
- Department of Biochemistry, Kangwon National University, Chuncheon, 24341, South Korea; Institute of Life Sciences (ILS), Kangwon National University, Chuncheon, 24341, South Korea; Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon, 24341, South Korea.
| |
Collapse
|
3
|
Moon GY, Dalkiran B, Park HS, Shin D, Son C, Choi JH, Bang S, Lee H, Doh I, Kim DH, Jeong WJ, Bu J. Dual Biomarker Strategies for Liquid Biopsy: Integrating Circulating Tumor Cells and Circulating Tumor DNA for Enhanced Tumor Monitoring. BIOSENSORS 2025; 15:74. [PMID: 39996976 PMCID: PMC11852634 DOI: 10.3390/bios15020074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/26/2025]
Abstract
The liquid biopsy has gained significant attention in cancer diagnostics, with circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) being recognized as key biomarkers for tumor detection and monitoring. However, each biomarker possesses inherent limitations that restrict its standalone clinical utility, such as the rarity and heterogeneity of CTCs and the variable sensitivity and specificity of ctDNA assays. This highlights the necessity of integrating both biomarkers to maximize diagnostic and prognostic potential, offering a more comprehensive understanding of the tumor biology and therapeutic response. In this review, we summarize clinical studies that have explored the combined analysis of CTCs and ctDNA as biomarkers, providing insights into their synergistic value in diverse tumor types. Specifically, this paper examines the individual advantages and limitations of CTCs and ctDNA, details the findings of combined biomarker studies across various cancers, highlights the benefits of dual biomarker approaches over single-biomarker strategies, and discusses future prospects for advancing personalized oncology through liquid biopsies. By offering a comprehensive overview of clinical studies combining CTCs and ctDNA, this review serves as a guideline for researchers and clinicians aiming to enhance biomarker-based strategies in oncology and informs biosensor design for improved biomarker detection.
Collapse
Affiliation(s)
- Ga Young Moon
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Basak Dalkiran
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Hyun Sung Park
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Dongjun Shin
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Chaeyeon Son
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Jung Hyun Choi
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Seha Bang
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Hosu Lee
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
| | - Il Doh
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Dong Hyung Kim
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, 267 Gajeongno, Yuseong-gu, Daejeon 34113, Republic of Korea; (I.D.); (D.H.K.)
| | - Woo-jin Jeong
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Jiyoon Bu
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (G.Y.M.); (B.D.); (H.S.P.); (D.S.); (C.S.); (J.H.C.); (S.B.); (H.L.)
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Biohybrid Systems Research Center, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
4
|
Bahmaie N, Ozensoy Guler O, Simsek E. A revolutionary era in advancing precision immuno-oncology; role of circulating tumor cells. THE JOURNAL OF LIQUID BIOPSY 2024; 6:100169. [PMID: 40027303 PMCID: PMC11863822 DOI: 10.1016/j.jlb.2024.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 03/05/2025]
Abstract
Despite a substantial progress in the development of strategies against cancer, cancer still remains a major global health issue due to a high recurrence rate, and severe side effects, leading basic medical scientists and clinical specialists toward more efficient diagnostics, prognostics, and therapeutics. Therefore, there is an imperative need for a comprehensive understanding on the cellular immunopathophysiology involved in the tumor microenvironment. In addition, results from a wide range of studies depicted that an aberration in the cellular mechanisms and immunopathophysiological interactions like Circulating Tumor Cells (CTCs) plays an indispensable role in the metastasis and tumor progression, revolutionizing cancer management by offering non-invasive detection methods and a real-time monitoring of tumor dynamics. Moreover, CTCs can clarify the tumor heterogeneity and the evolution of resistance mechanisms, aiding in the early detection of tumors and informing personalized treatment strategies. An increase in CTCs count can be associated with a worsened cancer prognosis, providing promising biomarkers for tumor phenotyping, tumor spreading or relapse, and monitoring the treatment response in patients with cancer. Hence, this systematic review aims to highlight the diagnostic, prognostic, and therapeutic potentials of CTCs, necessitating further investigations and an interdisciplinary collaboration among basic medical scientists and oncologists to address the current gaps in the strategies of cancer management, precisely improving patient-care and optimized clinical outcomes.
Collapse
Affiliation(s)
- Nazila Bahmaie
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University (AYBU), Turkey
| | - Ozen Ozensoy Guler
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University (AYBU), Turkey
| | - Ender Simsek
- Department of Medical Biology, Faculty of Medicine, Ankara Yildirim Beyazit University (AYBU), Turkey
| |
Collapse
|
5
|
Li J, Luo P, Liu S, Fu M, Lin A, Liu Y, He Z, Qiao K, Fang Y, Qu L, Yang K, Wang K, Wang L, Jiang A. Effective strategies to enhance the diagnosis and treatment of RCC: The application of biocompatible materials. Mater Today Bio 2024; 27:101149. [PMID: 39100279 PMCID: PMC11296058 DOI: 10.1016/j.mtbio.2024.101149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 08/06/2024] Open
Abstract
Renal cell carcinoma (RCC) is recognized as one of the three primary malignant tumors affecting the urinary system, posing a significant risk to human health and life. Despite advancements in understanding RCC, challenges persist in its diagnosis and treatment, particularly in early detection and diagnosis due to issues of low specificity and sensitivity. Consequently, there is an urgent need for the development of effective strategies to enhance diagnostic accuracy and treatment outcomes for RCC. In recent years, with the extensive research on materials for applications in the biomedical field, some materials have been identified as promising for clinical applications, e.g., in the diagnosis and treatment of many tumors, including RCC. Herein, we summarize the latest materials that are being studied and have been applied in the early diagnosis and treatment of RCC. While focusing on their adjuvant effects, we also discuss their technical principles and safety, thus highlighting the value and potential of their application. In addition, we also discuss the limitations of the application of these materials and possible future directions, providing new insights for improving RCC diagnosis and treatment.
Collapse
Affiliation(s)
- Jinxin Li
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Shiyang Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Meiling Fu
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361101, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Ying Liu
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Ziwei He
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Kun Qiao
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Le Qu
- Department of Urology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, 305 East Zhongshan Road, Nanjing, 210000, China
| | - Kaidi Yang
- Department of Oncology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, Hainan, 572000, China
- Department of Oncology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Kunpeng Wang
- Department of Urology, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, 222061, China
- Department of Urology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The first People's Hospital of Lianyungang, 222061, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| |
Collapse
|
6
|
Dong Z, Wang Y, Xu G, Liu B, Wang Y, Reboud J, Jajesniak P, Yan S, Ma P, Liu F, Zhou Y, Jin Z, Yang K, Huang Z, Zhuo M, Jia B, Fang J, Zhang P, Wu N, Yang M, Cooper JM, Chang L. Genetic and phenotypic profiling of single living circulating tumor cells from patients with microfluidics. Proc Natl Acad Sci U S A 2024; 121:e2315168121. [PMID: 38683997 PMCID: PMC11087790 DOI: 10.1073/pnas.2315168121] [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: 09/11/2023] [Accepted: 03/08/2024] [Indexed: 05/02/2024] Open
Abstract
Accurate prediction of the efficacy of immunotherapy for cancer patients through the characterization of both genetic and phenotypic heterogeneity in individual patient cells holds great promise in informing targeted treatments, and ultimately in improving care pathways and clinical outcomes. Here, we describe the nanoplatform for interrogating living cell host-gene and (micro-)environment (NICHE) relationships, that integrates micro- and nanofluidics to enable highly efficient capture of circulating tumor cells (CTCs) from blood samples. The platform uses a unique nanopore-enhanced electrodelivery system that efficiently and rapidly integrates stable multichannel fluorescence probes into living CTCs for in situ quantification of target gene expression, while on-chip coculturing of CTCs with immune cells allows for the real-time correlative quantification of their phenotypic heterogeneities in response to immune checkpoint inhibitors (ICI). The NICHE microfluidic device provides a unique ability to perform both gene expression and phenotypic analysis on the same single cells in situ, allowing us to generate a predictive index for screening patients who could benefit from ICI. This index, which simultaneously integrates the heterogeneity of single cellular responses for both gene expression and phenotype, was validated by clinically tracing 80 non-small cell lung cancer patients, demonstrating significantly higher AUC (area under the curve) (0.906) than current clinical reference for immunotherapy prediction.
Collapse
Affiliation(s)
- Zaizai Dong
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
- School of Engineering Medicine, Beihang University, Beijing100191, China
| | - Yusen Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
| | - Gaolian Xu
- Shanghai Sci-Tech InnoCenter for Infection and Immunity, Shanghai200438, China
| | - Bing Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing100142, China
| | - Yang Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
- School of Engineering Medicine, Beihang University, Beijing100191, China
| | - Julien Reboud
- Division of Biomedical Engineering, University of Glasgow, G12 8LTGlasgow, United Kingdom
| | - Pawel Jajesniak
- Division of Biomedical Engineering, University of Glasgow, G12 8LTGlasgow, United Kingdom
| | - Shi Yan
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing100142, China
| | - Pingchuan Ma
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
| | - Feng Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
| | - Yuhao Zhou
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
| | - Zhiyuan Jin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
| | - Kuan Yang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
| | - Zhaocun Huang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital and Institute, Beijing100142, China
| | - Bo Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital and Institute, Beijing100142, China
| | - Jian Fang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital and Institute, Beijing100142, China
| | - Panpan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital and Institute, Beijing100142, China
| | - Nan Wu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing100142, China
| | - Mingzhu Yang
- Beijing Research Institute of Mechanical Equipment, Beijing100143, China
| | - Jonathan M. Cooper
- Division of Biomedical Engineering, University of Glasgow, G12 8LTGlasgow, United Kingdom
| | - Lingqian Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing100191, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei230032, China
| |
Collapse
|
7
|
Hazra RS, Kale N, Boyle C, Molina KB, D'Souza A, Aland G, Jiang L, Chaturvedi P, Ghosh S, Mallik S, Khandare J, Quadir M. Magnetically-activated, nanostructured cellulose for efficient capture of circulating tumor cells from the blood sample of head and neck cancer patients. Carbohydr Polym 2024; 323:121418. [PMID: 37940250 DOI: 10.1016/j.carbpol.2023.121418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 11/10/2023]
Abstract
In this report, the relative efficiency of cellulose nanocrystals (CNCs) and nanofibers (CNFs) to capture circulating tumor cells (CTCs) from the blood sample of head and neck cancer (HNC) patients was evaluated. Detection and enumeration of CTCs are critical for monitoring cancer progression. Both types of nanostructured cellulose were chemically modified with Epithelial Cell Adhesion Molecule (EpCAM) antibody and iron oxide nanoparticles. The EpCAM antibody facilitated the engagement of CTCs, promoting entrapment within the cellulose cage structure. Iron oxide nanoparticles, on the other hand, rendered the cages activatable via the use of a magnet for the capture and separation of entrapped CTCs. The efficiency of the network structures is shown in head and neck cancer (HNC) patients' blood samples. It was observed that the degree of chemical functionalization of hydroxyl groups located within the CNCs or CNFs with anti-EpCAM determined the efficiency of the system's interaction with CTCs. Further, our result indicated that inflexible scaffolds of nanocrystals interacted more efficiently with CTCs than that of the fibrous CNF scaffolds. Network structures derived from CNCs demonstrated comparable CTC capturing efficiency to commercial standard, OncoDiscover®. The output of the work will provide the chemical design principles of cellulosic materials intended for constructing affordable platforms for monitoring cancer progression in 'real time'.
Collapse
Affiliation(s)
- Raj Shankar Hazra
- Department of Mechanical Engineering, North Dakota State University, Fargo, ND 58108, USA; Department of Coatings and Polymeric Materials, North Dakota State University, Fargo 58108, ND, USA
| | - Narendra Kale
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo 58108, ND, USA; Department of Pharmaceutical Sciences, North Dakota State University, Fargo 58108, ND, USA
| | - Camden Boyle
- Department of Engineering and Technology, Southeast Missouri State University, One University Plaza, MS6825, Cape Girardeau, MO 63701, USA
| | - Kayla B Molina
- Department of Biomedical Engineering, The University of Minnesota Twin Cities, Minneapolis, MN 55455, USA
| | - Alain D'Souza
- Actorius Innovations and Research, Pune, India; Actorius Innovations and Research, Simi Valley, CA 93063, USA
| | - Gourishankar Aland
- Actorius Innovations and Research, Pune, India; Actorius Innovations and Research, Simi Valley, CA 93063, USA
| | - Long Jiang
- Department of Mechanical Engineering, North Dakota State University, Fargo, ND 58108, USA
| | - Pankaj Chaturvedi
- Department of Head and Neck Surgical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Santaneel Ghosh
- Department of Engineering and Technology, Southeast Missouri State University, One University Plaza, MS6825, Cape Girardeau, MO 63701, USA
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo 58108, ND, USA
| | - Jayant Khandare
- Actorius Innovations and Research, Pune, India; School of Pharmacy, Dr. Vishwananth Karad MIT World Peace University, Pune 411038, India; School of Consciousness, Dr. Vishwananth Karad MIT World Peace University, Pune 411038, India; Actorius Innovations and Research, Simi Valley, CA 93063, USA.
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo 58108, ND, USA.
| |
Collapse
|
8
|
Poellmann MJ, Bu J, Kim D, Iida M, Hong H, Wang AZ, Wheeler DL, Kimple RJ, Hong S. Circulating tumor cell abundance in head and neck squamous cell carcinoma decreases with successful chemoradiation and cetuximab treatment. Cancer Lett 2023; 562:216187. [PMID: 37068555 PMCID: PMC10510654 DOI: 10.1016/j.canlet.2023.216187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common and deadly cancer. Circulating tumor cell (CTC) abundance may a valuable, prognostic biomarker in low- and intermediate-risk patients. However, few technologies have demonstrated success in detecting CTCs in these populations. We prospectively collected longitudinal CTC counts from two cohorts of patients receiving treatments at our institution using a highly sensitive device that purifies CTCs using biomimetic cell rolling and dendrimer-conjugated antibodies. In patients with intermediate risk human papillomavirus (HPV)-positive HNSCC, elevated CTC counts were detected in 13 of 14 subjects at screening with a median of 17 CTC/ml (range 0.2-2986.5). A second cohort of non-metastatic, HPV- HNSCC subjects received cetuximab monotherapy followed by surgical resection. In this cohort, all subjects had elevated baseline CTC counts median of 73 CTC/ml (range 5.4-332.9) with statistically significant declines during treatment. Interestingly, two patients with recurrent disease had elevated CTC counts during and following treatment, which also correlated with growth of size and ki67 expression in the primary tumor. The results suggest that our device may be a valuable tool for evaluating the success of less intensive treatment regimens.
Collapse
Affiliation(s)
- Michael J Poellmann
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, 53705, USA; Capio Biosciences, Madison, WI, 53719, USA; Capio Biosciences Korea, Incheon, South Korea
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, 53705, USA; Capio Biosciences, Madison, WI, 53719, USA; Capio Biosciences Korea, Incheon, South Korea
| | - DaWon Kim
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Heejoo Hong
- Department of Clinical Pharmacology & Therapeutics, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Andrew Z Wang
- Capio Biosciences, Madison, WI, 53719, USA; Capio Biosciences Korea, Incheon, South Korea; Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Deric L Wheeler
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, 53705, USA; Capio Biosciences, Madison, WI, 53719, USA; Capio Biosciences Korea, Incheon, South Korea; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA; Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, 53705, USA; Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA; Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul, South Korea.
| |
Collapse
|
9
|
Hong B, Zhang X, Du X, Yang D, Hu Z, Zhang X, Zhang N. Exploring the Potential Driver Gene Mutations That Promote Renal Cancer Cell Metastasis and Implantation Based on Circulating Tumor Cells Culture. Diagnostics (Basel) 2023; 13:diagnostics13111855. [PMID: 37296706 DOI: 10.3390/diagnostics13111855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Studies have shown that the circulating tumor cell (CTC) is a necessary condition for the invasion and distant metastasis of renal cell carcimona (RCC). However, few CTCs-related gene mutations have been developed which could promote the metastasis and implantation of RCC. The objective of this study is to explore the potential driver gene mutations that promote RCC metastasis and implantation based on CTCs culture. Fifteen patients with primary mRCC and three healthy subjects were included, and peripheral blood was obtained. After the preparation of synthetic biological scaffolds, peripheral blood CTCs were cultured. Successful cultured CTCs were applied to construct CTCs-derived xenograft (CDX) models, followed by DNA extraction, whole exome sequencing (WES) and bioinformatics analysis. Synthetic biological scaffolds were constructed based on previously applied techniques, and peripheral blood CTCs culture was successfully performed. We then constructed CDX models and performed WES, and explored the potential driver gene mutations that may promote RCC metastasis and implantation. Bioinformatics analysis showed that KAZN and POU6F2 may be closely related to the prognosis of RCC. We successfully performed the culture of peripheral blood CTCs and, on this basis we initially explored the potential driver mutations for the metastasis and implantation of RCC.
Collapse
Affiliation(s)
- Baoan Hong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xuezhou Zhang
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xin Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Urology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Dazhi Yang
- Acrogenic Biotechnologies INC, Rockville, MD 20850, USA
| | - Zhiyuan Hu
- 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 of China, Beijing 100190, China
| | - Xiuli Zhang
- 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 of China, Beijing 100190, China
| | - Ning Zhang
- Department of Urology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
10
|
Poellmann MJ, Bu J, Liu S, Wang AZ, Seyedin SN, Chandrasekharan C, Hong H, Kim Y, Caster JM, Hong S. Nanotechnology and machine learning enable circulating tumor cells as a reliable biomarker for radiotherapy responses of gastrointestinal cancer patients. Biosens Bioelectron 2023; 226:115117. [PMID: 36753988 PMCID: PMC10034717 DOI: 10.1016/j.bios.2023.115117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/13/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
A highly sensitive, circulating tumor cell (CTC)-based liquid biopsy was used to monitor gastrointestinal cancer patients during treatment to determine if CTC abundance was predictive of disease recurrence. The approach used a combination of biomimetic cell rolling on recombinant E-selectin and dendrimer-mediated multivalent immunocapture at the nanoscale to purify CTCs from peripheral blood mononuclear cells. Due to the exceptionally high numbers of CTCs captured, a machine learning algorithm approach was developed to efficiently and reliably quantify abundance of immunocytochemically-labeled cells. A convolutional neural network and logistic regression model achieved 82.9% true-positive identification of CTCs with a false positive rate below 0.1% on a validation set. The approach was then used to quantify CTC abundance in peripheral blood samples from 27 subjects before, during, and following treatments. Samples drawn from the patients either prior to receiving radiotherapy or early in chemotherapy had a median 50 CTC ml-1 whole blood (range 0.6-541.6). We found that the CTC counts drawn 3 months post treatment were predictive of disease progression (p = .045). This approach to quantifying CTC abundance may be a clinically impactful in the timely determination of gastrointestinal cancer progression or response to treatment.
Collapse
Affiliation(s)
- Michael J Poellmann
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, 53705, USA; Capio Biosciences, Inc., Madison, WI, 53719, USA and Capio Biosciences Korea, Incheon, 21983 South Korea
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, 53705, USA; Capio Biosciences, Inc., Madison, WI, 53719, USA and Capio Biosciences Korea, Incheon, 21983 South Korea; Department of Biological Engineering, Inha University, Incheon, 22212, South Korea
| | - Stanley Liu
- Capio Biosciences, Inc., Madison, WI, 53719, USA and Capio Biosciences Korea, Incheon, 21983 South Korea
| | - Andrew Z Wang
- Capio Biosciences, Inc., Madison, WI, 53719, USA and Capio Biosciences Korea, Incheon, 21983 South Korea; Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Steven N Seyedin
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA
| | | | - Heejoo Hong
- Department of Clinical Pharmacology & Therapeutics, Asan Medical Center, University of Ulsan, Seoul, 05505, South Korea
| | - YoungSoo Kim
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon, 21983, South Korea
| | - Joseph M Caster
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, 53705, USA; Capio Biosciences, Inc., Madison, WI, 53719, USA and Capio Biosciences Korea, Incheon, 21983 South Korea; Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon, 21983, South Korea; Lachman Institute for Pharmaceutical Development, University of Wisconsin-Madison, Madison, WI, 53705, USA; Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
11
|
Ghorbanizamani F, Moulahoum H, Guler Celik E, Zihnioglu F, Beduk T, Goksel T, Turhan K, Timur S. Design of Polymeric Surfaces as Platforms for Streamlined Cancer Diagnostics in Liquid Biopsies. BIOSENSORS 2023; 13:400. [PMID: 36979612 PMCID: PMC10046689 DOI: 10.3390/bios13030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 06/18/2023]
Abstract
Minimally invasive approaches for cancer diagnosis are an integral step in the quest to improve cancer survival. Liquid biopsies such as blood samples are matrices explored to extract valuable information about the tumor and its state through various indicators, such as proteins, peptides, tumor DNA, or circulating tumor cells. Although these markers are scarce, making their isolation and detection in complex matrices challenging, the development in polymer chemistry producing interesting structures, including molecularly imprinted polymers, branched polymers, nanopolymer composites, and hybrids, allowed the development of enhanced platforms with impressive performance for liquid biopsies analysis. This review describes the latest advances and developments in polymer synthesis and their application for minimally invasive cancer diagnosis. The polymer structures improve the operational performances of biosensors through various processes, such as increased affinity for enhanced sensitivity, improved binding, and avoidance of non-specific interactions for enhanced specificity. Furthermore, polymer-based materials can be a tremendous help in signal amplification of usually low-concentrated targets in the sample. The pros and cons of these materials, how the synthesis process affects their performance, and the device applications for liquid biopsies diagnosis will be critically reviewed to show the essentiality of this technology in oncology and clinical biomedicine.
Collapse
Affiliation(s)
- Faezeh Ghorbanizamani
- Biochemistry Department, Faculty of Science, Ege University, Bornova, 35100 Izmir, Türkiye
| | - Hichem Moulahoum
- Biochemistry Department, Faculty of Science, Ege University, Bornova, 35100 Izmir, Türkiye
| | - Emine Guler Celik
- Bioengineering Department, Faculty of Engineering, Ege University, Bornova, 35100 Izmir, Türkiye
- EGE SCIENCE PRO Scientific Research Inc., Ege University, IdeEGE Technology Development Zone, Bornova, 35100 Izmir, Türkiye
| | - Figen Zihnioglu
- Biochemistry Department, Faculty of Science, Ege University, Bornova, 35100 Izmir, Türkiye
| | - Tutku Beduk
- Silicon Austria Labs GmbH: Sensor Systems, Europastrasse 12, 9524 Villach, Austria
| | - Tuncay Goksel
- EGE SCIENCE PRO Scientific Research Inc., Ege University, IdeEGE Technology Development Zone, Bornova, 35100 Izmir, Türkiye
- Department of Pulmonary Medicine, Faculty of Medicine, Ege University, Bornova, 35100 Izmir, Türkiye
- EGESAM-Ege University Translational Pulmonary Research Center, Bornova, 35100 Izmir, Türkiye
| | - Kutsal Turhan
- EGE SCIENCE PRO Scientific Research Inc., Ege University, IdeEGE Technology Development Zone, Bornova, 35100 Izmir, Türkiye
- Department of Thoracic Surgery, Faculty of Medicine, Ege University, Bornova, 35100 Izmir, Türkiye
| | - Suna Timur
- Biochemistry Department, Faculty of Science, Ege University, Bornova, 35100 Izmir, Türkiye
- EGE SCIENCE PRO Scientific Research Inc., Ege University, IdeEGE Technology Development Zone, Bornova, 35100 Izmir, Türkiye
- Central Research Testing and Analysis Laboratory Research and Application Center, Ege University, Bornova, 35100 Izmir, Türkiye
| |
Collapse
|
12
|
Wu R, Wang K, Gai Y, Li M, Wang J, Wang C, Zhang Y, Xiao Z, Jiang D, Gao Z, Xia X. Nanomedicine for renal cell carcinoma: imaging, treatment and beyond. J Nanobiotechnology 2023; 21:3. [PMID: 36597108 PMCID: PMC9809106 DOI: 10.1186/s12951-022-01761-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023] Open
Abstract
The kidney is a vital organ responsible for maintaining homeostasis in the human body. However, renal cell carcinoma (RCC) is a common malignancy of the urinary system and represents a serious threat to human health. Although the overall survival of RCC has improved substantially with the development of cancer diagnosis and management, there are various reasons for treatment failure. Firstly, without any readily available biomarkers, timely diagnosis has been greatly hampered. Secondly, the imaging appearance also varies greatly, and its early detection often remains difficult. Thirdly, chemotherapy has been validated as unavailable for treating renal cancer in the clinic due to its intrinsic drug resistance. Concomitant with the progress of nanotechnological methods in pharmaceuticals, the management of kidney cancer has undergone a transformation in the recent decade. Nanotechnology has shown many advantages over widely used traditional methods, leading to broad biomedical applications ranging from drug delivery, prevention, diagnosis to treatment. This review focuses on nanotechnologies in RCC management and further discusses their biomedical translation with the aim of identifying the most promising nanomedicines for clinical needs. As our understanding of nanotechnologies continues to grow, more opportunities to improve the management of renal cancer are expected to emerge.
Collapse
Affiliation(s)
- Ruolin Wu
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Keshan Wang
- grid.33199.310000 0004 0368 7223Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yongkang Gai
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Mengting Li
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Jingjing Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Chenyang Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Yajing Zhang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zhiwei Xiao
- grid.413247.70000 0004 1808 0969Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dawei Jiang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zairong Gao
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Xiaotian Xia
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| |
Collapse
|
13
|
Couto-Cunha A, Jerónimo C, Henrique R. Circulating Tumor Cells as Biomarkers for Renal Cell Carcinoma: Ready for Prime Time? Cancers (Basel) 2022; 15:cancers15010287. [PMID: 36612281 PMCID: PMC9818240 DOI: 10.3390/cancers15010287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Renal cell carcinoma (RCC) is among the 15 most common cancers worldwide, with rising incidence. In most cases, this is a silent disease until it reaches advance stages, demanding new effective biomarkers in all domains, from detection to post-therapy monitoring. Circulating tumor cells (CTC) have the potential to provide minimally invasive information to guide assessment of the disease's aggressiveness and therapeutic strategy, representing a special pool of neoplastic cells which bear metastatic potential. In some tumor models, CTCs' enumeration has been associated with prognosis, but there is a largely unexplored potential for clinical applicability encompassing screening, diagnosis, early detection of metastases, prognosis, response to therapy and monitoring. Nonetheless, lack of standardization and high cost hinder the translation into clinical practice. Thus, new methods for collection and analysis (genomic, proteomic, transcriptomic, epigenomic and metabolomic) are needed to ascertain the role of CTC as a RCC biomarker. Herein, we provide a critical overview of the most recently published data on the role and clinical potential of CTCs in RCC, addressing their biology and the molecular characterization of this remarkable set of tumor cells. Furthermore, we highlight the existing and emerging techniques for CTC enrichment and detection, exploring clinical applications in RCC. Notwithstanding the notable progress in recent years, the use of CTCs in a routine clinical scenario of RCC patients requires further research and technological development, enabling multimodal analysis to take advantage of the wealth of information they provide.
Collapse
Affiliation(s)
- Anabela Couto-Cunha
- Integrated Master in Medicine, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Carmen Jerónimo
- Department of Pathology and Molecular Immunology, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Department of Pathology & Cancer Biology & Epigenetics Group—Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (P.CCC Raquel Seruca), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Rui Henrique
- Department of Pathology and Molecular Immunology, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
- Department of Pathology & Cancer Biology & Epigenetics Group—Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Centre Raquel Seruca (P.CCC Raquel Seruca), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Correspondence: or
| |
Collapse
|
14
|
Bu J, Jeong WJ, Jafari R, Kubiatowicz LJ, Nair A, Poellmann MJ, Hong RS, Liu EW, Owen RH, Rawding PA, Hopkins CM, Kim D, George DJ, Armstrong AJ, Král P, Wang AZ, Bruce J, Zhang T, Kimple RJ, Hong S. Bimodal liquid biopsy for cancer immunotherapy based on peptide engineering and nanoscale analysis. Biosens Bioelectron 2022; 213:114445. [PMID: 35679646 DOI: 10.1016/j.bios.2022.114445] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/13/2022] [Accepted: 05/30/2022] [Indexed: 11/02/2022]
Abstract
Despite its high potential, PD-L1 expressed by tumors has not been successfully utilized as a biomarker for estimating treatment responses to immunotherapy. Circulating tumor cells (CTCs) and tumor-derived exosomes that express PD-L1 can potentially be used as biomarkers; however, currently available assays lack clinically significant sensitivity and specificity. Here, a novel peptide-based capture surface is developed to effectively isolate PD-L1-expressing CTCs and exosomes from human blood. For the effective targeting of PD-L1, this study integrates peptide engineering strategies to enhance the binding strength and specificity of a β-hairpin peptide derived from PD-1 (pPD-1). Specifically, this study examines the effect of poly(ethylene glycol) spacers, the secondary peptide structure, and modification of peptide sequences (e.g., removal of biologically redundant amino acid residues) on capture efficiency. The optimized pPD-1 configuration captures PD-L1-expressing tumor cells and tumor-derived exosomes with 1.5-fold (p = 0.016) and 1.2-fold (p = 0.037) higher efficiencies, respectively, than their whole antibody counterpart (aPD-L1). This enhanced efficiency is translated into more clinically significant detection of CTCs (1.9-fold increase; p = 0.035) and exosomes (1.5-fold increase; p = 0.047) from patients' baseline samples, demonstrating stronger correlation with patients' treatment responses. Additionally, we confirmed that the clinical accuracy of our system can be further improved by co-analyzing the two biomarkers (bimodal CTC/exosome analysis). These data demonstrate that pPD-1-based capture is a promising approach for capturing PD-L1-expressing CTCs and exosomes, which can be used as a reliable biomarker for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiyoon Bu
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA; Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Woo-Jin Jeong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA; Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Roya Jafari
- Department of Chemistry, University of Illinois at Chicago, 845 W Taylor St, Chicago, IL, 60607, USA
| | - Luke J Kubiatowicz
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - Ashita Nair
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - Michael J Poellmann
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - Rachel S Hong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - Elizabeth W Liu
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - Randall H Owen
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - Piper A Rawding
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - Caroline M Hopkins
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - DaWon Kim
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA
| | - Daniel J George
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, 10 Bryan Searle Drive, Durham, NC, 27710, USA; Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, 20 Duke Medicine Cir, Durham, NC, 27710, USA
| | - Andrew J Armstrong
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, 10 Bryan Searle Drive, Durham, NC, 27710, USA; Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, 20 Duke Medicine Cir, Durham, NC, 27710, USA
| | - Petr Král
- Department of Chemistry, University of Illinois at Chicago, 845 W Taylor St, Chicago, IL, 60607, USA; Department of Physics, Department of Pharmaceutical Sciences, University of Illinois at Chicago, 845 W Taylar St, Chicage, IL, 60607, USA
| | - Andrew Z Wang
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Radiation Oncology and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Justine Bruce
- Department of Human Oncology, University of Wisconsin-Madison, Madison, 600 Highland Ave, WI, 53792, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, 600 Highland Ave, WI, 53792, USA
| | - Tian Zhang
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, 10 Bryan Searle Drive, Durham, NC, 27710, USA; Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University, 20 Duke Medicine Cir, Durham, NC, 27710, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin-Madison, Madison, 600 Highland Ave, WI, 53792, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, 600 Highland Ave, WI, 53792, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, University of Wisconsin - Madison, 777 Highland Ave, Madison, WI, 53705, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, 600 Highland Ave, WI, 53792, USA; Department of Biomedical Engineering, The University of Wisconsin-Madison, 1550 Engineering Dr., Madison, WI, 53705, USA; Yonsei Frontier Lab, Department of Pharmacy, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
15
|
Poellmann MJ, Rawding P, Kim D, Bu J, Kim Y, Hong S. Branched, dendritic, and hyperbranched polymers in liquid biopsy device design. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1770. [PMID: 34984833 PMCID: PMC9480505 DOI: 10.1002/wnan.1770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022]
Abstract
The development of minimally invasive tests for cancer diagnosis and prognosis will aid in the research of new treatments and improve survival rates. Liquid biopsies seek to derive actionable information from tumor material found in routine blood samples. The relative scarcity of tumor material in this complex mixture makes isolating and detecting cancerous material such as proteins, circulating tumor DNA, exosomes, and whole circulating tumor cells a challenge for device engineers. This review describes the chemistry and applications of branched and hyperbranched to improve the performance of liquid biopsy devices. These polymers can improve the performance of a liquid biopsy through several mechanisms. For example, polymers designed to increase the affinity of capture enhance device sensitivity. On the other hand, polymers designed to increase binding avidity or repel nonspecific adsorption enhance device specificity. Branched and hyperbranched polymers can also be used to amplify the signal from small amounts of detected material. The further development of hyperbranched polymers in liquid biopsy applications will enhance device capabilities and help these critical technologies reach the oncology clinic where they are sorely needed. This article is categorized under: Diagnostic Tools > Biosensing Diagnostic Tools > Diagnostic Nanodevices.
Collapse
Affiliation(s)
- Michael J Poellmann
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
- Capio Biosciences, Madison, Wisconsin, USA
| | - Piper Rawding
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - DaWon Kim
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
| | - YoungSoo Kim
- Department of Pharmacy, Yonsei University, Incheon, South Korea
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin, USA
- Capio Biosciences, Madison, Wisconsin, USA
- Department of Pharmacy, Yonsei University, Incheon, South Korea
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
16
|
Lee T, Rawding PA, Bu J, Hyun S, Rou W, Jeon H, Kim S, Lee B, Kubiatowicz LJ, Kim D, Hong S, Eun H. Machine-Learning-Based Clinical Biomarker Using Cell-Free DNA for Hepatocellular Carcinoma (HCC). Cancers (Basel) 2022; 14:2061. [PMID: 35565192 PMCID: PMC9103537 DOI: 10.3390/cancers14092061] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 02/08/2023] Open
Abstract
(1) Background: Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. Although various serum enzymes have been utilized for the diagnosis and prognosis of HCC, the currently available biomarkers lack the sensitivity needed to detect HCC at early stages and accurately predict treatment responses. (2) Methods: We utilized our highly sensitive cell-free DNA (cfDNA) detection system, in combination with a machine learning algorithm, to provide a platform for improved diagnosis and prognosis of HCC. (3) Results: cfDNA, specifically alpha-fetoprotein (AFP) expression in captured cfDNA, demonstrated the highest accuracy for diagnosing malignancies among the serum/plasma biomarkers used in this study, including AFP, aspartate aminotransferase, alanine aminotransferase, albumin, alkaline phosphatase, and bilirubin. The diagnostic/prognostic capability of cfDNA was further improved by establishing a cfDNA score (cfDHCC), which integrated the total plasma cfDNA levels and cfAFP-DNA expression into a single score using machine learning algorithms. (4) Conclusion: The cfDHCC score demonstrated significantly improved accuracy in determining the pathological features of HCC and predicting patients' survival outcomes compared to the other biomarkers. The results presented herein reveal that our cfDNA capture/analysis platform is a promising approach to effectively utilize cfDNA as a biomarker for the diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Taehee Lee
- Department of Biomedical Laboratory Science, Daegu Health College, Daegu 41453, Korea;
- Department of Senior Healthcare, Graduate School, Eulji University, Uijeongbu-si 11759, Korea;
| | - Piper A. Rawding
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin—Madison, Madison, WI 53705, USA; (P.A.R.); (J.B.); (L.J.K.); (D.K.)
- Wisconsin Center for NanoBioSystems (WisCNano), University of Wisconsin—Madison, Madison, WI 53705, USA
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin—Madison, Madison, WI 53705, USA; (P.A.R.); (J.B.); (L.J.K.); (D.K.)
- Wisconsin Center for NanoBioSystems (WisCNano), University of Wisconsin—Madison, Madison, WI 53705, USA
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Korea
- Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon 22212, Korea
| | - Sunghee Hyun
- Department of Senior Healthcare, Graduate School, Eulji University, Uijeongbu-si 11759, Korea;
| | - Woosun Rou
- Department of Internal Medicine, Chungnam National University Sejong Hospital (CNUSH), Sejong 30099, Korea; (W.R.); (H.J.)
| | - Hongjae Jeon
- Department of Internal Medicine, Chungnam National University Sejong Hospital (CNUSH), Sejong 30099, Korea; (W.R.); (H.J.)
| | - Seokhyun Kim
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon 35015, Korea; (S.K.); (B.L.)
| | - Byungseok Lee
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon 35015, Korea; (S.K.); (B.L.)
| | - Luke J. Kubiatowicz
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin—Madison, Madison, WI 53705, USA; (P.A.R.); (J.B.); (L.J.K.); (D.K.)
| | - Dawon Kim
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin—Madison, Madison, WI 53705, USA; (P.A.R.); (J.B.); (L.J.K.); (D.K.)
- Wisconsin Center for NanoBioSystems (WisCNano), University of Wisconsin—Madison, Madison, WI 53705, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin—Madison, Madison, WI 53705, USA; (P.A.R.); (J.B.); (L.J.K.); (D.K.)
- Wisconsin Center for NanoBioSystems (WisCNano), University of Wisconsin—Madison, Madison, WI 53705, USA
- Yonsei Frontier Lab, Department of Pharmacy, Yonsei University, Seoul 03722, Korea
| | - Hyuksoo Eun
- Yonsei Frontier Lab, Department of Pharmacy, Yonsei University, Seoul 03722, Korea
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| |
Collapse
|
17
|
Rawding PA, Bu J, Wang J, Kim D, Drelich AJ, Kim Y, Hong S. Dendrimers for cancer immunotherapy: Avidity-based drug delivery vehicles for effective anti-tumor immune response. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1752. [PMID: 34414690 PMCID: PMC9485970 DOI: 10.1002/wnan.1752] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy, or the utilization of a patient's own immune system to treat cancer, has shifted the paradigm of cancer treatment. Despite meaningful responses being observed in multiple studies, currently available immunotherapy platforms have only proven effective to a small subset of patients. To address this, nanoparticles have been utilized as a novel carrier for immunotherapeutic drugs, achieving robust anti-tumor effects with increased adaptive and durable responses. Specifically, dendrimer nanoparticles have attracted a great deal of scientific interest due to their versatility in various therapeutic applications, resulting from their unique physicochemical properties and chemically well-defined architecture. This review offers a comprehensive overview of dendrimer-based immunotherapy technologies, including their formulations, biological functionalities, and therapeutic applications. Common formulations include: (1) modulators of cytokine secretion of immune cells (adjuvants); (2) facilitators of the recognition of tumorous antigens (vaccines); (3) stimulators of immune effectors to selectively attack cells expressing specific antigens (antibodies); and (4) inhibitors of immune-suppressive responses (immune checkpoint inhibitors). On-going works and prospects of dendrimer-based immunotherapies are also discussed. Overall, this review provides a critical overview on rapidly growing dendrimer-based immunotherapy technologies and serves as a guideline for researchers and clinicians who are interested in this field. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Piper A Rawding
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jianxin Wang
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - DaWon Kim
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Adam J Drelich
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Youngsoo Kim
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA,Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA,Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
18
|
Jeong W, Bu J, Jafari R, Rehak P, Kubiatowicz LJ, Drelich AJ, Owen RH, Nair A, Rawding PA, Poellmann MJ, Hopkins CM, Král P, Hong S. Hierarchically Multivalent Peptide-Nanoparticle Architectures: A Systematic Approach to Engineer Surface Adhesion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103098. [PMID: 34894089 PMCID: PMC8811846 DOI: 10.1002/advs.202103098] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/04/2021] [Indexed: 05/20/2023]
Abstract
The multivalent binding effect has been the subject of extensive studies to modulate adhesion behaviors of various biological and engineered systems. However, precise control over the strong avidity-based binding remains a significant challenge. Here, a set of engineering strategies are developed and tested to systematically enhance the multivalent binding of peptides in a stepwise manner. Poly(amidoamine) (PAMAM) dendrimers are employed to increase local peptide densities on a substrate, resulting in hierarchically multivalent architectures (HMAs) that display multivalent dendrimer-peptide conjugates (DPCs) with various configurations. To control binding behaviors, effects of the three major components of the HMAs are investigated: i) poly(ethylene glycol) (PEG) linkers as spacers between conjugated peptides; ii) multiple peptides on the DPCs; and iii) various surface arrangements of HMAs (i.e., a mixture of DPCs each containing different peptides vs DPCs cofunctionalized with multiple peptides). The optimized HMA configuration enables significantly enhanced target cell binding with high selectivity compared to the control surfaces directly conjugated with peptides. The engineering approaches presented herein can be applied individually or in combination, providing guidelines for the effective utilization of biomolecular multivalent interactions using DPC-based HMAs.
Collapse
Affiliation(s)
- Woo‐jin Jeong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
- Department of Biological Sciences and BioengineeringInha University100 Inha‐ro, Michuhol‐guIncheon22212Republic of Korea
| | - Jiyoon Bu
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Roya Jafari
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Pavel Rehak
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Luke J. Kubiatowicz
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Adam J. Drelich
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Randall H. Owen
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Ashita Nair
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Piper A. Rawding
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Michael J. Poellmann
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Caroline M. Hopkins
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
| | - Petr Král
- Department of ChemistryUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
- Departments of Physics, Pharmaceutical Sciences and Chemical EngineeringUniversity of Illinois at Chicago845 W Taylor StChicagoIL60607USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division and Wisconsin Center for NanoBioSystems (WisCNano)School of PharmacyUniversity of Wisconsin‐Madison777 Highland AveMadisonWI53705USA
- Department of Biomedical EngineeringThe University of Wisconsin‐Madison1550 Engineering Dr.MadisonWI53705USA
- Yonsei Frontier LabDepartment of PharmacyYonsei University50 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| |
Collapse
|
19
|
Liu X, Ma L, Yan W, Aazmi A, Fang M, Xu X, Kang H, Xu X. A review of recent progress toward the efficient separation of circulating tumor cells via micro‐/nanostructured microfluidic chips. VIEW 2022. [DOI: 10.1002/viw.20210013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Xiaoshi Liu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Liang Ma
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou P. R. China
| | - Wenyuan Yan
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems Zhejiang University Hangzhou P. R. China
- School of Mechanical Engineering Zhejiang University Hangzhou P. R. China
| | - Minghe Fang
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Xiuzhen Xu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Hanyue Kang
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| | - Xiaobin Xu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education Shanghai Key Laboratory of D&A for Metal‐Functional Materials School of Materials Science and Engineering Institute for Advanced Study Tongji University Shanghai P. R. China
| |
Collapse
|
20
|
Cytochalasin B Treatment and Osmotic Pressure Enhance the Production of Extracellular Vesicles (EVs) with Improved Drug Loading Capacity. NANOMATERIALS 2021; 12:nano12010003. [PMID: 35009953 PMCID: PMC8746776 DOI: 10.3390/nano12010003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) have been highlighted as novel drug carriers due to their unique structural properties and intrinsic features, including high stability, biocompatibility, and cell-targeting properties. Although many efforts have been made to harness these features to develop a clinically effective EV-based therapeutic system, the clinical translation of EV-based nano-drugs is hindered by their low yield and loading capacity. Herein, we present an engineering strategy that enables upscaled EV production with increased loading capacity through the secretion of EVs from cells via cytochalasin-B (CB) treatment and reduction of EV intravesicular contents through hypo-osmotic stimulation. CB (10 µg/mL) promotes cells to extrude EVs, producing ~three-fold more particles than through natural EV secretion. When CB is induced in hypotonic conditions (223 mOsm/kg), the produced EVs (hypo-CIMVs) exhibit ~68% less intravesicular protein, giving 3.4-fold enhanced drug loading capacity compared to naturally secreted EVs. By loading doxorubicin (DOX) into hypo-CIMVs, we found that hypo-CIMVs efficiently deliver their drug cargos to their target and induce up to ~1.5-fold more cell death than the free DOX. Thus, our EV engineering offers the potential for leveraging EVs as an effective drug delivery vehicle for cancer treatment.
Collapse
|
21
|
Jiang M, Jin S, Han J, Li T, Shi J, Zhong Q, Li W, Tang W, Huang Q, Zong H. Detection and clinical significance of circulating tumor cells in colorectal cancer. Biomark Res 2021; 9:85. [PMID: 34798902 PMCID: PMC8605607 DOI: 10.1186/s40364-021-00326-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/27/2021] [Indexed: 02/08/2023] Open
Abstract
Histopathological examination (biopsy) is the "gold standard" for the diagnosis of colorectal cancer (CRC). However, biopsy is an invasive method, and due to the temporal and spatial heterogeneity of the tumor, a single biopsy cannot reveal the comprehensive biological characteristics and dynamic changes of the tumor. Therefore, there is a need for new biomarkers to improve CRC diagnosis and to monitor and treat CRC patients. Numerous studies have shown that "liquid biopsy" is a promising minimally invasive method for early CRC detection. A liquid biopsy mainly samples circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), microRNA (miRNA) and extracellular vesicles (EVs). CTCs are malignant cells that are shed from the primary tumors and/or metastases into the peripheral circulation. CTCs carry information on both primary tumors and metastases that can reflect dynamic changes in tumors in a timely manner. As a promising biomarker, CTCs can be used for early disease detection, treatment response and disease progression evaluation, disease mechanism elucidation, and therapeutic target identification for drug development. This review will discuss currently available technologies for plasma CTC isolation and detection, their utility in the management of CRC patients and future research directions.
Collapse
Affiliation(s)
- Miao Jiang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Shuiling Jin
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Jinming Han
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Tong Li
- BGI College, Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, Henan, China
| | - Jianxiang Shi
- BGI College, Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, Henan, China.,Precision Medicine Center, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052, China
| | - Qian Zhong
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Wen Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China
| | - Wenxue Tang
- Departments of Otolaryngology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Qinqin Huang
- Academy of medical science, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Hong Zong
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, NO.1 Eastern Jianshe Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
22
|
Fabrication of Formalin-Fixed, Paraffin-Embedded (FFPE) Circulating Tumor Cell (CTC) Block Using a Hydrogel Core-Mediated Method. MICROMACHINES 2021; 12:mi12091128. [PMID: 34577771 PMCID: PMC8466852 DOI: 10.3390/mi12091128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/24/2022]
Abstract
Circulating tumor cells (CTCs) are extremely low-frequency cells in the bloodstream. As those cells have detached from the primary tumor tissues and it circulates throughout the whole body, they are considered as promising diagnostic biomarkers for clinical application. However, the analysis of CTC is often restricted due to their rarity and heterogeneity, as well as their short-term presence. Here we proposed formalin-fixed, paraffin-embedded (FFPE) CTC block method, in combination manner with the hydrogel core-mediated CTC accumulation and conventional paraffin tissue block preparation. The hydrogel core specifically captures and releases cancer cells with high efficiency with an immunoaffinity manner. An additional shell structure protects the isolated cancer cells during the FFPE CTC block preparation process. The fabricated FFPE CTC block was sectioned and cytopathologically investigated just the same way as the conventional tissue block. Our results demonstrate that rare cells such as CTCs can also be prepared for FFPE cell blocks and shows great promise for cytopathological CTC studies.
Collapse
|
23
|
Bu J, Lee TH, Poellmann MJ, Rawding PA, Jeong W, Hong RS, Hyun SH, Eun HS, Hong S. Tri-modal liquid biopsy: Combinational analysis of circulating tumor cells, exosomes, and cell-free DNA using machine learning algorithm. Clin Transl Med 2021; 11:e499. [PMID: 34459134 PMCID: PMC8335965 DOI: 10.1002/ctm2.499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022] Open
Affiliation(s)
- Jiyoon Bu
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Center for NanoBioSystemsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Tae Hee Lee
- Research Institute for Future Medical ScienceChungnam National University Sejong Hospital (CNUSH)SejongRepublic of Korea
- Department of Senior HealthcareBK21 Plus ProgramGraduated SchoolEulji UniversityDaejeonRepublic of Korea
| | - Michael J. Poellmann
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Center for NanoBioSystemsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Piper A. Rawding
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Center for NanoBioSystemsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Woo‐Jin Jeong
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Center for NanoBioSystemsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Biological Sciences and BioengineeringInha UniversityIncheonRepublic of Korea
| | - Rachel S. Hong
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Sung Hee Hyun
- Department of Senior HealthcareBK21 Plus ProgramGraduated SchoolEulji UniversityDaejeonRepublic of Korea
| | - Hyuk Soo Eun
- Department of Internal MedicineChungnam National University School of MedicineDaejeonRepublic of Korea
| | - Seungpyo Hong
- Pharmaceutical Sciences DivisionSchool of PharmacyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Center for NanoBioSystemsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Yonsei Frontier Lab and Department of PharmacyYonsei UniversitySeoulRepublic of Korea
| |
Collapse
|
24
|
Nair A, Bu J, Bugno J, Rawding PA, Kubiatowicz LJ, Jeong WJ, Hong S. Size-Dependent Drug Loading, Gene Complexation, Cell Uptake, and Transfection of a Novel Dendron-Lipid Nanoparticle for Drug/Gene Co-delivery. Biomacromolecules 2021; 22:3746-3755. [PMID: 34319087 DOI: 10.1021/acs.biomac.1c00541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Dendron micelles have shown promising results as a multifunctional delivery system, owing to their unique molecular architecture. Herein, we have prepared a novel poly(amidoamine) (PAMAM) dendron-lipid hybrid nanoparticle (DLNP) as a nanocarrier for drug/gene co-delivery and examined how the dendron generation of DLNPs impacts their cargo-carrying capabilities. DLNPs, formed by a thin-layer hydration method, were internally loaded with chemo-drugs and externally complexed with plasmids. Compared to generation 2 dendron DLNP (D2LNPs), D3LNPs demonstrated a higher drug encapsulation efficiency (31% vs 87%) and better gene complexation (minimal N/P ratio of 20:1 vs 5:1 for complexation) due to their smaller micellar aggregation number and higher charge density, respectively. Furthermore, D3LNPs were able to avoid endocytosis and subsequent lysosomal degradation and demonstrated a higher cellular uptake than D2LNPs. As a result, D3LNPs exhibited significantly enhanced antitumor and gene transfection efficacy in comparison to D2LNPs. These findings provide design cues for engineering multifunctional dendron-based nanotherapeutic systems for effective combination cancer treatment.
Collapse
Affiliation(s)
- Ashita Nair
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States.,Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, The University of Wisconsin-Madison, 777 Highland Ave., Madison, Wisconsin 53705, United States
| | - Jiyoon Bu
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States.,Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, The University of Wisconsin-Madison, 777 Highland Ave., Madison, Wisconsin 53705, United States
| | - Jason Bugno
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois 60612, United States
| | - Piper A Rawding
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States.,Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, The University of Wisconsin-Madison, 777 Highland Ave., Madison, Wisconsin 53705, United States
| | - Luke J Kubiatowicz
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States.,Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, The University of Wisconsin-Madison, 777 Highland Ave., Madison, Wisconsin 53705, United States
| | - Woo-Jin Jeong
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States.,Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Seungpyo Hong
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States.,Wisconsin Center for NanoBioSystems (WisCNano), School of Pharmacy, The University of Wisconsin-Madison, 777 Highland Ave., Madison, Wisconsin 53705, United States.,Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, Illinois 60612, United States.,Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
25
|
Wang Q, Shi T, Wan M, Wei J, Wang F, Mao C. Research progress of using micro/nanomotors in the detection and therapy of diseases related to the blood environment. J Mater Chem B 2021; 9:283-294. [PMID: 33241834 DOI: 10.1039/d0tb02055a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Micro/nanomotors bring new possibilities for the detection and therapy of diseases related to the blood environment with their unique motion effect. This work reviews the research progress of using micro/nanomotors in the detection and therapy of diseases related to the blood environment. First, we outline the advantages of using micro/nanomotors in blood-related disease detection. To be specific, the motion capability of micro/nanomotors can increase plasma or blood fluid convection and accelerate the interaction between the sample and the capture probe. This allows the effective reduction of the amount of reagents and treatment steps. Therefore, the application of micro/nanomotors significantly improves the analytical performance. Second, we discuss the key challenges and future prospects of micro/nanomotors in the treatment of blood-environment related diseases. It is very important to design a unique treatment plan according to the etiology and specific microenvironment of the disease. The next generation of micro/nanomotors is expected to bring exciting progress to the detection and therapy of blood-environment related diseases.
Collapse
Affiliation(s)
- Qi Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China. and School of Geography, Nanjing Normal University, Nanjing, 210023, China
| | - Tao Shi
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.
| | - Jia Wei
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Fenghe Wang
- Jiangsu Province Key Laboratory of Environmental Engineering, School of Environment, Nanjing Normal University, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
26
|
Zhao C, Jian X, Zhang X, Guo J, Gao Z, Song YY. Rapid Capture and Photocatalytic Inactivation of Target Cells from Whole Blood by Rotating Janus Nanotubes. ACS APPLIED MATERIALS & INTERFACES 2021; 13:12972-12981. [PMID: 33689269 DOI: 10.1021/acsami.1c02042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Effective isolation and removal of target tumor cells from patients' peripheral blood are of great importance to clinical prognosis and recovery. However, the extremely low quantity of target cells in peripheral blood becomes one of the challenges in this respect. Herein, we design and synthesize an innovative nanostructure based on magnetic TiO2 nanotubes with Pt nanoparticles' asymmetrical decoration for effectively capturing and inactivating target cells. Using CCRF-CEM as the model cell, the resulting nanotubes with accurate modification of recognition probes exhibit high selectivity and cell-isolation efficiency upon real blood samples. Particularly, the target cells are selectively captured at a low concentration with a recovery rate of 73.0 ± 11.5% at five cells per milliliter for whole blood samples. Consequently, benefitting from the remarkable photocatalytic activity of the Janus nanotubes, these isolated cells can be rapidly inactivated via light-emitting diode (LED) irradiation with an ignorable effect on normal cells. This work offers a new paradigm for high-efficient isolating/killing target cells from a complex medium.
Collapse
Affiliation(s)
- Chenxi Zhao
- College of Sciences, Northeastern University, Shenyang 110004, China
| | - Xiaoxia Jian
- College of Sciences, Northeastern University, Shenyang 110004, China
| | - Xi Zhang
- College of Sciences, Northeastern University, Shenyang 110004, China
| | - Junli Guo
- College of Sciences, Northeastern University, Shenyang 110004, China
| | - Zhida Gao
- College of Sciences, Northeastern University, Shenyang 110004, China
| | - Yan-Yan Song
- College of Sciences, Northeastern University, Shenyang 110004, China
| |
Collapse
|
27
|
Bu J, Lee TH, Jeong WJ, Poellmann MJ, Mudd K, Eun HS, Liu EW, Hong S, Hyun SH. Enhanced detection of cell-free DNA (cfDNA) enables its use as a reliable biomarker for diagnosis and prognosis of gastric cancer. PLoS One 2020; 15:e0242145. [PMID: 33264292 PMCID: PMC7710035 DOI: 10.1371/journal.pone.0242145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
Although circulating cell-free DNA (cfDNA) is a promising biomarker for the diagnosis and prognosis of various tumors, clinical correlation of cfDNA with gastric cancer has not been fully understood. To address this, we developed a highly sensitive cfDNA capture system by integrating polydopamine (PDA) and silica. PDA-silica hybrids incorporated different molecular interactions to a single system, enhancing cfDNA capture by 1.34-fold compared to the conventional silica-based approach (p = 0.001), which was confirmed using cell culture supernatants. A clinical study using human plasma samples revealed that the diagnostic accuracy of the new system to be superior than the commercially available cfDNA kit, as well as other serum antigen tests. Among the cancer patients, plasma cfDNA levels exhibited a good correlation with the size of a tumor. cfDNA was also predicative of distant metastasis, as the median cfDNA levels of metastatic cancer patients were ~60-fold higher than those without metastasis (p = 0.008). Furthermore, high concordance between tissue biopsy and cfDNA genomic analysis was found, as HER2 expression in cfDNA demonstrated an area under ROC curve (AUC) of 0.976 (p <0.001) for detecting patients with HER2-positive tumors. The new system also revealed high prognostic capability of cfDNA, as the concentration of cfDNA was highly associated with the survival outcomes. Our novel technology demonstrates the potential to achieve efficient detection of cfDNA that may serve as a reliable biomarker for gastric tumor.
Collapse
Affiliation(s)
- Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Tae Hee Lee
- Department of Senior Healthcare, BK21 plus program, Graduated School, Eulji University, Daejeon, Republic of Korea
- Research Institute for Future Medical Science, Chungnam National University Sejong Hospital (CNUSH), Sejong, Republic of Korea
| | - Woo-jin Jeong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biological Engineering, Inha University, Incheon, Republic of Korea
| | - Michael J. Poellmann
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Kara Mudd
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Hyuk Soo Eun
- Divison of Gastroenterology and Hepatology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Elizabeth W. Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
- Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul, Republic of Korea
- * E-mail: (SH); (SHH)
| | - Sung Hee Hyun
- Department of Senior Healthcare, BK21 plus program, Graduated School, Eulji University, Daejeon, Republic of Korea
- * E-mail: (SH); (SHH)
| |
Collapse
|
28
|
Li H, Sun J, Zhu H, Wu H, Zhang H, Gu Z, Luo K. Recent advances in development of dendritic polymer-based nanomedicines for cancer diagnosis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1670. [PMID: 32949116 DOI: 10.1002/wnan.1670] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 02/05/2023]
Abstract
Dendritic polymers have highly branched three-dimensional architectures, the fourth type apart from linear, cross-linked, and branched one. They possess not only a large number of terminal functional units and interior cavities, but also a low viscosity with weak or no entanglement. These features endow them with great potential in various biomedicine applications, including drug delivery, gene therapy, tissue engineering, immunoassay and bioimaging. Most review articles related to bio-related applications of dendritic polymers focus on their drug or gene delivery, while very few of them are devoted to their function as cancer diagnosis agents, which are essential for cancer treatment. In this review, we will provide comprehensive insights into various dendritic polymer-based cancer diagnosis agents. Their classification and preparation are presented for readers to have a precise understanding of dendritic polymers. On account of physical/chemical properties of dendritic polymers and biological properties of cancer, we will suggest a few design strategies for constructing dendritic polymer-based diagnosis agents, such as active or passive targeting strategies, imaging reporters-incorporating strategies, and/or internal stimuli-responsive degradable/enhanced imaging strategies. Their recent applications in in vitro diagnosis of cancer cells or exosomes and in vivo diagnosis of primary and metastasis tumor sites with the aid of single/multiple imaging modalities will be discussed in great detail. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging Diagnostic Tools > in vitro Nanoparticle-Based Sensing.
Collapse
Affiliation(s)
- Haonan Li
- Laboratory of Stem Cell Biology, and Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayu Sun
- Laboratory of Stem Cell Biology, and Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hongyan Zhu
- Laboratory of Stem Cell Biology, and Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Haoxing Wu
- Laboratory of Stem Cell Biology, and Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, California, USA
| | - Zhongwei Gu
- Laboratory of Stem Cell Biology, and Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kui Luo
- Laboratory of Stem Cell Biology, and Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Barth DA, Drula R, Ott L, Fabris L, Slaby O, Calin GA, Pichler M. Circulating Non-coding RNAs in Renal Cell Carcinoma-Pathogenesis and Potential Implications as Clinical Biomarkers. Front Cell Dev Biol 2020; 8:828. [PMID: 33042985 PMCID: PMC7523432 DOI: 10.3389/fcell.2020.00828] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
Liquid biopsy-the determination of circulating cells, proteins, DNA or RNA from biofluids through a "less invasive" approach-has emerged as a novel approach in all cancer entities. Circulating non-(protein) coding RNAs including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and YRNAs can be passively released by tissue or cell damage or actively secreted as cell-free circulating RNAs, bound to lipoproteins or carried by exosomes. In renal cell carcinoma (RCC), a growing body of evidence suggests circulating non-coding RNAs (ncRNAs) such as miRNAs, lncRNAs, and YRNAs as promising and easily accessible blood-based biomarkers for the early diagnosis of RCC as well as for the prediction of prognosis and treatment response. In addition, circulating ncRNAs could also play a role in RCC pathogenesis and progression. This review gives an overview over the current study landscape of circulating ncRNAs and their involvement in RCC pathogenesis as well as their potential utility as future biomarkers in RCC diagnosis and treatment.
Collapse
Affiliation(s)
- Dominik A Barth
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, Graz, Austria.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rares Drula
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Research Centre for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Leonie Ott
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Fabris
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czechia.,Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, Graz, Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
30
|
Bu J, Nair A, Iida M, Jeong WJ, Poellmann MJ, Mudd K, Kubiatowicz LJ, Liu EW, Wheeler DL, Hong S. An Avidity-Based PD-L1 Antagonist Using Nanoparticle-Antibody Conjugates for Enhanced Immunotherapy. NANO LETTERS 2020; 20:4901-4909. [PMID: 32510959 PMCID: PMC7737517 DOI: 10.1021/acs.nanolett.0c00953] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Upregulation of programmed death ligand 1 (PD-L1) allows cancer cells to evade antitumor immunity. Despite tremendous efforts in developing PD-1/PD-L1 immune checkpoint inhibitors (ICIs), clinical trials using such ICIs have shown inconsistent benefits. Here, we hypothesized that the ICI efficacy would be dictated by the binding strength of the inhibitor to the target proteins. To assess this, hyperbranched, multivalent poly(amidoamine) dendrimers were employed to prepare dendrimer-ICI conjugates (G7-aPD-L1). Binding kinetics measurements using SPR, BLI, and AFM revealed that G7-aPD-L1 exhibits significantly enhanced binding strength to PD-L1 proteins, compared to free aPD-L1. The binding avidity of G7-aPD-L1 was translated into in vitro efficiency and in vivo selectivity, as the conjugates improved the PD-L1 blockade effect and enhanced accumulation in tumor sites. Our results demonstrate that the dendrimer-mediated multivalent interaction substantially increases the binding avidity of the ICIs and thereby improves the antagonist effect, providing a novel platform for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ashita Nair
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mari Iida
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Woo-jin Jeong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Michael J. Poellmann
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kara Mudd
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Luke J. Kubiatowicz
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Elizabeth W. Liu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Deric L. Wheeler
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Yonsei Frontier Lab and Department of Pharmacy, Yonsei University, Seoul 03722, Republic of Korea
- Address all correspondence to: Prof. Seungpyo Hong, Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin – Madison, 7121 Rennebohm Hall 777 Highland Avenue, Madison, WI 53705, USA, / phone: (608) 890-0699
| |
Collapse
|