1
|
Kumar S, Senapati S, Chang HC. Extracellular vesicle and lipoprotein diagnostics (ExoLP-Dx) with membrane sensor: A robust microfluidic platform to overcome heterogeneity. BIOMICROFLUIDICS 2024; 18:041301. [PMID: 39056024 PMCID: PMC11272220 DOI: 10.1063/5.0218986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024]
Abstract
The physiological origins and functions of extracellular vesicles (EVs) and lipoproteins (LPs) propel advancements in precision medicine by offering non-invasive diagnostic and therapeutic prospects for cancers, cardiovascular, and neurodegenerative diseases. However, EV/LP diagnostics (ExoLP-Dx) face considerable challenges. Their intrinsic heterogeneity, spanning biogenesis pathways, surface protein composition, and concentration metrics complicate traditional diagnostic approaches. Commonly used methods such as nanoparticle tracking analysis, enzyme-linked immunosorbent assay, and nuclear magnetic resonance do not provide any information about their proteomic subfractions, including active proteins/enzymes involved in essential pathways/functions. Size constraints limit the efficacy of flow cytometry for small EVs and LPs, while ultracentrifugation isolation is hampered by co-elution with non-target entities. In this perspective, we propose a charge-based electrokinetic membrane sensor, with silica nanoparticle reporters providing salient features, that can overcome the interference, long incubation time, sensitivity, and normalization issues of ExoLP-Dx from raw plasma without needing sample pretreatment/isolation. A universal EV/LP standard curve is obtained despite their heterogeneities.
Collapse
Affiliation(s)
- Sonu Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Satyajyoti Senapati
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Hsueh-Chia Chang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| |
Collapse
|
2
|
Kumar MA, Baba SK, Sadida HQ, Marzooqi SA, Jerobin J, Altemani FH, Algehainy N, Alanazi MA, Abou-Samra AB, Kumar R, Al-Shabeeb Akil AS, Macha MA, Mir R, Bhat AA. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct Target Ther 2024; 9:27. [PMID: 38311623 PMCID: PMC10838959 DOI: 10.1038/s41392-024-01735-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/20/2023] [Accepted: 12/24/2023] [Indexed: 02/06/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, membranous structures secreted into the extracellular space. They exhibit diverse sizes, contents, and surface markers and are ubiquitously released from cells under normal and pathological conditions. Human serum is a rich source of these EVs, though their isolation from serum proteins and non-EV lipid particles poses challenges. These vesicles transport various cellular components such as proteins, mRNAs, miRNAs, DNA, and lipids across distances, influencing numerous physiological and pathological events, including those within the tumor microenvironment (TME). Their pivotal roles in cellular communication make EVs promising candidates for therapeutic agents, drug delivery systems, and disease biomarkers. Especially in cancer diagnostics, EV detection can pave the way for early identification and offers potential as diagnostic biomarkers. Moreover, various EV subtypes are emerging as targeted drug delivery tools, highlighting their potential clinical significance. The need for non-invasive biomarkers to monitor biological processes for diagnostic and therapeutic purposes remains unfulfilled. Tapping into the unique composition of EVs could unlock advanced diagnostic and therapeutic avenues in the future. In this review, we discuss in detail the roles of EVs across various conditions, including cancers (encompassing head and neck, lung, gastric, breast, and hepatocellular carcinoma), neurodegenerative disorders, diabetes, viral infections, autoimmune and renal diseases, emphasizing the potential advancements in molecular diagnostics and drug delivery.
Collapse
Affiliation(s)
- Mudasir A Kumar
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Sadaf K Baba
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Sara Al Marzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Faisal H Altemani
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh Algehainy
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammad A Alanazi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Rashid Mir
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
3
|
Zhang Y, Tao K, Ding L, Zhao Y. Assessing biomarkers for post-surgical wound healing: A meta-analysis of exosome-based CircRNA in breast cancer recovery. Int Wound J 2024; 21:e14723. [PMID: 38379248 PMCID: PMC10830351 DOI: 10.1111/iwj.14723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/07/2024] [Accepted: 01/13/2024] [Indexed: 02/22/2024] Open
Abstract
To evaluate the diagnostic potential of exosome-based circular RNAs (circRNAs) as biomarkers for wound healing in patients after breast cancer surgery, we conducted a comprehensive meta-analysis of studies that measured exosome-based circRNA levels in breast cancer patients post-surgery. Data sources included several biomedical databases up to April 2023. Two independent reviewers extracted the data and assessed study quality. Sensitivity, specificity and diagnostic odds ratios were synthesized using random-effects model with subgroup analyses performed based on study characteristics. Seventeen studies met the inclusion criteria, encompassing a total of 1234 patients. The pooled sensitivity and specificity of exosome-based circRNA for detecting wound healing complications were 0.85 (95% CI: 0.77-0.91) and 0.83 (95% CI: 0.78-0.88), respectively. The area under the summary receiver operating characteristic (SROC) curve was 0.90, indicating high diagnostic accuracy. Subgroup analyses revealed that diagnostic performance was consistent across studies of different geographic regions and sample types but indicated potential variability related to patient age and study design. Exosome-based circRNA profiles exhibited the high diagnostic accuracy for monitoring wound healing in breast cancer post-operative care. These findings supported the potential utility of circRNA as non-invasive biomarkers for post-surgical recovery. However, variability among studies suggested the need for standardized protocols in biomarker measurement. Future research should focus on longitudinal studies to validate the prognostic value of these biomarkers and investigate their role in personalized patient management.
Collapse
Affiliation(s)
- Yunsuo Zhang
- Department of Breast SurgeryThe Fourth People's Hospital of ZhenjiangZhenjiangChina
| | - Kai Tao
- Department of Breast SurgeryThe Fourth People's Hospital of ZhenjiangZhenjiangChina
| | - Lachun Ding
- Department of InformationThe Fourth People's Hospital of ZhenjiangZhenjiangChina
| | - Yi Zhao
- Department of Breast SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
4
|
Cheng W, Yao Y, Li D, Duan C, Wang Z, Xiang Y. Asymmetrically split DNAzyme-based colorimetric and electrochemical dual-modal biosensor for detection of breast cancer exosomal surface proteins. Biosens Bioelectron 2023; 238:115552. [PMID: 37542978 DOI: 10.1016/j.bios.2023.115552] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/07/2023]
Abstract
Exosomal surface proteins are potentially useful for breast cancer diagnosis and awareness of risk. However, some detection techniques involving complex operations and expensive instrumentation are limited to advance to clinical applications. To solve this problem, we develop a dual-modal sensor combining naked-eye detection and electrochemical assay of exosomal surface proteins from breast cancer. Most of existing sensors rely on aptamers recognizing exosomes and generating amplified signals at the same time, which require well-designed aptamer probes to avoid difficulties in identifying exosomes. In our work, aptamers not bound by the exosomes can serve as complete templates to induce formation of G quadruplexes. The peroxidase activity of the G-quadruplex/hemin DNAzyme catalyze substrates can generate both color and electrochemical signals. The developed dual-modal sensor offers a remarkable capability to differentiate nonmetastatic, metastatic breast cancer patients, and healthy individuals through the analysis of exosomal surface proteins. The sensor's distinctive features, including its universality, simplicity, and cost-effectiveness, position it as a promising diagnostic tool in breast cancer research and clinical practice.
Collapse
Affiliation(s)
- Wenting Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Yanheng Yao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Dayong Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Chengjie Duan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Zhongyun Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Yang Xiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
5
|
Mugoni V, Ciani Y, Quaini O, Tomasini S, Notarangelo M, Vannuccini F, Marinelli A, Leonardi E, Pontalti S, Martinelli A, Rossetto D, Pesce I, Mansy SS, Barbareschi M, Ferro A, Caffo O, Attard G, Di Vizio D, D'Agostino VG, Nardella C, Demichelis F. Integrating extracellular vesicle and circulating cell-free DNA analysis using a single plasma aliquot improves the detection of HER2 positivity in breast cancer patients. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e108. [PMID: 38046436 PMCID: PMC10688391 DOI: 10.1002/jex2.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/19/2023] [Accepted: 08/11/2023] [Indexed: 12/05/2023]
Abstract
Multi-analyte liquid biopsies represent an emerging opportunity for non-invasive cancer assessment. We developed ONCE (One Aliquot for Circulating Elements), an approach for the isolation of extracellular vesicles (EV) and cell-free DNA (cfDNA) from a single aliquot of blood. We assessed ONCE performance to classify HER2-positive early-stage breast cancer (BrCa) patients by combining EV-associated RNA (EV-RNA) and cfDNA signals on n = 64 healthy donors (HD) and non-metastatic BrCa patients. Specifically, we isolated EV-enriched samples by a charge-based (CB) method and investigated EV-RNA and cfDNA by next-generation sequencing (NGS) and by digital droplet PCR (ddPCR). Sequencing of cfDNA and EV-RNA from HER2- and HER2+ patients demonstrated concordance with in situ molecular analyses of matched tissues. Combined analysis of the two circulating analytes by ddPCR showed increased sensitivity in ERBB2/HER2 detection compared to single nucleic acid components. Multi-analyte liquid biopsy prediction performance was comparable to tissue-based sequencing results from TCGA. Also, imaging flow cytometry analysis revealed HER2 protein on the surface of EV isolated from the HER2+ BrCa plasma, thus corroborating the potential relevance of studying EV as companion analyte to cfDNA. This data confirms the relevance of combining cfDNA and EV-RNA for HER2 cancer assessment and supports ONCE as a valuable tool for multi-analytes liquid biopsies' clinical implementation.
Collapse
Affiliation(s)
- Vera Mugoni
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Yari Ciani
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Orsetta Quaini
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Simone Tomasini
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Michela Notarangelo
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Federico Vannuccini
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Alessia Marinelli
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Elena Leonardi
- Unit of Surgical Pathology, Santa Chiara Hospital, APSSTrentoItaly
| | - Stefano Pontalti
- Department of Medical OncologySanta Chiara Hospital, APSSTrentoItaly
| | - Angela Martinelli
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Daniele Rossetto
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Isabella Pesce
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Sheref S. Mansy
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | | | - Antonella Ferro
- Department of Medical OncologySanta Chiara Hospital, APSSTrentoItaly
| | - Orazio Caffo
- Department of Medical OncologySanta Chiara Hospital, APSSTrentoItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | | | - Caterina Nardella
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative BiologyUniversity of TrentoTrentoItaly
| |
Collapse
|
6
|
Vilella-Figuerola A, Cordero A, Mirabet S, Muñoz-García N, Suades R, Padró T, Badimon L. Platelet-Released Extracellular Vesicle Characteristics Differ in Chronic and in Acute Heart Disease. Thromb Haemost 2023; 123:892-903. [PMID: 37075787 DOI: 10.1055/s-0043-57017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs), shed in response to cell activation, stress, or injury, are increased in the blood of patients with cardiovascular disease. EVs are characterized by expressing parental-cell antigens, allowing the determination of their cellular origin. Platelet-derived EVs (pEVs) are the most abundant in blood. Although not universally given, EVs generally express phosphatidylserine (PS) in their membrane. OBJECTIVES To investigate pEVs in chronic and acute conditions, such as chronic heart failure (CHF) and first-onset acute coronary syndrome (ACS), in patients treated as per guidelines. METHODS EVs in CHF patients (n = 119), ACS patients (n = 58), their respective controls (non-CHF [n = 21] and non-ACS [n = 24], respectively), and a reference control group (n = 31) were characterized and quantified by flow cytometry, using monoclonal antibodies against platelet antigens, and annexin V (AV) to determine PS exposure. RESULTS CHF patients had higher EVs-PS- numbers, while ACS had predominantly EVs-PS+. In contrast to ACS, CHF patients had significantly reduced numbers of pEVs carrying PECAM and αIIb-integrin epitopes (CD31+/AV+, CD41a+/AV+, and CD31+/CD41a+/AV+), while no differences were observed in P-selectin-rich pEVs (CD62P+/AV+) compared with controls. Additionally, background etiology of CHF (ischemic vs. nonischemic) or ACS type (ST-elevation myocardial infarction [STEMI] vs. non-STEMI [NSTEMI]) did not affect pEV levels. CONCLUSION PS exposure in EV and pEV-release differ between CHF and ACS patients, with tentatively different functional capacities beyond coagulation to inflammation and cross-talk with other cell types.
Collapse
Affiliation(s)
- Alba Vilella-Figuerola
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Alberto Cordero
- Cardiology Department, Hospital Universitario de San Juan, Alicante, Spain
- Unidad de Investigación en Cardiología, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), València, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Sònia Mirabet
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- Heart Failure Group, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Natàlia Muñoz-García
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Rosa Suades
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program-ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- UAB-Chair Cardiovascular Research, Barcelona, Spain
| |
Collapse
|
7
|
Abdelmohsen K, Herman AB, Carr AE, Henry‐Smith CA, Rossi M, Meng Q, Yang J, Tsitsipatis D, Bangura A, Munk R, Martindale JL, Nogueras‐Ortiz CJ, Hao J, Gong Y, Liu Y, Cui C, Hartnell LM, Price NL, Ferrucci L, Kapogiannis D, de Cabo R, Gorospe M. Survey of organ-derived small extracellular vesicles and particles (sEVPs) to identify selective protein markers in mouse serum. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e106. [PMID: 37744304 PMCID: PMC10512735 DOI: 10.1002/jex2.106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 09/26/2023]
Abstract
Extracellular vesicles and particles (EVPs) are secreted by organs across the body into different circulatory systems, including the bloodstream, and reflect pathophysiologic conditions of the organ. However, the heterogeneity of EVPs in the blood makes it challenging to determine their organ of origin. We hypothesized that small (s)EVPs (<100 nm in diameter) in the bloodstream carry distinctive protein signatures associated with each originating organ, and we investigated this possibility by studying the proteomes of sEVPs produced by six major organs (brain, liver, lung, heart, kidney, fat). We found that each organ contained distinctive sEVP proteins: 68 proteins were preferentially found in brain sEVPs, 194 in liver, 39 in lung, 15 in heart, 29 in kidney, and 33 in fat. Furthermore, we isolated sEVPs from blood and validated the presence of sEVP proteins associated with the brain (DPP6, SYT1, DNM1L), liver (FABPL, ARG1, ASGR1/2), lung (SFPTA1), heart (CPT1B), kidney (SLC31), and fat (GDN). We further discovered altered levels of these proteins in serum sEVPs prepared from old mice compared to young mice. In sum, we have cataloged sEVP proteins that can serve as potential biomarkers for organ identification in serum and show differential expression with age.
Collapse
Affiliation(s)
- Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Allison B. Herman
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Angelica E. Carr
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Charnae’ A. Henry‐Smith
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Martina Rossi
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Qiong Meng
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Jen‐Hao Yang
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Alhassan Bangura
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Jennifer L. Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | | | - Jon Hao
- Poochon ScientificFrederickMarylandUSA
| | - Yi Gong
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Yie Liu
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Chang‐Yi Cui
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| | - Lisa M. Hartnell
- Translational Gerontology Branch, NIA IRPNIHBaltimoreMarylandUSA
| | - Nathan L. Price
- Translational Gerontology Branch, NIA IRPNIHBaltimoreMarylandUSA
| | - Luigi Ferrucci
- Translational Gerontology Branch, NIA IRPNIHBaltimoreMarylandUSA
| | | | - Rafael de Cabo
- Translational Gerontology Branch, NIA IRPNIHBaltimoreMarylandUSA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program (NIA IRP)National Institutes of Health (NIH)BaltimoreMarylandUSA
| |
Collapse
|
8
|
Yang C, Yang Y, Zhao G, Wang H, Dai Y, Huang X. A Low-Cost Microfluidic-Based Detection Device for Rapid Identification and Quantification of Biomarkers-Based on a Smartphone. BIOSENSORS 2023; 13:753. [PMID: 37504151 PMCID: PMC10377552 DOI: 10.3390/bios13070753] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/11/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
The sensitive and rapid detection of microsamples is crucial for early diagnosis of diseases. The short response times and low sample volume requirements of microfluidic chips have shown great potential in early diagnosis, but there are still shortcomings such as complex preparation processes and high costs. We developed a low-cost smartphone-based fluorescence detection device (Smartphone-BFDD) without precision equipment for rapid identification and quantification of biomarkers on glass capillary. The device combines microfluidic technology with RGB image analysis, effectively reducing the sample volume to 20 μL and detection time to only 30 min. For the sensitivity of the device, we constructed a standard sandwich immunoassay (antibody-antigen-antibody) in a glass capillary using the N-protein of SARS-CoV-2 as a biological model, realizing a low limit of detection (LOD, 40 ng mL-1). This device provides potential applications for different biomarkers and offers wide use for rapid biochemical analysis in biomedical research.
Collapse
Affiliation(s)
- Chonghui Yang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Yujing Yang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Gaozhen Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Huan Wang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Yang Dai
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Xiaowen Huang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| |
Collapse
|
9
|
Adduri RSR, Cai K, Velasco‐Alzate K, Vasireddy R, Miller JW, de Frías SP, de Frías FP, Horimasu Y, Iwamoto H, Hattori N, Zhang Y, Gibson KF, Pal AK, Chen Z, Nicastro D, Li L, Cherian S, Sholl LM, Shetty S, Ndetan H, Maeda AH, Ferretto MAP, Hunninghake GM, Schwartz DA, Kass DJ, Rosas IO, Konduru NV. Plasma extracellular vesicle proteins as promising noninvasive biomarkers for diagnosis of idiopathic pulmonary fibrosis. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e98. [PMID: 38939072 PMCID: PMC11080873 DOI: 10.1002/jex2.98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 05/03/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2024]
Abstract
High-resolution computed tomography (HRCT) imaging is critical for diagnostic evaluation of Idiopathic Pulmonary Fibrosis (IPF). However, several other interstitial lung diseases (ILDs) often exhibit radiologic pattern similar to IPF on HRCT making the diagnosis of the disease difficult. Therefore, biomarkers that distinguish IPF from other ILDs can be a valuable aid in diagnosis. Using mass spectrometry, we performed proteomic analysis of plasma extracellular vesicles (EVs) in patients diagnosed with IPF, chronic hypersensitivity pneumonitis, nonspecific interstitial pneumonitis, and healthy subjects. A five-protein signature was identified by lasso regression and was validated in an independent cohort using ELISA. The five-protein signature derived from mass spectrometry data showed an area under the receiver operating characteristic curve of 0.915 (95%CI: 0.819-1.011) and 0.958 (95%CI: 0.882-1.034) for differentiating IPF from other ILDs and from healthy subjects, respectively. Stepwise backwards elimination yielded a model with 3 and 2 proteins for discriminating IPF from other ILDs and healthy subjects, respectively, without compromising diagnostic accuracy. In summary, we discovered and validated EV protein biomarkers for differential diagnosis of IPF in independent cohorts. Interestingly, the biomarker panel could also distinguish IPF and healthy subjects with high accuracy. The biomarkers need to be evaluated in large prospective cohorts to establish their clinical utility.
Collapse
Affiliation(s)
- Raju S. R. Adduri
- Department of Cellular and Molecular BiologyUniversity of Texas Health Science Center at TylerTylerTexasUSA
| | - Kai Cai
- Departments of Cell Biology and BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Karen Velasco‐Alzate
- Department of Cellular and Molecular BiologyUniversity of Texas Health Science Center at TylerTylerTexasUSA
| | - Ravikiran Vasireddy
- Department of Cellular and Molecular BiologyUniversity of Texas Health Science Center at TylerTylerTexasUSA
| | - Jeffrey W. Miller
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Sergio Poli de Frías
- Pulmonary Critical Care Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Internal MedicineMount Sinai Medical CenterMiami BeachFloridaUSA
| | - Fernando Poli de Frías
- Pulmonary Critical Care Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Pulmonary, Critical Care, and Sleep MedicineBaylor College of MedicineHoustonTexasUSA
| | - Yasushi Horimasu
- Department of Molecular and Internal MedicineGraduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal MedicineGraduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Noboru Hattori
- Department of Molecular and Internal MedicineGraduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Yingze Zhang
- Division of Pulmonary, Allergy and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kevin F. Gibson
- Division of Pulmonary, Allergy and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Zhe Chen
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Daniela Nicastro
- Departments of Cell Biology and BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Li Li
- Division of Environmental and Occupational Health SciencesDivision of Pulmonary Sciences and Critical Care MedicineDepartment of MedicineSchool of MedicineUniversity of Colorado DenverDenverColoradoUSA
| | - Sujith Cherian
- Department of Internal MedicineMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Lynette M. Sholl
- Department of PathologyBrigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Sreerama Shetty
- Department of Cellular and Molecular BiologyUniversity of Texas Health Science Center at TylerTylerTexasUSA
| | - Harrison Ndetan
- Department of BiostatisticsSchool of Health ProfessionsUniversity of Texas Health Science Center at TylerTylerTexasUSA
| | - Anthony H. Maeda
- Pulmonary Critical Care Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | | - Gary M. Hunninghake
- Pulmonary Critical Care Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - David A. Schwartz
- Department of MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Daniel J. Kass
- Division of Pulmonary, Allergy and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ivan O. Rosas
- Pulmonary Critical Care Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Pulmonary, Critical Care, and Sleep MedicineBaylor College of MedicineHoustonTexasUSA
| | - Nagarjun V. Konduru
- Department of Cellular and Molecular BiologyUniversity of Texas Health Science Center at TylerTylerTexasUSA
| |
Collapse
|
10
|
Safari F, Kehelpannala C, Safarchi A, Batarseh AM, Vafaee F. Biomarker Reproducibility Challenge: A Review of Non-Nucleotide Biomarker Discovery Protocols from Body Fluids in Breast Cancer Diagnosis. Cancers (Basel) 2023; 15:2780. [PMID: 37345117 DOI: 10.3390/cancers15102780] [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: 02/24/2023] [Revised: 05/02/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Breast cancer has now become the most commonly diagnosed cancer, accounting for one in eight cancer diagnoses worldwide. Non-invasive diagnostic biomarkers and associated tests are superlative candidates to complement or improve current approaches for screening, early diagnosis, or prognosis of breast cancer. Biomarkers detected from body fluids such as blood (serum/plasma), urine, saliva, nipple aspiration fluid, and tears can detect breast cancer at its early stages in a minimally invasive way. The advancements in high-throughput molecular profiling (omics) technologies have opened an unprecedented opportunity for unbiased biomarker detection. However, the irreproducibility of biomarkers and discrepancies of reported markers have remained a major roadblock to clinical implementation, demanding the investigation of contributing factors and the development of standardised biomarker discovery pipelines. A typical biomarker discovery workflow includes pre-analytical, analytical, and post-analytical phases, from sample collection to model development. Variations introduced during these steps impact the data quality and the reproducibility of the findings. Here, we present a comprehensive review of methodological variations in biomarker discovery studies in breast cancer, with a focus on non-nucleotide biomarkers (i.e., proteins, lipids, and metabolites), highlighting the pre-analytical to post-analytical variables, which may affect the accurate identification of biomarkers from body fluids.
Collapse
Affiliation(s)
- Fatemeh Safari
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Cheka Kehelpannala
- BCAL Diagnostics Ltd., Suite 506, 50 Clarence St, Sydney, NSW 2000, Australia
- BCAL Dx, The University of Sydney, Sydney Knowledge Hub, Merewether Building, Sydney, NSW 2006, Australia
| | - Azadeh Safarchi
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
- Microbiomes for One Systems Health, Health and Biosecurity, CSIRO, Westmead, NSW 2145, Australia
| | - Amani M Batarseh
- BCAL Diagnostics Ltd., Suite 506, 50 Clarence St, Sydney, NSW 2000, Australia
- BCAL Dx, The University of Sydney, Sydney Knowledge Hub, Merewether Building, Sydney, NSW 2006, Australia
| | - Fatemeh Vafaee
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
- UNSW Data Science Hub (uDASH), University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
- OmniOmics.ai Pty Ltd., Sydney, NSW 2035, Australia
| |
Collapse
|
11
|
Lee SH, Cha B, Ko J, Afzal M, Park J. Acoustofluidic separation of proteins from platelets in human blood plasma using aptamer-functionalized microparticles. BIOMICROFLUIDICS 2023; 17:024105. [PMID: 37153865 PMCID: PMC10162022 DOI: 10.1063/5.0140096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 05/10/2023]
Abstract
Microfluidic liquid biopsy has emerged as a promising clinical assay for early diagnosis. Herein, we propose acoustofluidic separation of biomarker proteins from platelets in plasma using aptamer-functionalized microparticles. As model proteins, C-reactive protein and thrombin were spiked in human platelet-rich plasma. The target proteins were selectively conjugated with their corresponding aptamer-functionalized microparticles of different sizes, and the particle complexes served as a mobile carrier for the conjugated proteins. The proposed acoustofluidic device was composed of an interdigital transducer (IDT) patterned on a piezoelectric substrate and a disposable polydimethylsiloxane (PDMS) microfluidic chip. The PDMS chip was placed in a tilted arrangement with the IDT to utilize both vertical and horizontal components of surface acoustic wave-induced acoustic radiation force (ARF) for multiplexed assay at high-throughput. The two different-sized particles experienced the ARF at different magnitudes and were separated from platelets in plasma. The IDT on the piezoelectric substrate could be reusable, while the microfluidic chip can be replaceable for repeated assays. The sample processing throughput with the separation efficiency >95% has been improved such that the volumetric flow rate and flow velocity were 1.6 ml/h and 37 mm/s, respectively. For the prevention of platelet activation and protein adsorption to the microchannel, polyethylene oxide solution was introduced as sheath flows and coating on to the walls. We conducted scanning electron microscopy, x-ray photoemission spectroscopy , and sodium dodecyl sulfate- analysis before and after the separation to confirm the protein capture and separation. We expect that the proposed approach will provide new prospects for particle-based liquid biopsy using blood.
Collapse
Affiliation(s)
- Song Ha Lee
- Department of Mechanical Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Beomseok Cha
- Department of Mechanical Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Jeongu Ko
- Department of Mechanical Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Muhammad Afzal
- Center of Immunology Marseille-Luminy, Aix-Marseille University, 171 Av, De Luminy, 13009 Marseille, France
| | - Jinsoo Park
- Department of Mechanical Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| |
Collapse
|
12
|
Lopez K, Lai SWT, Lopez Gonzalez EDJ, Dávila RG, Shuck SC. Extracellular vesicles: A dive into their role in the tumor microenvironment and cancer progression. Front Cell Dev Biol 2023; 11:1154576. [PMID: 37025182 PMCID: PMC10071009 DOI: 10.3389/fcell.2023.1154576] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/10/2023] [Indexed: 04/08/2023] Open
Abstract
Extracellular vesicles (EVs) encompass a diverse set of membrane-derived particles released from cells and are found in numerous biological matrices and the extracellular space. Specific classes of EVs include apoptotic bodies, exosomes, and microvesicles, which vary in their size, origin, membrane protein expression, and interior cargo. EVs provide a mechanism for shuttling cargo between cells, which can influence cell physiology by transporting proteins, DNA, and RNA. EVs are an abundant component of the tumor microenvironment (TME) and are proposed to drive tumor growth and progression by communicating between fibroblasts, macrophages, and tumor cells in the TME. The cargo, source, and type of EV influences the pro- or anti-tumoral role of these molecules. Therefore, robust EV isolation and characterization techniques are required to ensure accurate elucidation of their association with disease. Here, we summarize different EV subclasses, methods for EV isolation and characterization, and a selection of current clinical trials studying EVs. We also review key studies exploring the role and impact of EVs in the TME, including how EVs mediate intercellular communication, drive cancer progression, and remodel the TME.
Collapse
|
13
|
Telekes A, Horváth A. The Role of Cell-Free DNA in Cancer Treatment Decision Making. Cancers (Basel) 2022; 14:6115. [PMID: 36551600 PMCID: PMC9776613 DOI: 10.3390/cancers14246115] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
The aim of this review is to evaluate the present status of the use of cell-free DNA and its fraction of circulating tumor DNA (ctDNA) because this year July 2022, an ESMO guideline was published regarding the application of ctDNA in patient care. This review is for clinical oncologists to explain the concept, the terms used, the pros and cons of ctDNA; thus, the technical aspects of the different platforms are not reviewed in detail, but we try to help in navigating the current knowledge in liquid biopsy. Since the validated and adequately sensitive ctDNA assays have utility in identifying actionable mutations to direct targeted therapy, ctDNA may be used for this soon in routine clinical practice and in other different areas as well. The cfDNA fragments can be obtained by liquid biopsy and can be used for diagnosis, prognosis, and selecting among treatment options in cancer patients. A great proportion of cfDNA comes from normal cells of the body or from food uptake. Only a small part (<1%) of it is related to tumors, originating from primary tumors, metastatic sites, or circulating tumor cells (CTCs). Soon the data obtained from ctDNA may routinely be used for finding minimal residual disease, detecting relapse, and determining the sites of metastases. It might also be used for deciding appropriate therapy, and/or emerging resistance to the therapy and the data analysis of ctDNA may be combined with imaging or other markers. However, to achieve this goal, further clinical validations are inevitable. As a result, clinicians should be aware of the limitations of the assays. Of course, several open questions are still under research and because of it cfDNA and ctDNA testing are not part of routine care yet.
Collapse
Affiliation(s)
- András Telekes
- Omnimed-Etosz, Ltd., 81 Széher Rd., 1021 Budapest, Hungary
- Semmelweis University, 26. Üllői Rd., 1085 Budapest, Hungary
| | - Anna Horváth
- Department of Internal Medicine and Haematology, Semmelweis University, 46. Szentkirályi Rd., 1088 Budapest, Hungary
| |
Collapse
|
14
|
Serhii K, Anastasiia H, Oksana M, Kyrylo L, Liubov S, Nataliia V, Iryna I, Rostyslav S, Alla R. Tristetraprolin expression levels and methylation status in breast cancer. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Wu HJ, Chu PY. Current and Developing Liquid Biopsy Techniques for Breast Cancer. Cancers (Basel) 2022; 14:2052. [PMID: 35565189 PMCID: PMC9105073 DOI: 10.3390/cancers14092052] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer and leading cause of cancer mortality among woman worldwide. The techniques of diagnosis, prognosis, and therapy monitoring of breast cancer are critical. Current diagnostic techniques are mammography and tissue biopsy; however, they have limitations. With the development of novel techniques, such as personalized medicine and genetic profiling, liquid biopsy is emerging as the less invasive tool for diagnosing and monitoring breast cancer. Liquid biopsy is performed by sampling biofluids and extracting tumor components, such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), cell-free mRNA (cfRNA) and microRNA (miRNA), proteins, and extracellular vehicles (EVs). In this review, we summarize and focus on the recent discoveries of tumor components and biomarkers applied in liquid biopsy and novel development of detection techniques, such as surface-enhanced Raman spectroscopy (SERS) and microfluidic devices.
Collapse
Affiliation(s)
- Hsing-Ju Wu
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan;
- Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua 505, Taiwan
- Department of Biology, National Changhua University of Education, Changhua 500, Taiwan
| | - Pei-Yi Chu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
| |
Collapse
|
16
|
Morasso C, Ricciardi A, Sproviero D, Truffi M, Albasini S, Piccotti F, Sottotetti F, Mollica L, Cereda C, Sorrentino L, Corsi F. Fast quantification of extracellular vesicles levels in early breast cancer patients by Single Molecule Detection Array (SiMoA). Breast Cancer Res Treat 2021; 192:65-74. [PMID: 34935096 PMCID: PMC8841315 DOI: 10.1007/s10549-021-06474-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/02/2021] [Indexed: 10/25/2022]
Abstract
PURPOSE Preliminary reports suggest that extracellular vesicles (EVs) might be a promising biomarker for breast cancer (BC). However, the quantification of plasmatic levels of EVs is a complex task. To overcome these limitations, we developed a new, fast, and easy to use assay for the quantification of EVs directly in plasma based on the use of Single-Molecule Array (SiMoA). METHODS By using SiMoA to identify CD9+/CD63+ EVs, we analyzed plasma samples of 181 subjects (95 BC and 86 healthy controls, HC). A calibration curve, made of a serial dilution of lyophilized standards from human plasma, was used in each run to ensure the obtainment of quantitative results from the assay. In a subgroup of patients, EVs concentrations were estimated in plasma before and after 30 days from cancer surgery. Additional information on the size of EVs were also acquired using a Nanosight system to obtain a clearer understanding of the mechanism underlying the releases of EVs associated with the presence of cancer. RESULTS The measured levels of EVs resulted significantly higher in BC patients (median values 1179.1 ng/µl vs 613.0 ng/µl, p < 0.0001). ROC curve was used to define the optimal cut-off level of the test at 1034.5 ng/µl with an AUC of 0.75 [95% CI 0.68-0.82]. EVs plasmatic concentrations significantly decreased after cancer surgery compared to baseline values (p = 0.014). No correlation was found between EVs concentration and clinical features of BC. CONCLUSION SiMoA assay allows plasmatic EVs levels detection directly without any prior processing. EVs levels are significantly higher in BC patients and significantly decreases after cancer surgery.
Collapse
Affiliation(s)
- Carlo Morasso
- Laboratory of Nanomedicine, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Alessandra Ricciardi
- Laboratory of Nanomedicine, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Daisy Sproviero
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Marta Truffi
- Laboratory of Nanomedicine, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Sara Albasini
- Breast Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Francesca Piccotti
- Laboratory of Nanomedicine, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Federico Sottotetti
- Medical Oncology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Ludovica Mollica
- Medical Oncology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Luca Sorrentino
- Colorectal Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Fabio Corsi
- Breast Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy. .,Dipartimento di Scienze Biomediche e Cliniche "L. Sacco", Università di Milano, Via G.B. Grassi, 74, 20157, Milan, Italy.
| |
Collapse
|