Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Semin Nucl Med ; 53(3): 449-456, 2023 05.
Article in English | MEDLINE | ID: mdl-36344325

ABSTRACT

More than 250,000 patients die from Hodgkin or non-Hodgkin lymphoma each year. Currently, molecular imaging with 18F-FDG-PET/CT is the standard of care for lymphoma staging and therapy response assessment. In this review, we will briefly summarize the role of molecular imaging for lymphoma diagnosis, staging, outcome prediction, and prognostication. We discuss future directions in response assessment and surveillance with quantitative PET parameters, the utility of interim assessment, and the differences with response assessment to immunomodulatory therapy. Lastly, we will cover innovations in the field regarding novel tracers and artificial intelligence.


Subject(s)
Hodgkin Disease , Lymphoma , Humans , Hodgkin Disease/therapy , Positron Emission Tomography Computed Tomography/methods , Artificial Intelligence , Fluorodeoxyglucose F18 , Lymphoma/diagnostic imaging , Lymphoma/therapy , Lymphoma/pathology , Positron-Emission Tomography , Molecular Imaging , Neoplasm Staging
2.
MethodsX ; 8: 101414, 2021.
Article in English | MEDLINE | ID: mdl-34430309

ABSTRACT

A miniaturized optoelectronic sensor is demonstrated that measures total protein concentration in serum and urine with sensitivity and accuracy comparable to gold-standard methods. The sensor is comprised of a vertical cavity surface emitting laser (VCSEL), photodetector and other custom optical components and electronics that can be hybrid packaged into a portable, handheld form factor. In conjunction, a custom fluorescence assay has been developed based on the protein-induced fluorescence enhancement (PIFE) phenomenon, enabling real-time sensor response to changes in protein concentration. Methods are described for the following:•Standard curves: Used to determine the sensitivity, dynamic range, and linearity of the VCSEL biosensor/PIFE assay system in buffer as well as in human blood and urine samples.•Comparison of VCSEL biosensor performance with a benchtop fluorimetric microplate reader.•Accuracy of the VCSEL biosensor/PIFE assay system: Evaluated by comparing sensor measurements with gold-standard clinical laboratory measurements of total protein in serum and urine samples from patients with diabetes.

3.
Sci Transl Med ; 13(597)2021 06 09.
Article in English | MEDLINE | ID: mdl-34108250

ABSTRACT

Continuous health monitoring and integrated diagnostic devices, worn on the body and used in the home, will help to identify and prevent early manifestations of disease. However, challenges lie ahead in validating new health monitoring technologies and in optimizing data analytics to extract actionable conclusions from continuously obtained health data.


Subject(s)
Precision Medicine
4.
Biosens Bioelectron ; 180: 112823, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33715946

ABSTRACT

Measurement of total protein in urine is key to monitoring kidney health in diabetes. However, most total protein assays are performed using large, expensive laboratory chemistry analyzers that are not amenable to point-of-care analysis or home monitoring and cannot provide real-time readouts. We developed a miniaturized optoelectronic biosensor using a vertical cavity surface-emitting laser (VCSEL), coupled with a fast protein assay based on protein-induced fluorescence enhancement (PIFE), that can dynamically measure protein concentrations in protein-spiked buffer, serum, and urine in seconds with excellent sensitivity (urine LOD = 0.023 g/L, LOQ = 0.075 g/L) and over a broad range of physiologically relevant concentrations. Comparison with gold standard clinical assays and standard fluorimetry tools showed that the sensor can accurately and reliably quantitate total protein in clinical urine samples from patients with diabetes. Our VCSEL biosensor is amenable to integration with miniaturized electronics, which could afford a portable, low-cost, easy-to-use device for sensitive, accurate, and real-time total protein measurements from small biofluid volumes.


Subject(s)
Biosensing Techniques , Biological Assay , Humans , Lasers , Point-of-Care Systems , Proteins
5.
Clin Cancer Res ; 27(4): 1058-1068, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33087332

ABSTRACT

PURPOSE: Immunomonitoring of chimeric antigen receptor (CAR) T cells relies primarily on their quantification in the peripheral blood, which inadequately quantifies their biodistribution and activation status in the tissues. Noninvasive molecular imaging of CAR T cells by PET is a promising approach with the ability to provide spatial, temporal, and functional information. Reported strategies rely on the incorporation of reporter transgenes or ex vivo biolabeling, significantly limiting the application of CAR T-cell molecular imaging. In this study, we assessed the ability of antibody-based PET (immunoPET) to noninvasively visualize CAR T cells. EXPERIMENTAL DESIGN: After analyzing human CAR T cells in vitro and ex vivo from patient samples to identify candidate targets for immunoPET, we employed a syngeneic, orthotopic murine tumor model of lymphoma to assess the feasibility of in vivo tracking of CAR T cells by immunoPET using the 89Zr-DFO-anti-ICOS tracer, which we have previously reported. RESULTS: Analysis of human CD19-CAR T cells during activation identified the Inducible T-cell COStimulator (ICOS) as a potential target for immunoPET. In a preclinical tumor model, 89Zr-DFO-ICOS mAb PET-CT imaging detected significantly higher signal in specific bone marrow-containing skeletal sites of CAR T-cell-treated mice compared with controls. Importantly, administration of ICOS-targeting antibodies at tracer doses did not interfere with CAR T-cell persistence and function. CONCLUSIONS: This study highlights the potential of ICOS-immunoPET imaging for monitoring of CAR T-cell therapy, a strategy readily applicable to both commercially available and investigational CAR T cells.See related commentary by Volpe et al., p. 911.


Subject(s)
Immunotherapy, Adoptive/methods , Inducible T-Cell Co-Stimulator Protein/metabolism , Lymphoma, Large B-Cell, Diffuse/therapy , T-Lymphocytes/transplantation , Animals , Biological Products/therapeutic use , Cell Line, Tumor , Coculture Techniques , Datasets as Topic , Disease Models, Animal , Humans , Inducible T-Cell Co-Stimulator Protein/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Mice , Mice, Transgenic , Molecular Imaging/methods , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography/methods , RNA-Seq , Receptors, Chimeric Antigen/immunology , Retrospective Studies , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
Cancer Res ; 80(21): 4780-4790, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32900772

ABSTRACT

Graft-versus-host disease (GvHD) is a major complication of allogeneic hematopoietic cell transplantation (HCT), mediated primarily by donor T cells that become activated and attack host tissues. Noninvasive strategies detecting T-cell activation would allow for early diagnosis and possibly more effective management of HCT recipients. PET imaging is a sensitive and clinically relevant modality ideal for GvHD diagnosis, and there is a strong rationale for the use of PET tracers that can monitor T-cell activation and expansion with high specificity. The TNF receptor superfamily member OX40 (CD134) is a cell surface marker that is highly specific for activated T cells, is upregulated during GvHD, and mediates disease pathogenesis. We recently reported the development of an antibody-based activated T-cell imaging agent targeting OX40. In the present study, we visualize the dynamics of OX40 expression in an MHC-mismatch mouse model of acute GvHD using OX40-immunoPET. This approach enabled visualization of T-cell activation at early stages of disease, prior to overt clinical symptoms with high sensitivity and specificity. This study highlights the potential utility of the OX40 PET imaging as a new strategy for GvHD diagnosis and therapy monitoring. SIGNIFICANCE: OX40-immunoPET imaging is a promising noninvasive strategy for early detection of GvHD, capable of detecting signs of GvHD pathology even prior to the development of overt clinical symptoms.


Subject(s)
Graft vs Host Disease/immunology , Positron Emission Tomography Computed Tomography/methods , Radiopharmaceuticals/pharmacology , Receptors, OX40/analysis , T-Lymphocytes , Animals , Antibodies, Monoclonal/pharmacology , Copper Radioisotopes/pharmacology , Lymphocyte Activation , Mice , Receptors, OX40/metabolism , Tissue Distribution
7.
Proc Natl Acad Sci U S A ; 117(23): 12674-12685, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32430322

ABSTRACT

Robust cytotoxic T cell infiltration has proven to be difficult to achieve in solid tumors. We set out to develop a flexible protocol to efficiently transfect tumor and stromal cells to produce immune-activating cytokines, and thus enhance T cell infiltration while debulking tumor mass. By combining ultrasound with tumor-targeted microbubbles, membrane pores are created and facilitate a controllable and local transfection. Here, we applied a substantially lower transmission frequency (250 kHz) than applied previously. The resulting microbubble oscillation was significantly enhanced, reaching an effective expansion ratio of 35 for a peak negative pressure of 500 kPa in vitro. Combining low-frequency ultrasound with tumor-targeted microbubbles and a DNA plasmid construct, 20% of tumor cells remained viable, and ∼20% of these remaining cells were transfected with a reporter gene both in vitro and in vivo. The majority of cells transfected in vivo were mucin 1+/CD45- tumor cells. Tumor and stromal cells were then transfected with plasmid DNA encoding IFN-ß, producing 150 pg/106 cells in vitro, a 150-fold increase compared to no-ultrasound or no-plasmid controls and a 50-fold increase compared to treatment with targeted microbubbles and ultrasound (without IFN-ß). This enhancement in secretion exceeds previously reported fourfold to fivefold increases with other in vitro treatments. Combined with intraperitoneal administration of checkpoint inhibition, a single application of IFN-ß plasmid transfection reduced tumor growth in vivo and recruited efficacious immune cells at both the local and distant tumor sites.


Subject(s)
Immunotherapy/methods , Interferon-beta/genetics , Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , Transfection/methods , Ultrasonic Waves , Animals , Cell Line, Tumor , Cell Membrane/radiation effects , Cell Movement , Humans , Interferon-beta/metabolism , Mice , Microbubbles/therapeutic use , T-Lymphocytes/physiology
8.
Photoacoustics ; 14: 77-98, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31293884

ABSTRACT

Photoacoustic is an emerging biomedical imaging modality, which allows imaging optical absorbers in the tissue by acoustic detectors (light in - sound out). Such a technique has an immense potential for clinical translation since it allows high resolution, sufficient imaging depth, with diverse endogenous and exogenous contrast, and is free from ionizing radiation. In recent years, tremendous developments in both the instrumentation and imaging agents have been achieved. These opened avenues for clinical imaging of various sites allowed applications such as brain functional imaging, breast cancer screening, diagnosis of psoriasis and skin lesions, biopsy and surgery guidance, the guidance of tumor therapies at the reproductive and urological systems, as well as imaging tumor metastases at the sentinel lymph nodes. Here we survey the various clinical and pre-clinical literature and discuss the potential applications and hurdles that still need to be overcome.

9.
Nat Biomed Eng ; 2(9): 696-705, 2018 09.
Article in English | MEDLINE | ID: mdl-30505627

ABSTRACT

The detection and analysis of rare blood biomarkers is necessary for early diagnosis of cancer and to facilitate the development of tailored therapies. However, current methods for the isolation of circulating tumour cells (CTCs) or nucleic acids present in a standard clinical sample of only 5-10 ml of blood provide inadequate yields for early cancer detection and comprehensive molecular profiling. Here, we report the development of a flexible magnetic wire that can retrieve rare biomarkers from the subject's blood in vivo at a much higher yield. The wire is inserted and removed through a standard intravenous catheter and captures biomarkers that have been previously labelled with injected magnetic particles. In a proof-of-concept experiment in a live porcine model, we demonstrate the in vivo labelling and single-pass capture of viable model CTCs in less than 10 s. The wire achieves capture efficiencies that correspond to enrichments of 10-80 times the amount of CTCs in a 5-ml blood draw, and 500-5,000 times the enrichments achieved using the commercially available Gilupi CellCollector.

10.
Mol Imaging Biol ; 20(5): 705-715, 2018 10.
Article in English | MEDLINE | ID: mdl-29916118

ABSTRACT

Intraoperative imaging (IOI) is performed to guide delineation and localization of regions of surgical interest. While oncological surgical planning predominantly utilizes x-ray computed tomography (CT), magnetic resonance imaging (MRI), and ultrasound (US), intraoperative guidance mainly remains on surgeon interpretation and pathology for confirmation. Over the past decades however, intraoperative guidance has evolved significantly with the emergence of several novel imaging technologies, including fluorescence-, Raman, photoacoustic-, and radio-guided approaches. These modalities have demonstrated the potential to further optimize precision in surgical resection and improve clinical outcomes for patients. Not only can these technologies enhance our understanding of the disease, they can also yield large imaging datasets intraoperatively that can be analyzed by deep learning approaches for more rapid and accurate pathological diagnosis. Unfortunately, many of these novel technologies are still under preclinical or early clinical evaluation. Organizations like the Intra-Operative Imaging Study Group of the European Society for Molecular Imaging (ESMI) support interdisciplinary interactions with the aim to improve technical capabilities in the field, an approach that can succeed only if scientists, engineers, and physicians work closely together with industry and regulatory bodies to resolve roadblocks to clinical translation. In this review, we provide an overview of a variety of novel IOI technologies, discuss their challenges, and present future perspectives on the enormous potential of IOI for oncological surgical navigation.


Subject(s)
Diagnostic Imaging/trends , Inventions/trends , Monitoring, Intraoperative/trends , Surgery, Computer-Assisted/trends , Diagnostic Imaging/methods , Humans , Imaging, Three-Dimensional/methods , Imaging, Three-Dimensional/trends , Magnetic Resonance Imaging/methods , Monitoring, Intraoperative/methods , Precision Medicine/methods , Precision Medicine/trends , Surgery, Computer-Assisted/methods , Tomography, X-Ray Computed/methods
11.
Sci Transl Med ; 10(430)2018 02 28.
Article in English | MEDLINE | ID: mdl-29491186

ABSTRACT

Health care systems primarily focus on patients after they present with disease, not before. The emerging field of precision health encourages disease prevention and earlier detection by monitoring health and disease based on an individual's risk. Active participation in health care can be encouraged with continuous health-monitoring devices, providing a higher-resolution picture of human health and disease. However, the development of monitoring technologies must prioritize the collection of actionable data and long-term user engagement.


Subject(s)
Delivery of Health Care/methods , Precision Medicine/methods , Humans
12.
J Clin Invest ; 128(6): 2569-2580, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29596062

ABSTRACT

In situ cancer vaccines are under active clinical investigation, given their reported ability to eradicate both local and disseminated malignancies. Intratumoral vaccine administration is thought to activate a T cell-mediated immune response, which begins in the treated tumor and cascades systemically. In this study, we describe a PET tracer (64Cu-DOTA-AbOX40) that enabled noninvasive and longitudinal imaging of OX40, a cell-surface marker of T cell activation. We report the spatiotemporal dynamics of T cell activation following in situ vaccination with CpG oligodeoxynucleotide in a dual tumor-bearing mouse model. We demonstrate that OX40 imaging was able to predict tumor responses on day 9 after treatment on the basis of tumor tracer uptake on day 2, with greater accuracy than both anatomical and blood-based measurements. These studies provide key insights into global T cell activation following local CpG treatment and indicate that 64Cu-DOTA-AbOX40 is a promising candidate for monitoring clinical cancer immunotherapy strategies.


Subject(s)
Cancer Vaccines/immunology , Copper Radioisotopes/pharmacology , Lymphocyte Activation , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/immunology , Positron-Emission Tomography , Receptors, OX40/immunology , T-Lymphocytes/immunology , Animals , Cancer Vaccines/pharmacology , Cell Line, Tumor , HEK293 Cells , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/therapy , Oligodeoxyribonucleotides/pharmacology
14.
ACS Nano ; 11(11): 10712-10723, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29090896

ABSTRACT

Circulating tumor-derived extracellular vesicles (EVs) have emerged as a promising source for identifying cancer biomarkers for early cancer detection. However, the clinical utility of EVs has thus far been limited by the fact that most EV isolation methods are tedious, nonstandardized, and require bulky instrumentation such as ultracentrifugation (UC). Here, we report a size-based EV isolation tool called ExoTIC (exosome total isolation chip), which is simple, easy-to-use, modular, and facilitates high-yield and high-purity EV isolation from biofluids. ExoTIC achieves an EV yield ∼4-1000-fold higher than that with UC, and EV-derived protein and microRNA levels are well-correlated between the two methods. Moreover, we demonstrate that ExoTIC is a modular platform that can sort a heterogeneous population of cancer cell line EVs based on size. Further, we utilize ExoTIC to isolate EVs from cancer patient clinical samples, including plasma, urine, and lavage, demonstrating the device's broad applicability to cancers and other diseases. Finally, the ability of ExoTIC to efficiently isolate EVs from small sample volumes opens up avenues for preclinical studies in small animal tumor models and for point-of-care EV-based clinical testing from fingerprick quantities (10-100 µL) of blood.


Subject(s)
Biomarkers, Tumor/blood , Early Detection of Cancer , Exosomes/genetics , Ultracentrifugation/methods , Blood Proteins/isolation & purification , Exosomes/chemistry , Extracellular Vesicles/genetics , Humans , MicroRNAs/blood , Neoplasms/blood , Neoplasms/pathology , Neoplastic Cells, Circulating/chemistry , Neoplastic Cells, Circulating/pathology
15.
Nat Rev Mater ; 2(5)2017 May.
Article in English | MEDLINE | ID: mdl-29876137

ABSTRACT

Nanodiagnostics as a field makes use of fundamental advances in nanobiotechnology to diagnose, characterize and manage disease at the molecular scale. As these strategies move closer to routine clinical use, a proper understanding of different imaging modalities, relevant biological systems and physical properties governing nanoscale interactions is necessary to rationally engineer next-generation bionanomaterials. In this Review, we analyse the background physics of several clinically relevant imaging modalities and their associated sensitivity and specificity, provide an overview of the materials currently used for in vivo nanodiagnostics, and assess the progress made towards clinical translation. This work provides a framework for understanding both the impressive progress made thus far in the nanodiagnostics field as well as presenting challenges that must be overcome to obtain widespread clinical adoption.

16.
Proc Natl Acad Sci U S A ; 113(52): E8379-E8386, 2016 12 27.
Article in English | MEDLINE | ID: mdl-27956614

ABSTRACT

Circulating tumor cells (CTCs) are established cancer biomarkers for the "liquid biopsy" of tumors. Molecular analysis of single CTCs, which recapitulate primary and metastatic tumor biology, remains challenging because current platforms have limited throughput, are expensive, and are not easily translatable to the clinic. Here, we report a massively parallel, multigene-profiling nanoplatform to compartmentalize and analyze hundreds of single CTCs. After high-efficiency magnetic collection of CTC from blood, a single-cell nanowell array performs CTC mutation profiling using modular gene panels. Using this approach, we demonstrated multigene expression profiling of individual CTCs from non-small-cell lung cancer (NSCLC) patients with remarkable sensitivity. Thus, we report a high-throughput, multiplexed strategy for single-cell mutation profiling of individual lung cancer CTCs toward minimally invasive cancer therapy prediction and disease monitoring.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/blood , Lung Neoplasms/blood , Neoplastic Cells, Circulating , Adult , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Cell Count , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Leukocyte Common Antigens/blood , Lung Neoplasms/pathology , Male , Microfluidics , Middle Aged , Mutation , Nanotechnology , Reverse Transcriptase Polymerase Chain Reaction , Single-Cell Analysis
17.
Nanomedicine ; 12(2): 287-307, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26707817

ABSTRACT

Nanomedicine, the integration of nanotechnological tools in medicine demonstrated promising potential to revolutionize the diagnosis and treatment of various human health conditions. Nanoparticles (NPs) have shown much promise in diagnostics of cancer, especially since they can accommodate targeting molecules on their surface, which search for specific tumor cell receptors upon injection into the blood stream. This concentrates the NPs in the desired tumor location. Furthermore, such receptor-specific targeting may be exploited for detection of potential metastases in an early stage. Some NPs, such as superparamagnetic iron oxide NPs (SPIONs), are also compatible with magnetic resonance imaging (MRI), which makes their clinical translation and application rather easy and accessible for tumor imaging purposes. Furthermore, multifunctional and/or theranostic NPs can be used for simultaneous imaging of cancer and drug delivery. In this review article, we will specifically focus on the application of SPIONs in early detection and imaging of major cancer types. FROM THE CLINICAL EDITOR: Super-paramagnetic iron oxide nanoparticles (SPIONs) have been reported by many to be useful as an MRI contrast agent in the detection of tumors. To further enhance the tumor imaging, SPIONs can be coupled with tumor targeting motifs. In this article, the authors performed a comprehensive review on the current status of using targeted SPIONS in tumor detection and also the potential hurdles to overcome.


Subject(s)
Contrast Media/chemistry , Ferric Compounds/chemistry , Magnetic Resonance Imaging/methods , Magnets/chemistry , Nanoparticles/chemistry , Neoplasms/diagnosis , Theranostic Nanomedicine/methods , Animals , Drug Delivery Systems/methods , Early Detection of Cancer/methods , Humans , Neoplasm Metastasis/diagnosis , Neoplasms/therapy
18.
ACS Nano ; 9(2): 1868-1877, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25625373

ABSTRACT

Ultrasound imaging is a powerful tool in medicine because of the millisecond temporal resolution and submillimeter spatial resolution of acoustic imaging. However, the current generation of acoustic contrast agents is primarily limited to vascular targets due to their large size. Nanosize particles have the potential to be used as a contrast agent for ultrasound molecular imaging. Silica-based nanoparticles have shown promise here; however, their slow degradation rate may limit their applications as a contrast agent. Phosphate-based glasses are an attractive alternative with controllable degradation rate and easily metabolized degradation components in the body. In this study, biodegradable P2O5-CaO-Na2O phosphate-based glass nanospheres (PGNs) were synthesized and characterized as contrast agents for ultrasound imaging. The structure of the PGNs was characterized using scanning electron microscopy (SEM), energy-dispersive X-ray spectroscopy (EDX), X-ray diffraction (XRD), (31)P magic angle spinning nuclear magnetic resonance ((31)P MAS NMR), and Fourier transform infrared (FTIR) spectroscopy. The SEM images indicated a spherical shape with a diameter size range of 200-500 nm. The XRD, (31)P NMR, and FTIR results revealed the amorphous and glassy nature of PGNs that consisted of mainly Q(1) and Q(2) phosphate units. We used this contrast to label mesenchymal stem cells and determined in vitro and in vivo detection limits of 5 and 9 µg/mL, respectively. Cell counts down to 4000 could be measured with ultrasound imaging with no cytoxicity at doses needed for imaging. Importantly, ion-release studies confirmed these PGNs biodegrade into aqueous media with degradation products that can be easily metabolized in the body.


Subject(s)
Electricity , Glass/chemistry , Mesenchymal Stem Cells/diagnostic imaging , Nanospheres/chemistry , Nanotechnology/methods , Oxides/chemistry , Calcium Compounds/chemistry , Cell Survival , Chemistry Techniques, Synthetic , Contrast Media/chemistry , Contrast Media/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Nanospheres/metabolism , Oxides/metabolism , Phosphorus Compounds/chemistry , Sodium Compounds/chemistry , Temperature , Ultrasonography
20.
Lab Chip ; 10(22): 3157-62, 2010 Nov 21.
Article in English | MEDLINE | ID: mdl-20924527

ABSTRACT

We describe an automated, self-powered chip based on lateral flow immunoassay for rapid, quantitative, and multiplex protein detection from pinpricks of whole blood. The device incorporates on-chip purification of blood plasma by employing inertial forces to focus blood cells away from the assay surface, where plasma proteins are captured and detected on antibody "barcode" arrays. Power is supplied from the capillary action of a piece of adsorbent paper, and sequentially drives, over a 40 minute period, the four steps required to capture serum proteins and then develop a multiplex immunoassay. An 11 protein panel is assayed from whole blood, with high sensitivity and high reproducibility. This inexpensive, self-contained, and easy to operate chip provides a useful platform for point-of-care diagnoses, particularly in resource-limited settings.


Subject(s)
Blood Proteins/analysis , Microfluidic Analytical Techniques/instrumentation , Blood Proteins/metabolism , DNA/chemistry , Enzyme-Linked Immunosorbent Assay/instrumentation , Enzyme-Linked Immunosorbent Assay/methods , Humans , Image Processing, Computer-Assisted , Immunoassay/methods , Microfluidic Analytical Techniques/methods , Oligonucleotide Array Sequence Analysis , Point-of-Care Systems , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...