Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Lasers Surg Med ; 50(9): 908-916, 2018 09.
Article in English | MEDLINE | ID: mdl-29799134

ABSTRACT

OBJECTIVES: To measure and compare microvascular responses within the skin of the upper arm to local stimuli, such as heating or rubbing, through the use of optical coherence tomography angiography (OCTA), and to investigate its impact on blood volume collection. MATERIALS AND METHODS: With the use of heat packs or rubbing, local stimulation was applied to the skin of either the left or right upper arm. Data from the stimulated sites were obtained using OCTA comparing pre- and post-stimulation microvascular parameters, such as vessel density, mean vessel diameter, and mean avascular pore size. Additionally, blood was collected using a newly designed collection device and volume was recorded to evaluate the effect of the skin stimulation. RESULTS: Nineteen subjects were recruited for local stimulation study (including rubbing and heating) and 21 subjects for blood drawn study. Of these subjects, 14 agreed to participate in both studies. OCTA was successful in monitoring and measuring minute changes in the microvasculature of the stimulated skin. Compared to baseline, significant changes after local heating and rubbing were respectively found in vessel density (16% [P = 0.0004] and 33% [P < 0.0001] increase), mean vessel diameter (14% and 11% increase) and mean avascular pore size (5% [P = 0.0068] and 8% [P = 0.0005] decrease) after stimulations. A gradual recovery was recorded for each parameter, with no difference being measured after 30 minutes. Blood collection volumes significantly increased after stimulations of heating (48% increase; P = 0.049) and rubbing (78% increase; P = 0.048). Significant correlations were found between blood volume and microvascular parameters except mean avascular pore size under the heating condition. CONCLUSIONS: OCTA can provide important information regarding microvascular adaptations to local stimuli. With that, both heating and rubbing of the skin have positive effects on blood collection capacity, with rubbing having the most significant effect. Lasers Surg. Med. 50:908-916, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Angiography , Dermis/blood supply , Dermis/diagnostic imaging , Microvessels/diagnostic imaging , Physical Stimulation , Tomography, Optical Coherence , Adult , Blood Specimen Collection , Female , Humans , Male , Middle Aged , Upper Extremity , Young Adult
2.
Clin Cancer Res ; 23(3): 746-756, 2017 02.
Article in English | MEDLINE | ID: mdl-27401243

ABSTRACT

PURPOSE: There is a critical clinical need for new predictive and pharmacodynamic biomarkers that evaluate pathway activity in patients treated with targeted therapies. A microscale platform known as VERSA (versatile exclusion-based rare sample analysis) was developed to integrate readouts across protein, mRNA, and DNA in circulating tumor cells (CTC) for a comprehensive analysis of the androgen receptor (AR) signaling pathway. EXPERIMENTAL DESIGN: Utilizing exclusion-based sample preparation principles, a handheld chip was developed to perform CTC capture, enumeration, quantification, and subcellular localization of proteins and extraction of mRNA and DNA. This technology was validated across integrated endpoints in cell lines and a cohort of patients with castrate-resistant prostate cancer (CRPC) treated with AR-targeted therapies and chemotherapies. RESULTS: The VERSA was validated in cell lines to analyze AR protein expression, nuclear localization, and gene expression targets. When applied to a cohort of patients, radiographic progression was predicted by the presence of multiple AR splice variants and activity in the canonical AR signaling pathway. AR protein expression and nuclear localization identified phenotypic heterogeneity. Next-generation sequencing with the FoundationOne panel detected copy number changes and point mutations. Longitudinal analysis of CTCs identified acquisition of multiple AR variants during targeted treatments and chemotherapy. CONCLUSIONS: Complex mechanisms of resistance to AR-targeted therapies, across RNA, DNA, and protein endpoints, exist in patients with CRPC and can be quantified in CTCs. Interrogation of the AR signaling pathway revealed distinct patterns relevant to tumor progression and can serve as pharmacodynamic biomarkers for targeted therapies. Clin Cancer Res; 1-11. ©2016 AACR.

3.
Anal Chem ; 86(13): 6355-62, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24927449

ABSTRACT

Sample preparation is a major bottleneck in many biological processes. Paramagnetic particles (PMPs) are a ubiquitous method for isolating analytes of interest from biological samples and are used for their ability to thoroughly sample a solution and be easily collected with a magnet. There are three main methods by which PMPs are used for sample preparation: (1) removal of fluid from the analyte-bound PMPs, (2) removal of analyte-bound PMPs from the solution, and (3) removal of the substrate (with immobilized analyte-bound PMPs). In this paper, we explore the third and least studied method for PMP-based sample preparation using a platform termed Sliding Lid for Immobilized Droplet Extractions (SLIDE). SLIDE leverages principles of surface tension and patterned hydrophobicity to create a simple-to-operate platform for sample isolation (cells, DNA, RNA, protein) and preparation (cell staining) without the need for time-intensive wash steps, use of immiscible fluids, or precise pinning geometries. Compared to other standard isolation protocols using PMPs, SLIDE is able to perform rapid sample preparation with low (0.6%) carryover of contaminants from the original sample. The natural recirculation occurring within the pinned droplets of SLIDE make possible the performance of multistep cell staining protocols within the SLIDE by simply resting the lid over the various sample droplets. SLIDE demonstrates a simple easy to use platform for sample preparation on a range of complex biological samples.


Subject(s)
Cell Separation/instrumentation , Chemical Fractionation/instrumentation , DNA/isolation & purification , Magnets/chemistry , Proteins/isolation & purification , RNA, Viral/isolation & purification , Animals , Cell Line , Equipment Design , Green Fluorescent Proteins/isolation & purification , HIV/genetics , HIV/isolation & purification , Humans , Luminescent Proteins/isolation & purification , RNA, Viral/genetics , Red Fluorescent Protein
4.
Integr Biol (Camb) ; 6(6): 627-635, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24791272

ABSTRACT

In prostate cancer, bone is a frequent site of metastasis; however, the molecular mechanisms of this tumor tropism remain unclear. Here, we integrate a microfluidic coculture platform with multi-photon imaging based techniques to assess both phenotypic cell behavior and FAD fluorescence intensity and fluorescence lifetime in the same cell. This platform combines two independent assays normally performed with two different cell populations into a single device, allowing us to simultaneously assess both phenotypic cell behavior and enzyme activity. We observed that the osteotropic prostate cancer cell line (C4-2B), when in a coculture with bone marrow stromal cells (MC3T3-E1), has increased protrusive phenotype and increased total and protein-bound FAD compared to its parent cell line (LNCaP). We hypothesized that an increase in ROS-generating APAO activity may be responsible for these effects, and found that the effects were decreased in the presence of the antioxidant N-Acetyl Cysteine (NAC). This suggests that an ROS-related signaling mechanism at the bone metastatic site may be correlated with and play a role in increased invasion of metastasizing prostate cancer cells. The studies performed using this combined platform will lead to new insights into the mechanisms that drive prostate cancer metastasis.


Subject(s)
Flavin-Adenine Dinucleotide/metabolism , Mesenchymal Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Cell Line, Tumor , Coculture Techniques , Humans , Male , Mesenchymal Stem Cells/cytology , Microfluidics , Microscopy, Fluorescence, Multiphoton , Reactive Oxygen Species/antagonists & inhibitors
5.
Lab Chip ; 14(1): 24-31, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24190548

ABSTRACT

The great hope in circulating tumor cell (CTC) research lies in the use of these rare cells as an accessible "fluid biopsy" that would permit frequent, minimally invasive sampling of tumor cells for similar molecular assays that are performed on traditional biopsies. Given the rarity of CTCs in peripheral circulation, microscale methods show great promise and superiority to capture and analyze these cells from patients with solid tumors. Novel technologies that produce validated CTC biomarkers may finally provide medical oncologists the tools needed to provide precise, personalized medical care for patients with advanced cancer. However, few CTC technologies demonstrate both experimental and clinical evidence of an accurate, reliable and reproducible assay that also meets the regulatory requirements to enter routine clinical practice. Many opportunities exist to incorporate clinical needs and regulatory benchmarks into technology development to more quickly garner FDA approval to direct decisions on patient care. This review will address: 1) device development tailored to address predictive, prognostic and/or therapeutic needs across the multitude of malignancies and disease stages; 2) validation benchmarks for clinical assay development; 3) early establishment of standard operating procedures for sample acquisition and analysis; 4) demonstration of clinical utility; 5) clinical qualification of a novel biomarker; and 6) integration of a newly validated and qualified technology into routine clinical practice. Early understanding and incorporation of these regulatory requirements into assay development can simplify and speed the integration of these novel technologies into patient care. Meeting these benchmarks will lead to the true personalization of cancer therapies, directing initial and subsequent treatments for each individual based on initial tumor characteristics while monitoring for emerging mechanisms of resistance in these continually evolving tumors.


Subject(s)
Biomarkers, Tumor/blood , Neoplastic Cells, Circulating/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood , Breast Neoplasms/pathology , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Female , Humans , Male , Prostatic Neoplasms/blood , Prostatic Neoplasms/pathology
6.
Lab Chip ; 14(1): 99-105, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24158597

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths in the United States and worldwide. This has led to major research initiatives focusing on improving early diagnosis rate, as well as the development of pharmacodynamic biomarkers. However, broad clinical integration of these approaches is limited due to the invasive nature of lung biopsies, needle aspirates and resections. Recently, an advance for sampling suspicious lung nodules to collect mini-bronchoalveolar lavage (mBAL) samples was shown to be diagnostically relevant but limited by standard cytology techniques leading to low sensitivity and specificity. In addition, a second non-invasive method that holds great promise is the collection of circulating tumor cells, a rare population of tumor cells that have shed into peripheral circulation from primary or metastatic tumor sites, from blood. Here, we utilize a recently published platform, VerIFAST, for the capture and proteomic analysis of rare cells, to isolate cells of interest from lung cancer patients using both mBAL and blood samples. The VerIFAST platform leverages surface tension at the microscale to pin aqueous and oil fluids in adjacent chambers to create a virtual filter between two aqueous fluids. In this manuscript, the VerIFAST was further enhanced to include oil pinning, which allowed on-device tumbling, further eliminating a laborious and time consuming step that could result in increased sample loss. Finally, we further developed the base assays used in standard histopathologic assays for diagnostic and pharmacodynamic analysis of these rare lung cancer cells. Specifically, we examined thyroid transcription factor-1 (TTF-1) signal intensity, in which loss is associated with more aggressive disease, and epidermal growth factor receptor (EGFR) signal intensity, which is a high value therapeutic target in lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Immunomagnetic Separation/methods , Lung Neoplasms/diagnosis , Microfluidic Analytical Techniques/methods , Neoplastic Cells, Circulating/metabolism , Antibodies, Immobilized/chemistry , Antibodies, Immobilized/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Bronchoalveolar Lavage Fluid/cytology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , ErbB Receptors/immunology , ErbB Receptors/metabolism , Humans , Immunomagnetic Separation/instrumentation , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microfluidic Analytical Techniques/instrumentation , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Oils/chemistry , Proteomics , Thyroid Nuclear Factor 1 , Transcription Factors/immunology , Transcription Factors/metabolism
7.
Anal Chem ; 85(20): 9764-70, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24016179

ABSTRACT

The path from gene (DNA) to gene product (RNA or protein) is the foundation of genotype giving rise to phenotype. Comparison of genomic analyses (DNA) with paired transcriptomic studies (mRNA) is critical to evaluating the pathogenic processes that give rise to human disease. The ability to analyze both DNA and mRNA from the same sample is not only important for biologic interrogation but also to minimize variance (e.g., sample loss) unrelated to the biology. Existing methods for RNA and DNA purification from a single sample are typically time-consuming and labor intensive or require large sample sizes to split for separate RNA and DNA extraction procedures. Thus, there is a need for more efficient and cost-effective methods to purify both RNA and DNA from a single sample. To address this need, we have developed a technique, termed SNARE (Selective Nucleic Acid Removal via Exclusion), that uses pinned oil interfaces to simultaneous purify mRNA and DNA from a single sample. A unique advantage of SNARE is the elimination of dilutive wash and centrifugation processes that are fundamental to conventional methods where sample is typically discarded. This minimizes loss and maximizes recovery by allowing nondilutive reinterrogation of the sample. We demonstrate that SNARE is more sensitive than commercially available kits, robustly and repeatably achieving mRNA and DNA purification from extremely low numbers of cells for downstream analyses. In addition to sensitivity, SNARE is fast, easy to use, and cost-effective and requires no laboratory infrastructure or hazardous chemicals. We demonstrate the clinical utility of the SNARE with prostate cancer circulating tumor cells to demonstrate its ability to perform both genomic and transcriptomic interrogation on rare cell populations that would be difficult to achieve with any current method.


Subject(s)
Chemical Fractionation/methods , DNA/isolation & purification , Microfluidic Analytical Techniques/methods , Base Sequence , Buffers , Cell Line, Tumor , DNA/genetics , Gravitation , Humans , Male , Mutation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Surface Tension
8.
Proc Natl Acad Sci U S A ; 110(25): 10111-6, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23729815

ABSTRACT

Although the field of microfluidics has made significant progress in bringing new tools to address biological questions, the accessibility and adoption of microfluidics within the life sciences are still limited. Open microfluidic systems have the potential to lower the barriers to adoption, but the absence of robust design rules has hindered their use. Here, we present an open microfluidic platform, suspended microfluidics, that uses surface tension to fill and maintain a fluid in microscale structures devoid of a ceiling and floor. We developed a simple and ubiquitous model predicting fluid flow in suspended microfluidic systems and show that it encompasses many known capillary phenomena. Suspended microfluidics was used to create arrays of collagen membranes, mico Dots (µDots), in a horizontal plane separating two fluidic chambers, demonstrating a transwell platform able to discern collective or individual cellular invasion. Further, we demonstrated that µDots can also be used as a simple multiplexed 3D cellular growth platform. Using the µDot array, we probed the combined effects of soluble factors and matrix components, finding that laminin mitigates the growth suppression properties of the matrix metalloproteinase inhibitor GM6001. Based on the same fluidic principles, we created a suspended microfluidic metabolite extraction platform using a multilayer biphasic system that leverages the accessibility of open microchannels to retrieve steroids and other metabolites readily from cell culture. Suspended microfluidics brings the high degree of fluidic control and unique functionality of closed microfluidics into the highly accessible and robust platform of open microfluidics.


Subject(s)
Microfluidic Analytical Techniques , Microfluidics/instrumentation , Microfluidics/methods , Models, Biological , Adrenal Cortex/cytology , Breast Neoplasms/pathology , Capillaries/metabolism , Cell Biology/instrumentation , Cell Line, Tumor , Cell Membrane/physiology , Cell Movement , Collagen Type I/metabolism , Computer Simulation , Female , Humans , Hydrocortisone/analysis , Hydrocortisone/metabolism , Male , Matrix Metalloproteinase Inhibitors/pharmacology , Metabolomics/instrumentation , Metabolomics/methods , Prostatic Neoplasms/pathology , Steroids/analysis , Steroids/metabolism , Toxicology/instrumentation , Toxicology/methods
9.
Methods ; 64(2): 137-43, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23806645

ABSTRACT

Circulating tumor cells (CTCs) exist in the peripheral blood stream of metastatic cancer patients at rates of approximately 1 CTC per billion background cells. In order to capture and analyze this rare cell population, various techniques exist that range from antibody-based surface marker positive selection to methods that use physical properties of CTCs to negatively exclude background cells from a CTC population. However, methods to capture cells for functional downstream analyses are limited due to inaccessibility of the captured sample or labeling techniques that may be prohibitive to cell function. Here, we present a negative selection method that leverages a Microfluidic Cell Concentrator (MCC) to allow collection and analysis of this rare cell population without needing cell adhesion or other labeling techniques to keep the cells within the chamber. Because the MCC is designed to allow collection and analysis of non-adherent cell populations, multiple staining steps can be applied in parallel to a given CTC population without losing any of the population. The ability of the MCC for patient sample processing of CTCs for enumeration was demonstrated with five patient samples, revealing an average of 0.31 CTCs/mL. The technique was compared to a previously published method - the ELISPOT - that showed similar CTC levels among the five patient samples tested. Because the MCC method does not use positive selection, the method can be applied across a variety of tumor types with no changes to the process.


Subject(s)
Cell Separation/methods , Microfluidic Analytical Techniques/methods , Neoplastic Cells, Circulating/pathology , Antigens, Neoplasm/blood , Cell Adhesion Molecules/blood , Cell Count , Enzyme-Linked Immunospot Assay/methods , Epithelial Cell Adhesion Molecule , Humans , Male , Microfluidic Analytical Techniques/instrumentation , Neoplasm Metastasis , Neoplasms/blood
10.
Lab Chip ; 13(3): 391-6, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23223939

ABSTRACT

Isolation and characterization of a specific subset of cells from a large heterogeneous population is necessary for studying rare subpopulations of cells. Existing methods require transfer or wash steps that risk causing loss of the rare cell population of interest. Integrated methods reduce loss, making these methods especially useful for reliable isolation of rare cell populations. In this report, we demonstrate the VerIFAST, a device that builds upon the simplified workflow of the Immiscible Filtration Assisted by Surface Tension (IFAST) to integrate a method for cellular isolation with methods for extra- and intracellular staining. First, a front-end purification step allows cells and unwanted particulates to passively settle out of the operational path of the paramagnetic particles, resulting in good efficiency of capture (>80%) and purity (>70%) with a single virtual wall traverse. Second, a Sieve Chamber is used downstream of the isolation chamber that removes excess unbound paramagnetic particles (PMPs) and performs complex multi-step washing procedures without centrifugation or transfer steps. Further, cellular staining can be performed in the device and is demonstrated for extracellular epithelial cell adhesion molecule (EpCAM), intracellular pan-cytokeratins, and Ki-67.


Subject(s)
Cell Separation/methods , Filtration/instrumentation , Staining and Labeling/methods , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Equipment Design , Filtration/methods , Flow Cytometry , Humans , Immunohistochemistry , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Surface Tension
11.
J Lab Autom ; 18(3): 206-11, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23015519

ABSTRACT

The purification of analytes is an important prerequisite for many analytical processes. Although automated infrastructure has dramatically increased throughput for many of these processes, the upstream analyte purification throughput has lagged behind, partially due to the complexity of conventional isolation processes. Here, we demonstrate automated operation of arrays of a new sample preparation technology--immiscible filtration assisted by surface tension (IFAST). IFAST uses surface tension to position an immiscible liquid barrier between a biological sample and downstream buffer. Paramagnetic particles are used to capture analytes of interest and draw them across the immiscible barrier, thus resulting in purification in a single step. Furthermore, the planarity of the IFAST design enables facile and simultaneous operation of multiple IFAST devices. To demonstrate the application of automation to IFAST, we successfully perform an array of 48 IFAST-based assays to detect the presence of a specific antibody. This assay array uses only a commercial automated liquid handler to load the devices and a custom-built magnet actuator to operate the assays. Automated operation of the IFAST devices resulted in more repeatable results relative to manual operation.


Subject(s)
Antibodies/analysis , Complex Mixtures/chemistry , Filtration/instrumentation , Microarray Analysis/statistics & numerical data , Animals , Antibodies/isolation & purification , Automation, Laboratory , Chemistry Techniques, Analytical , Filtration/methods , High-Throughput Screening Assays , Humans , Magnetic Phenomena , Surface Tension
12.
Sci Transl Med ; 4(141): 141ps13, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22764205

ABSTRACT

Recent insights into circulating tumor cells (CTCs) have been driven by numerous technological innovations aimed at isolating, purifying, and analyzing these rare cells. However, the information density within these cells has yet to be truly accessed and exploited for patient benefit. A device reported by Issadore et al. in this issue of Science Translational Medicine proposes a highly sensitive methodology that may both extend CTC capture to a broader patient population and provide greater understanding of biological targets for personalized medical therapies.


Subject(s)
Biosensing Techniques/methods , Microfluidic Analytical Techniques/methods , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...