Extracellular vesicles (EVs), including exosomes (EXOs) and microvesicles (MVs), have become a research subject of great excitement as a potential source of biomarkers in medicine. EVs are membrane bound vesicles, with EXOs derived from multivesicular bodies and MVs from plasma membrane. EVs carrying molecular constituents including proteins and nucleic acids of their originating cells represent an important mode of intercellular communication. A growing body of research has shown that cancer-derived EVs can transfer oncogenic activity and regulate angiogenesis, immunity, and metastasis to promote tumorigenesis and progression. EVs are present in various body fluids, such as blood, urine, saliva, and cerebrospinal fluids. Probing tumor-derived EVs in body fluids can therefore offer a non-invasive way to diagnose cancer, assess cancer progression, and monitor treatment responses.
The clinical use of EVs as cancer biomarkers has been limited by certain technical challenges. One such challenge is molecular detection and analysis of EVs. Due to their small size, EVs cannot be histologically examined using routine optical imaging, and they cannot be analyzed by traditional flow cytometry because of size limits (>200 nm). Western blot, enzyme-linked immunosorbent assays (ELISA), and mass spectrometry are commonly used to analyze EV proteins. These traditional approaches are impractical for longitudinal studies and clinical use because they are time-consuming, labor-intensive, and require relatively large amounts of samples. Despite recent advancements, technically simple, low cost, portable, rapid, efficient, sensitive, and specific technologies for EV molecular detection and surface protein analysis are needed.
As described herein, the present invention features compositions and methods related to the detection and profiling of extracellular vesicles (e.g., exosomes, microvesicles) using the Raman Extracellular Vesicle Assay (REVA) that is technically simple, inexpensive, portable, rapid, efficient, highly sensitive, and highly specific. The method involves the use of highly sensitive and specific surface enhanced Raman scattering (SERS) nanotags (e.g. SERS gold nanorod (AuNR) tags) to detect and quantify surface proteins on membrane bound vesicles that are captured on a substrate (e.g., an array, Au-coated glass microscope slide, bead). The invention features the first application of SERS nanotags in the analysis of membrane bound vesicles from any biological sample (e.g., any cell or tissue, including body fluids, such as blood, urine, saliva, cerebrospinal fluid), or from any biological source (e.g., a human or non-human mammal).
In some embodiments, REVA is performed in at least two different ways, referred to herein as direct REVA (dREVA) and capture REVA (cREVA). In dREVA, EVs are immobilized on a lipophilic substrate, labeled with target-specific SERS nanotags (e.g. antibody-conjugated SERS AuNR tags), and detected with a portable Raman spectrometer. In cREVA, EVs are captured on target-specific substrate (e.g. antibody-conjugated Au-coated glass microscope slide), labeled with SERS nanotags (e.g. SERS AuNRs), and detected with a portable Raman spectrometer.
Some aspects of the present disclosure provide a lipophilic substrate comprising an amphiphilic polymer having a thiolated hydrophilic portion and a hydrophobic tail covalently bound to a silver or gold film, wherein the film is fixed to a solid support. In some aspects, a lipophilic substrate is provided that comprises 1,2-distearoyl-sn-glycero-3-phosphoethanolamine conjugated polyethylene glycol thiol (DSPE-PEG-SH) and 11-mercaptoundecyl tetra (ethylene glycol) (MU-TEG) covalently bound to a gold film, wherein the film is fixed to a solid support. In some embodiments, the solid support can be a microscope slide, membrane, or wafer. In some embodiments, the film is optically transparent or opaque, and in some embodiments, the film is gold or silver.
In some aspects, an array device is provided comprising a planar substrate that has an amphiphilic polymer containing a thiolated hydrophilic portion and a hydrophobic tail covalently bound to a film. The film is fixed to a planar support, and a flexible array interface contacts the planar substrate. The interface comprises a plurality of holes. A rigid array template comprising a plurality of holes is in contact with the interface, and the holes of the interface and the holes of the array are aligned.
Provided herein is an array device comprising a planar substrate comprising 1,2-distearoyl-sn-glycero-3-phosphoethanolamine conjugated polyethylene glycol thiol (DSPE-PEG-SH) and 11-mercaptoundecyl tetra (ethylene glycol) (MU-TEG) that is covalently bound to gold film. The film is fixed to the planar substrate, which is in contact with a flexible array interface that comprises a plurality of holes. A rigid array template in contact with the interface also comprises a plurality of holes, and the holes of the interface and the holes of the array are aligned. In some embodiments, the planar substrate is a glass plate or silicon wafer, and in some embodiments, the flexible array interface comprises rubber or silicone. In some embodiments the rigid array template comprises plastic or resin.
The array device comprises wells, wherein each well is at least 1 mm in diameter and the inter-well distance is at least 0.5 mm. The substrate, interface, and template are arranged to form fluid-tight wells.
Also provided herein are surface-enhanced Raman scattering nanotags. The nanotag comprising a plasmonic nanoparticle, a 16-mercaptohexadecanoic acid-linked polyethylene glycol covalently bound at the thiol terminal to a surface of the nanoparticle, an antibody bound to the PEG thiol with the thiol terminal bound to a surface of the nanoparticle, and a Raman reporter that is incorporated into the MHDA pocket on the surface of the nanoparticle. In some embodiments, the Raman reporter is an organic or inorganic dye, and in some embodiments the organic dye is selected from QSY21, IR820, IR783, BHQ, QXL 680, and DTTC. The inorganic dye may be a pyridine or aminothiophenol. In some embodiments, the Raman reporter is QSY21.
In some embodiments, the nanoparticle is gold or silver. In some embodiments, the nanoparticle is a core-shell nanoparticle. The core-shell nanoparticle can be a magnetic-metallic core-shell nanoparticle.
In some embodiments, the Raman reporter that is incorporated into the MHDA pocket is on the surface of a carbon nanosphere or nanotube. The nanoparticle is a gold or silver nanorod in some embodiments and can be between 10 nm and 100 nm.
Another aspect of the present disclosure provides a surface-enhanced Raman scattering nanotag comprising a plasmonic nanoparticle, a Raman reporter and a cetyltrimethylammonium bromide (CTAB) bilayer. And in some aspects, a surface-enhanced Raman scattering nanotag is provided that comprises a plasmonic nanoparticle, a Raman reporter and cetyltrimethylammonium bromide (CTAB) bilayer. The Raman reporter in some embodiments is QSY21.
Methods are also provided in the present disclosure. For example, one aspect provides a method for producing a target-specific capture array, the method comprising providing a device comprising a planar substrate comprising an amphiphilic polymer containing a thiolated hydrophilic segment and a hydrophobic tail covalently bound to a film. The film is fixed to the planar support. A flexible array interface is in contact with the planar substrate, and the interface comprises a plurality of holes. A rigid array template is in contact with the interface, and the rigid array comprises a plurality of holes. The holes of the interface and the holes of the array are aligned, thereby forming a well. Lastly, the method comprises depositing a target-specific capture molecule into each well of the array, thereby forming a capture array. In some embodiments, the capture molecule is an antibody, a single-chain antibody, a nanobody, or an aptamer, and the capture molecule specifically binds an antigen of interest.
Methods are also provided for producing an array device comprising a plurality of cells or membrane bound vesicles, the method comprising providing an array device comprising a planar substrate comprising 1,2-distearoyl-sn-glycero-3-phosphoethanolamine conjugated polyethylene glycol thiol (DSPE-PEG-SH) and 11-mercaptoundecyl tetra (ethylene glycol) (MU-TEG) covalently bound to a gold film in each well. The film is fixed to the planar substrate, and a flexible array interface is in contact with the planar substrate, wherein the interface comprises a plurality of holes. A rigid array template is in contact with the interface, and the rigid array comprises a plurality of holes. The holes of the interface and the holes of the array are aligned thereby forming a well. A cell or membrane bound vesicle is deposited into each well of the array device. In some embodiments, the cell is a cancer cell, blood cell, bacterial cell, epithelial cell, or a parasitic cell. In some embodiments, the membrane bound vesicle is an exosome, microvesicle, an oncosome, microsome, or cellular organelle. Some aspects of the present disclosure contemplate an array device comprising a cell or membrane bound vesicle produced as described supra.
A method is also provided for characterizing biomarkers on a plurality of cells or membrane bound vesicles, the method comprising contacting an array device with a nanotag of claim and detecting a biomarker present on the cell or membrane bound vesicle using Raman spectroscopy. The membrane bound vesicle is an exosome, microvesicles, oncosome, microsome, or cellular organelle.
Other aspects provide a method for characterizing biomarkers on a plurality of cells or membrane bound vesicles, the method comprising contacting the array device described supra with a sample comprising a cell or membrane bound vesicle under conditions suitable for binding. The bound cell or membrane bound vesicle is contacted with a nanotag, and a biomarker present on the cell or membrane bound vesicle is detected using Raman spectroscopy. In some embodiments, the membrane bound vesicle is an exosome, microvesicles, oncosome, microsome, or cellular organelle.
The present disclosure also provides methods for characterizing disease in a subject, the method comprising obtaining a biological sample comprises an extracellular vesicle from the subject and contacting a lipophilic substrate or an array device as disclosed herein with the biological sample under conditions suitable for binding a cell or membrane bound vesicle to the substrate or array device. The bound extracellular vesicle is contacted with a nanotag, and a biomarker present on the cell or membrane bound vesicle is detected using Raman spectroscopy.
A method is also provided for characterizing a disease in a subject, the method comprising obtaining a biological sample from the subject, wherein the sample comprises an extracellular vesicle. The array device is contacted with the biological sample under conditions suitable for binding the extracellular vesicle to the array device, and the bound extracellular vesicle is contacted with a nanotag. A biomarker present on the membrane bound vesicle is detected using Raman spectroscopy. In some embodiments, the biological sample is cell culture media, urine, blood, serum, plasma, cerebral spinal fluid, saliva, or ascites.
A method for characterizing biomarkers on a single membrane bound vesicle is also provide as an aspect of the present disclosure. The method comprises contacting a membrane bound vesicle with a nanotag and exposing the membrane bound vesicle to a light source. A dark field image is acquired of the membrane bound vesicle, and this image serves as a mask to localize the membrane bound vesicle. The localized membrane bound vesicle to a wavelength sufficient to elicit a signal from the nanotag and the brightness of the signal from the nanotag is detected. The presence or absence of a biomarker on the membrane bound vesicle is detected using Raman spectroscopy, thereby characterizing the exosome. In some embodiments, the membrane bound vesicle is an exosome. In some embodiments, the method further comprises contacting the exosome with an antibody that specifically binds an antigen associated with an exosome. In some embodiments, the antibody is conjugated to a polyethylene glycol thiol (PEG-SH) moiety, and the thiol portion of the PEG-SH moiety of the antibody is bound to a functionalized surface of an array or particle. The array, in some embodiments, has multiple wells comprising the bound antibody that specifically binds an antigen associated with an exosome. And in some embodiments of the present method, the array is contacted with a sample comprising exosomes, the exosomes are captured in the array wells.
Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, the following terms have the meanings ascribed to them below, unless specified otherwise.
By “alteration” is meant a change (increase or decrease) in an analyte as detected by methods such as those described herein. In one embodiment, the alteration is in the level of a protein biomarker present on a membrane bound vesicle. As used herein, an alteration includes a 10% change in expression levels, preferably a 25% change, more preferably a 40% change, and most preferably a 50% or greater change in expression levels.
In this disclosure, “comprises,” “comprising,” “containing” and “having” and the like can have the meaning ascribed to them in U.S. Patent law and can mean “includes,” “including,” and the like; “consisting essentially of” or “consists essentially” likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments.
“Detect” refers to characterizing the presence, absence or amount of the analyte to be detected.
By “detectable label” is meant a composition that when linked to a molecule of interest renders the latter detectable, via spectroscopic, photochemical, biochemical, immunochemical, or chemical means. For example, useful labels include radioactive isotopes, magnetic beads, metallic beads, colloidal particles, fluorescent dyes, electron-dense reagents, enzymes (for example, as commonly used in an ELISA), biotin, digoxigenin, or haptens.
By “disease” is meant any condition or disorder that damages or interferes with the normal function of a cell, tissue, or organ. Exemplary diseases that can be evaluated using a method of the invention include, but are not limited to, cancer and neurodegenerative diseases.
By “extracellular vesicle” is meant a membrane bound vesicle that is present extracellularly. Exemplary extracellular vesicles include exosomes and microvesicles.
By “marker” is meant any protein or polynucleotide having an alteration in expression level or activity that is associated with a disease or disorder.
By “mask” is meant an image that only includes pixels that match certain criteria, and subsequent analysis can be directed only to those areas on the mask image. By “membrane bound vesicle” (MBV or MBVs) is meant any vesicle comprising a membrane structure. Exemplary membrane bound vesicles include, but are not limited to, extracellular vesicles (EV or EVs), microvesicles (MV or MVs), exosomes, and apoptotic bodies.
By “Raman Spectroscopy” is meant the spectroscopic technique used to observe vibrational, rotational, and other low-frequency modes in a system. Raman spectroscopy has been commonly used in chemistry to provide a structural fingerprint by which molecules are identified.
By “surface enhanced Raman scattering spectroscopy” is meant the spectroscopic technique in which the Raman scattering signals of a small organic molecule, such as an organic dye, are enhanced by a plasmonic nanoparticle when the small organic molecule is present on, or close to, the surface of the plasmonic nanoparticle. Surface enhanced Raman scattering spectroscopy has been commonly used in chemistry to provide a structural fingerprint of the small organic molecules, or to detect a target with the use of surface enhanced Raman scattering nanotags.
By “surface enhanced Raman scattering nanotags” is meant a plasmonic nanoparticle coated with a Raman reporter. In one embodiment, the plasmonic nanoparticle is a silver or gold nanoparticles surrounded by a metal oxide shell containing a fluorophore. Surface enhanced Raman scattering nanotags provide for the detection and quantification of a target of interest via specific binding of the surface enhanced Raman scattering nanotags and the target of interest. This allows a unique “fingerprint” to be generated that includes the signals of the Raman reporters present on the target.
By “reduces” is meant a negative alteration of at least 10%, 25%, 50%, 75%, or 100%.
By “reference” is meant a standard or control condition.
By “subject” is meant a mammal, including, but not limited to, a human or non-human mammal, such as a bovine, equine, canine, ovine, or feline.
Unless specifically stated or obvious from context, as used herein, the term “or” is understood to be inclusive. Unless specifically stated or obvious from context, as used herein, the terms “a”, “an”, and “the” are understood to be singular or plural.
Unless specifically stated or obvious from context, as used herein, the term “about” is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. About can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05%, or 0.01% of the stated value. Unless otherwise clear from context, all numerical values provided herein are modified by the term about.
The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable or aspect herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.
Any compositions or methods provided herein can be combined with one or more of any of the other compositions and methods provided herein.
This invention features a transformative technology for the detection and quantitative surface protein profiling of extracellular vesicle (EV or EVs) (e.g. exosome (EXO or EXOs), microvesicle (MV or MVs), apoptotic body) using surface enhanced Raman scattering (SERS) nanotags.
This technology, named Raman Extracellular Vesicle Assay (REVA), features the use of highly sensitive and highly specific surface enhanced Raman scattering gold nanorod (SERS AuNR) tags to label EVs and quantitatively detect EV surface proteins with SERS spectroscopy. The assay is advantageously efficient and can be used in combination with a low cost portable EV array device that provides for the analysis of the molecular expression pattern of target-specific surface proteins present on EVs and other membrane-bound vesicles from any biological sample (e.g., any cell or tissue, including body fluids, such as blood, urine, saliva, cerebrospinal fluid), or from any biological source (e.g., a human or non-human mammal). REVA may be used to detect many types of diseases (e.g., cancer, neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease) and characterize the molecular expression patterns of proteins from any biological sample. REVA provides the first application of SERS nanotags for the analysis of EVs and membrane bound vesicles.
REVA involves four major components: (1) extracellular vesicles (EVs) (or any other membrane bound vesicles (MBVs), cell, bacteria, virus, or similar particle isolated from a biological sample; (2) a device that immobilizes or captures EVs in a multiplex fashion (“EV array device”); (3) a labelling agent (e.g., Raman reporter) that provides for EV detection by SERS spectroscopy; and (4) a Raman spectrometer that collect signals. Depending on how the EVs are labeled with the labeling agent, the REVA is typically performed by direct Raman Extracellular Vesicle Assay (dREVA) (
Combining SERS detection with high sensitivity and specificity, and with an EV array device having high portability and high efficiency, allows for the innovative REVA technology to perform dozens of tests on a single palm size device from microliter sized samples with high sensitivity. For example, as described below, dREVA can detect EXOs at a concentration of 1×106 EXO/mL that is over 1000 times lower than the concentration of EXOs in human plasma (≥109 EXO/mL). This easy-to-operate, low cost, portable, efficient, highly sensitive, and highly specific REVA technology will facilitate molecular analysis of EVs, especially EXOs, and is useful in basic and clinical EV research, not only for marker discovery, but for providing insights into the role of EVs in disease development. It will open new avenues for developing new generation cancer liquid biopsy to diagnose cancer, monitor cancer progression, and monitor patient treatment responses in real-time. The REVA technology can be used world-wide, especially in limited-resource research and clinical environments and will advantageously impact cancer diagnostics and personalized treatment.
Another feature of the invention is the use of high throughput 3D printing technology to print a protein array to capture membrane bound vesicles in a target-specific manner on a functionalized gold chip, and label and detect membrane bound vesicles in a high throughput fashion with highly sensitive surface enhanced Raman scattering (SERs) small gold nanorods. This simple, inexpensive, and portable assay offers dozens of test sites on a single palm size chip from microliter samples within two hours, with an unprecedented limit of detection. For example, as described below, the methods have a limit of detection down to 200 exosomes.
The invention provides the first application of surface enhanced Raman scattering (SERS) nanotags for EV analysis. SERS provides for the enhancement of Raman signals of small organic molecules by roughened metallic surface via electromagnetic and chemical mechanism (K. Kneipp et al. J. Phys. Condens. Matter 2001, 14, R597). It can be used to detect EXO molecular constitutes, such as protein, carbohydrates and lipids by enhancing the Raman signals of the molecular constitutes of EXOs (L. Tirinato et al. Microelectron. Eng. 2012, 97, 337; C. Lee et al. Nanoscale 2015, 7, 9290; S. Stremersch et al. Small 2016, 12(24), 3292; J. Park et al. Anal. Chem. 2017, 89, 6695). In contrast, this invention features the use of SERS nanotags for quantitative surface protein profiling of EVs. SERS nanotags are plasmonic nanoparticles (e.g. gold and silver nanoparticles), such as gold nanoparticles coated with Raman reporters such as organic dyes. SERS nanotags provide for the highly sensitive detection of targets of interest with a known SERS spectrum of the Raman reporter (Y. Wang et al. Chem. Rev. 2013, 113(13), 1391). For example, circulating tumor cells in whole blood can be detected at a LOD of 1-2 cell/mL blood using iron oxide-gold core-shell nanoparticles carrying QSY21 reporter (S. Bhana et al. Nanomedicine(Lond) 2014, 9(5), 593). This high sensitivity is due to the strong Raman enhancement of the Raman reporter by the plasmonic nanoaprticles and the abundacy of the Raman reporters on the plasmonic nanoparticles.
Compared to current methods for surface protein analysis of EVs including surface plasmon resonance sensing (SPR technique) (H. Im et al., Nat. Biotechnol. 2014, 32(5), 490; L. Grasso et al., Anal. Bioanal. Chem. 2015, 407, 5425; A. A. I. Sina et al., Sci. Rep. 2016, 6, 30460; A. Thakur et al, Bioelectron. 2017, 94, 400) and resonance light scattering sensing (K. Liang et al. Nat. Biomed. Engineer. 2017, 1, 0021), the use of SERS nanotags for detection has at least two major advantages. First, data analysis is extremely simple. SERS provides fingerprint signals that distinguish interferences from biological background. The SERS spectrum only requires a simple baseline correction using a multi-segment polynomial fitting to subtract SERS background (broad continuum emission). This baseline correction is usually incorporated in the signal correction software and thus the as-acquired spectrum does not need further signal separation process for quantitative analysis. The peak intensity of the SERS spectrum from the Raman report is used to express the level of target protein on EVs. Second, signal collection is extremely fast (e.g., about a second) due to the high sensitivity of the SERS nanotags. For example, signals from 50 samples on a single device can be collected within about 1 minute, which is extremely fast and efficient.
An example of the plasmonic nanoparticles is anisotropic small gold nanorods (
Another feature of the invention is an EV device that allows for simultaneous processing and detection of multiple samples. An example of this EV device was fabricated with an Au slide and a template array (
In dREVA, EVs are immobilized on an array using a lipophilic chemical layer on the device and then labeled and detected using SERS nanotags. Lipophilic molecules with an alkyl chain have high affinity for the lipid bilayer of molecules (e.g., EVs, cells, organelles, membranes) through hydrophobic interactions between the lipid membrane of the target and the lipophilic molecules on the substrate. This invention features lipophilic molecule 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-conjugated polyethylene glycol thiol (DSPE-PEG-SH, MW 5000) combined with a hydrophilic short chain of 11-mercaptoundecyl tetra(ethylene glycol) (MU-TEG) (
In cREVA, EVs are captured on the array device by fixing target specific capture molecules (e.g., ligands), such as antibodies, on the Au surface of the device. For example, the antibodies can be conjugated to a PEG-SH linker in advance via an amide bond by linking commercially available HS-PEG-NHS MW5000 (e.g. Nanocs Inc.) with antibodies. The HS-PEG-Ab binds to the Au slide surface via Au—S bond, leaving external antibodies for specific recognition of the surface proteins on EVs. After functionalization with HS-PEG-Ab, the Au slide is then saturated with MU-TEG to minimize nonspecific interactions (
Labelling EVs with SERS AuNR Tags
In dREVA, the immobilized EVs via lipophilic capture were labeled with target-specific SERS AuNRs with QSY21 dye as the reporter (
In cREVA, the ligand-captured EVs are labeled with SERS AuNR tags via electrostatic interactions of SERS AuNRs and the lipid membrane of EVs (
Signal Collection with a High-Performance Raman System
In some embodiments, a Raman spectrometer can be used for signal collection from the SERS nanotags attached on EVs. Any Raman spectrometer or Raman microscope can be used for signal collection. In some embodiments, a Raman spectrometer is portable, low cost and high throughput. An example of such Raman system is ProRaman-L high performance spectrometer from TSI (
In some aspects of the present disclosure, methods for detecting single vesicles are provided that use single vesicle technology (SVT), which is based on surface enhanced Raman scattering (SERS) imaging to probe tumor-derived exosomes in the presence of non-tumor exosomes. This approach is referred to as SERS-SVT. In some embodiments, small SERS gold nanorod (AuNR) tags are used to label targeted surface protein markers on exosomes that will be captured directly from body fluids. Dark field imaging is used to localize the captured exosomes in a multi-well chamber slide and SERS imaging is used to detect the proteins on single exosomes. By analyzing the dark field mask image and the SERS target image, the expression profile of targeted proteins may be obtained that informs the amount and the protein level of the exosome subpopulation positive to the targeted protein. SVT is much more sensitive and provide valuable information that is not available in current bulk methods. SVT can identify cancer-derived EXOs that are undetectable by current bulk methods, thereby detecting cancer early. SVT can quantify the fraction of tumor-derived EXOs, which is critical in monitoring tumor progression. Further, SVT can reveal EXO subpopulations and discern compositional heterogeneity, which are very useful to understand tumor heterogeneity and help personalized treatment.
Tetraspanin CD81 is an EXO marker that differentiates EXOs from other types of extracellular vesicles; therefore, CD81 antibody can be used to capture EXOs from a biofluid. Other markers can be used to isolate EXOs including, but not limited to, ALIX, TSG101, and other tetraspanins such as CD63 and CD9. The method can directly capture EXOs with, for example, monoclonal antibodies from plasma and other biofluids without EXO pre-purification. In some embodiments, the antibody is conjugated to a polyethylene glycol thiol (PEG-SH) linker (MW=5000) by reacting HS-PEG-NHS with antibody. In some embodiments, the antibody conjugated to the linker may be purified by filtration centrifugation.
In some embodiments, capture of EXOs comprises immersing a chamber slide having an Au surface in composition comprising an antibody that specifically binds an antigen associated with an exosome, wherein the antibody is linked with PEG-SH. In some embodiments, this step is followed by a wash step with PBS. The chamber slide may then be immersed in a composition comprising an agent that inhibits or reduces nonspecific binding to the slide. In some embodiments, the agent is 11-mercaptoundecyl tetra (ethylene glycol) (MU-TEG). To capture the EXOs in a sample, the sample is incubated on the chip for a sufficient period of time to capture the EXOs in the sample. In some embodiments, the incubation period is about 2 hours. After immobilization, EXOs can be visualized with membrane staining agent such as DiO and DIB.
In some embodiments, the profiling of MBVs and/or EVs may be used as a diagnostic tool. Subjects having or at risk of developing a disease are diagnosed using any method known in the art. In particular embodiments, a subject is identified as being at risk to develop the disease. For example, the molecular profiling of labelled MBVs and/or EVs on a Raman spectrometer of a sample may be used to determine a subject who is at risk of acquiring a disease by comparing the subject's molecular profile to a different subject who has already been determined to not be at risk of acquiring the disease. In other embodiments, a subject is identified as having a disease.
For example, the molecular profiling of labelled MBVs and/or EVs on a Raman spectrometer of a sample may be used to determine a subject who has a disease by comparing the subject's molecular profile to a different subject who has already been determined to have the disease.
The invention provides kits that include a device (e.g., a microcopy slide, a chip, an Au-array device, or a bead) and an agent (e.g., a long chain lipophilic polymer and a short chain hydrophilic molecule). In some embodiments, the device contains a gold-coated glass microscope slide, an array template, and a rubber array interface.
In some embodiments, the kit comprises a sterile container which contains AuNRs, a Raman reporter, a nanotag stabilizer, and one or more target-specific functionalized antibodies. Such containers can be boxes, ampoules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art. Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding medicaments.
The practice of the present invention employs, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are well within the purview of the skilled artisan. Such techniques are explained fully in the literature, such as, “Molecular Cloning: A Laboratory Manual”, second edition (Sambrook, 1989); “Oligonucleotide Synthesis” (Gait, 1984); “Animal Cell Culture” (Freshney, 1987); “Methods in Enzymology” “Handbook of Experimental Immunology” (Weir, 1996); “Gene Transfer Vectors for Mammalian Cells” (Miller and Calos, 1987); “Current Protocols in Molecular Biology” (Ausubel, 1987); “PCR: The Polymerase Chain Reaction”, (Mullis, 1994); “Current Protocols in Immunology” (Coligan, 1991). These techniques are applicable to the production of the polynucleotides and polypeptides of the invention, and, as such, may be considered in making and practicing the invention. Particularly useful techniques for particular embodiments will be discussed in the sections that follow.
The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the assay, screening, and therapeutic methods of the invention, and are not intended to limit the scope of what the inventors regard as their invention.
Schematic illustrations of the methodology of the direct Raman Extracellular Assay (dREVA) is shown in
The Au slide is fabricated by depositing 10-nm thick Au film onto a standard glass microscope slide with a magnetron sputtering technique (
To immobilize EVs, the surface of the Au slide was grafted sequentially with long chain commercially available (e.g. Nanocs) DSPE-PEG-SH MW5000 and commercially available (e.g. Sigma Aldrich) short chain MU-TEG (
To label EVs, the experiments of this example developed and used unique antibody-conjugated SERS AuNRs using QSY21 as the Raman reporter (
The labeled EVs were detected with a TSI ProRaman-L high performance spectrometer with a 785 nm laser. The Raman probe was 200 μm in diameter which covers many EVs in the well of the device. The laser beam was focused in the center of each well to collect signals of each sample. Typical signal collection parameters include integration time of 1 s and laser power of 50 mW. Baseline correction should be enabled in the signal collection software EZRaman Reader V8.1.8 MV. The signal intensity of the strongest peak at 1497 cm−1 of the SERS spectrum, 11497, is used for analysis.
To account for the variations from instrumentation response and batch-to-batch nanotags, the spectrum of each nanotag solution (0.1 nM) needs to be collected before use and the 11497 value needs to be normalized to 2000 a.u., the typical value of a 0.1 nM nanotag solution. This gives a correction factor for each nanotag to correct 11497 of each sample labeled with that nanotag. The corrected values represent the level of targeted protein on EVs.
The specificity of dREVA to immobilize membrane-bound vesicles was examined by comprising EXO immobilization between surface modification of DSPE-PEG-SH/MU-TEG and MU-PEG only. EXOs were derived from MM231 cells.
Typically, AuNRs were synthesized in two steps: formation of small Au seed and growth of Au seed in an Au growth solution for 2 h to obtain AuNRs (X. Huang et al. 2016, 128(6), 2115). The AuNRs of this example were synthesized using the traditional seed-mediated growth method, but the growth time was controlled to 10 min. At this early stage of growth time, the size of AuNRs were small. The small size of AuNRs was neccessage to efficiently label the small size of EXOs.
Using the AuNRs, a QSY21 reporter, CD63 antibodies and a MHDA-PEG stabilizer, the target-specific SERS AuNR tags were synthesized based on the procedure described in Example 1 (
In the experiments of this example, the specificity and sensitivity of dREVA for EV detection was examined using MM231 EXO as the model EV and CD63 as the EXO marker.
The ability of dREVA for EV protein profiling was tested and validated with traditional ELISA using MM231 EXO model. In the experiments of this example, six surface proteins were analyzed including one epithelial marker (EpCAM), two breast cancer markers (CD44 and HER2) and three EXO markers (CD81, CD63, and CD9).
The dREVA was validated using the gold standard ELISA. ELISA was carried using the indirect approach, in which exosomes were adsorbed onto 96 well plates and then labeled with antibodies targeting each protein. The antibodies were recognized with HRP-conjugated secondary IgG antibody and then detected with the chromogenic substrate TMB.
Compared to the traditional ELISA, the dREVA is much faster. The assay takes 2 about 3 h compared to >24 h for ELISA. It is also simpler by combining labeling and signal amplification into a single agent (i.e. the antibody-conjugated SERS nanotag). It is more sensitive, >10 times sensitive than ELISA. In addition, the dREVA provides point-of-care capability because of the portable nature of the Au chip and Raman spectrometer.
To investigate whether EXOs reflect their originating cells on biomarker expression, the expression of EpCAM, CD44, and HER2 was analyzed on the surface of MM231 cells via flow cytometry analysis. Phycoerythrin (PE)-conjugated antibodies and IgG were used for the fluorescent labeling and signal readout. The results show that the MM231 cells have very low expression of EpCAM and HER2, but high expression of CD44 (
Schematic illustrations of the methodology of the capture Raman Extracellular Assay (cREVA) are shown in
The EV device is described in Example 1. The antibody functionalization is performed by incubating 50 μg/mL targeting-specific HS-PEG-Ab for 5 h at RT followed by incubation with 0.1 mM MU-TEG for 30 min at RT (
To label EVs, SERS AuNR tags are prepared by mixing 2 nM of AuNRs solution with 10 μM QSY21 for 15 min at RT (
EV detection, signal collection, and data analyses follow the description in Example 1.
The cREVA specifically capture EVs based on the targeting proteins.
The specificity and sensitivity of cREVA for EV detection was examined using MM231 EXO as the model EV and CD63 as the EXO marker.
Compared to dREVA, the cREVA is less sensitive, probably due to the limited amount of antibodies on the surface of Au slide. It takes 4 to 5 h longer than dREVA because of the elongated time on antibody binding on the Au slide.
The ability of cREVA for EV protein profiling is tested and validated with traditional ELISA using MM231 EXO model. We analyzed six including one epithelial marker (EpCAM), two breast cancer markers (CD44 and HER2) and three EXO markers (CD81, CD63, and CD9).
The cREVA was validated using the gold standard ELISA. A quantitative comparison shows that our Raman assay has high correlation to ELISA, with correlation coefficient R2 of 0.96 (
The cREVA has been tested to detect cancer markers on different cell lines. In these studies, we profiled EpCAM, CD44, HER2, CD81, CD63, and CD on breast cancer MM231 and SKBR3 and normal breast cells MCF12A. Flow cytometry analysis showed that SKBR3 cells have high expression EpCAM and HER2 and low expression of CD44 (
Flow cytometry analysis showed that the normal MCF12A cells positive for EpCAM and low expression of CD44 and HER2 (
Using cREVA to analyze the surface markers on multiple cell lines, the experiments of this example have demonstrated that EXOs reflect their originating cells on surface protein marker expressions. The cancer-specific marker (CD44 for MM231 cells and HER2 for SKBR3 cells) are presented on cancer-derived EXOs, but not on normal cell-derived EXOs (
The cREVA has been tested for breast cancer diagnostics. Due to the heterogeneous breast cancer types, we chose HER2-positive patients (n=10) for a proof-of-concept study. The disease includes invasive lobular carcinoma, infiltrating ductal carcinoma, and adenocarcinoma of the breast in stages I, II, and III. We obtained patient plasma samples from the XpressBank at Asterand Bioscience. To collect plasma samples from healthy donors (n=5), we obtained fresh whole blood and extracted exosomes by differential centrifugation. By profiling different proteins, we found EpCAM and HER2 are biomarkers to distinguish breast cancer patients from normal controls. As shown in
In conclusion, this aspect of the present disclosure provides a simple, rapid, inexpensive, highly sensitive, and highly specific Raman-based assay for point-of-care detection and molecular profiling of EVs and other membrane bound vesicles. The assay can be performed in two ways, direct Raman extracellular assay (dREVA) and capture Raman extracellular assay (cREVA). Using the assays (both dREVA and cREVA) and model EXOs from breast cancer cells, the experiments of the preceding examples showed that EXOs express cancer markers in a similar pattern to their donor cancer cells, suggesting the potential use of screening EXOs for biomarkers for cancer detection and investigation. The assay can be widely used for basic and clinical cancer research.
The dREVA can be technically modified for automatic and high throughput clinical test of large scale of samples in real-time by using an EV microarray platform. The EXOs can be directly deposited onto the lipophilic Au slide with pico- to lower nanoliter EVs using the well-established high speed and high throughput microdrop printing technology. The microdrop printing can make over 800 EV spots on the micrometer size scale on one Au slide. This next generation REVA has the potential to revolutionize EV research and realize a novel cancer liquid biopsy approach for cancer research and diagnosis.
The results described herein above, were obtained using the following methods and materials.
All chemicals were purchased from Sigma-Aldrich unless specified. Antibodies were purchased from Biolegend (San Diego, Calif.). QSY21 carboxylic acid-succinimidyl ester was purchased from Thermo Fisher Scientific. PE-labeled antibodies were purchased from Miltenyi Biotec (Auburn, Calif.). All cell lines were purchased from ATCC (Manassas, Va.). Cell culture media were purchased from VWR (Radnor, Pa.) and fetal bovine serum (FBS) was purchased from Fisher Scientific (Waltham, Mass.).
Au NRs were synthesized in two steps: preparation of Au seeds and growth of Au seeds into AuNRs in a growth solution. To make the Au seed solution, 0.5 mL of 1 mM chloroauric acid (HAuCl4) was added to 1.5 mL of 0.13 M cetyltrimethylammonium bromide (CTAB) solution with constant stirring. 120 μL of 10 mM ice-cold sodium borohydride (NaBH4) was quickly injected and the solution was stirred for 3 min to form the Au seed solution. The Au seed solution was kept undisturbed for 3 hours in 25° C. water bath before its use. In a different glass vial, 5 ml of 1 mM HAuCl4 was added 5 mL of 0.2 M CTAB solution followed by addition of 125 μl of 4 mM silver nitrate (AgNO3). After mixing with stirring, 12 μl of Au seed solution was quickly injected into the solution and left undisturbed for 10 min to form small AuNRs. The solution was centrifuged at 14000 rpm for 10 min and the AuNR pellet was resuspended with ultrapure water for further use.
To a 0.25 mL of 1 nM AuNR solution, 10 μL of 25 μM HS-PEG-Ab was added and gently stirred for 5 h at RT. Then 25 μL of 100 μM QSY21 carboxylic acid (hydrolyzed from QSY21 carboxylic acid-succinimidyl ester) was added and stirred for 15 min at RT. At last, 25 μL of 1 mM MHDA-PEG was added and stirred for 1 h at RT. The solution was centrifuged at 14,000 rpm for 10 min to precipitate down the antibody-conjugated SERS AuNR tags. The HS-PEG-Ab was prepared in advance by reacting 10 μL of 1 mg/mL antibodies with 10 μL of 1 mM HS-PEG-NHS MW 5000 in PBS for overnight at 4° C. After reaction, the free HS-PEG-NHS was separated by membrane filtration with a 10 KD Nanosep filter (PALL Life Sciences).
Preparation of SERS AuNR tags
100 μL of 100 μM QSY21 carboxylic acid aqueous solution was added to 1 mL of 2 nM AuNRs and the mixture was stirred for 15 min at RT. After purification by centrifugation (14000 rpm, 10 min), the SERS AuNR tags were resuspended in PBS to make 1 nM solution. The solution was aged at room temperature (RT) for 2 h before use.
A standard microscopy glass slide (75×25×1 mm) was coated with 10 nm thick Au film by magnetron sputtering technique using an ORION-AJA system from a 99.99% pure Au target. The deposition of the Au layer was performed on a 4 nm titanium layer previously deposited from a 99.99% pure titanium target on the glass slide. The slide-target distance was kept at 15 cm during the process. The film thickness was controlled by an INFICON SQM-160 quartz crystal monitor/controller equipment. The rotating substrate-holder was kept at 80 rpm. The films were grown in an atmosphere of argon at 3.0 mTorr and a gas flow of 15 sccm, with the DC power supply set to 100 W and the pressure before inserting the argon was 4.0×10−8 Torr. The whole process took 4 h.
Plastic (polylactic acid) array templates with specified well size and inter-well distance were fabricated using a MakerBot Replicator PC 3D printer. The template was attached with a rubber array via a layer of glue composed of 60% silicone and 40% mineral spirit. This rubber array was made from 1.6 mm thick rubber sheet in the same dimensions as the template via punctuation. The assembled plastic and rubber arrays were used as a template array to make antibody array on the Au-coated glass slides.
Plastic (polylactic acid) array templates with specified well size and inter-well distance were fabricated using a MakerBot Replicator PC 3D printer. The template was attached with a rubber array via a layer of glue composed of 60% silicone and 40% mineral spirit. This rubber array was made from 1.6 mm thick rubber sheet in the same dimensions as the template. The rubber was punctured with 2 mm 0 perforations to make the array. The assembled plastic/rubber array was used to make EV array on the Au-coated glass slide.
The template array was attached onto the surface of the Au-coated glass slide with ¾″ wide heavy-duty binder clips. Into each well, 20 μL of 1 mM DSPE-PEG-SH was added and incubated for 1 h at RT. Then, 5 μL of 0.5 mM MU-TEG was added and incubated for 30 min at RT. The unbound chemicals were removed by washing three times with PBS.
The template array was attached onto the surface of the Au-coated glass slide with ¾″ wide heavy-duty binder clips. 25 μL of 50 μg/mL target-specific antibody-linked HS-PEG-Ab in PBS was added into the wells and incubated for 5 h at RT. The antibody-treated wells were washed for three times with PBST (100 mL PBS+0.5 mL Tween 20 (0.5%)) to get rid of unbound proteins. Then, 15 μL of 0.1 mM MU-TEG was added into the wells and incubated for 30 min at RT to saturate the Au surface. The antibody-functionalized wells were washed three times with PBST and stored at 4° C. for further use. Isotype IgG was used as the negative control.
Human breast MDA-MB-231 (MM231) cancer cells were cultured in DMEM with high glucose with 10% fetal bovine serum (FBS) at 37° C. under 5% CO2. Human breast SKBR3 cancer cells were cultured in RPMI 1640 medium with 10% fetal bovine serum (FBS) at 37° C. under 5% CO2. Human breast normal cells MCF12A (immortalized) were cultured in DMEM/F-12 medium with 5% fetal horse serum, 1% Pen/Strep (100×), 0.5 mg/mL hydrocortisone, 10 μg/mL bovine insulin, 100 ng/mL cholera toxin, 20 ng/mL EGF.
Cells were grown in conditioned cell culture media (media+10% exosome-free FBS) for 48 h. The EXO-free FBS was obtained by separating EXOs from FBS with two times of ultracentrifugation (100,000 g, 70 min). To collect EXOs, the conditioned cell culture supernatant was collected and centrifuged at 430 g at RT for 10 min. The supernatant was collected and centrifuged at 16,500 g at 4° C. for 20 min. The supernatant was collected and centrifuged at 100,000 g at 4° C. for 70 min. After removing supernatant, the exosome pellet was resuspended in cold sterile PBS and centrifuged again at 100,000 g at 4° C. for 70 min. The exosome pellet was resuspended in cold sterile PBS, filtered with a 0.20 μm filtered with a 0.2 μm PES filter (Agilent Technologies), and stored at −80° C. before use. The concentration and size distribution of exosomes were characterized using NTA with a NanoSight LM10 microscope (Malvern Instruments, Inc).
Exosome Immobilization on the Lipophilic EV Array Device, Fluoresce Imaging and Labeling with the Target-Specific SERS AuNRs
25 μL of 6.25×107/mL EXOs were added to the lipophilic Au array wells and incubated for 30 min at RT. After washing the wells three times with PBS, EXOs were labeled with 1 mM 3,3′ Dioctadecyloxacrbocyanine perchlorate (DiO) in PBS for 15 min at RT. EXOs were then washed with PBS and examined by a fluorescent microscope (Olympus IX 71) with a Prior Lumen 200 illumination system. The excitation was 482/35 nm and emission was 536/40 nm. For labeling with SERS AuNRs, 25 μL of 1 nM target-specific antibody-conjugated SERS AuNR tags were added and incubated for 30 min at RT. The wells were washed three times with PBS and immersed in 20 μL PBS for detection.
Exosome Binding on the Antibody-Functionalized EV Array Device, Fluoresce Imaging, and Labeling with SERS AuNRs
25 μL of 6.25×107/mL EXOs were added to the antibody-functionalized Au array wells and incubated for 30 min at RT. After washing the wells three times with PBS, EXOs were labeled with 1 mM DiO in PBS for 15 min at RT. EXOs were then washed with PBS and examined by a fluorescent microscope (Olympus IX 71) with a Prior Lumen 200 illumination system. The excitation was 482/35 nm and emission was 536/40 nm. For labeling with SERS AuNRs, 25 μL of 1 nM SERS AuNR tags were added into each well and incubated for 30 min at RT. The wells were washed three times with PBS and immersed in 20 μL PBS for detection.
Raman signals were collected with a TSI ProRaman spectrometer (X=785 nm). The laser beam size at focus was 200 μm. Each spectrum was collected with the laser power of 50 mW and acquisition time of 1 s. A baseline correction using a multi-segment polynomial fitting was automatically performed by the signal acquisition software (EZRaman Reader v8.1.8) to subtract SERS background (broad continuum emission). The peak at 1497 cm−1, which is the strongest one among all the peaks of the QSY21 SERS spectrum, was used as the representative peak for analysis. To account for the variations from instrumentation response and batch-to-batch nanotag preparation, the spectrum of the SERS nanotag solution (0.1 nM) during each experiment was collected and the intensity of the 1497 cm−1 peak was normalized to 2000 a.u., the typical value of a 0.1 nM nanotag solution. This gave a correction factor for each nanotag to correct the signal intensity from EXOs labeled with that nanotag during each experiment. The corrected intensity of the 1497 cm−1 peak was used for analysis.
50 μl of 6.25×108/mL MM231 EXOs were added into 96-well polystyrene plate (Corning Incorporated) wells and incubated at 4° C. for overnight. The wells were washed three times with Dulbecco's phosphate-buffered saline (DPBS) followed by incubation with 100 μl of blocking solution (DPBS with 4% BSA) at RT for 2.0 h. After washing three times with DPBS, each well was treated with the following solutions subsequently, 50 μL of 2 μg/ml target-specific antibodies (2 h, RT), 50 μl of HRP-conjugated anti-mouse IgG antibody (ThermoFisher, 1:60 dilution in blocking solution) (1 h, RT), and 100 μl of 3,3,5,5-tetramethylbenzidine solution (TMB, Sigma-Aldrich) (30 min, RT). The wells were washed three times with DBPS between steps. After the TMB incubation, 100 μl of 2 M sulfuric acid (H2SO4) was added to stop the reaction. The optical density of each well was measured at 450 nm using a BioTEK ELx800 absorbance microplate reader. Isotype IgG was used as the control.
One aspect of the present disclosure describes methodologies for protein profiling of membrane-bound single vesicles focusing on exosomes (EXOs) using SERS imaging with SERS nanotags as contrast agent (SERS-Single Vesicle Technology or SERS-SVT).
EXOs are captured and analyzed on Au-coated standard microscope glass slide (75×25×1 mm) (
Direct Capture of EXOs from Biofluids
Tetraspanin CD81 is an EXO marker that differentiates EXOs from other types of extracellular vesicles; therefore, CD81 antibody was used to capture EXOs from biofluid. The method can directly capture EXOs with CD81 monoclonal antibodies from plasma and other biofluids without EXO pre-purification. The CD81 antibody was conjugated to a polyethylene glycol thiol (PEG-SH) linker (MW=5000) by reacting HS-PEG-NHS with CD81 antibody (100:1 molar ratio) at 4° C. for overnight and then purified by filtration centrifugation. EXOs were diluted in conditioned cell culture medium (cell culture medium without fetal bovine serum) with phosphate buffer solution (PBS) and filter with 0.2 micron membrane filter.
The procedure used to capture EXOs from plasma included the following steps (
Preparation of SERS AuNR tagged secondary antibodies was performed as described supra.
For specific protein detection on exosomes, an indirect assay was used (
EXOs can be detected using a commercial Raman microscope with dark field modality. Alternatively, a versatile optical microscopic system for single EXO SERS analysis was developed by integrating an optical microscope (Nikon, LV 150N) with an excitation laser and confocal micro-Raman setup.
For Raman measurements, the samples were excited by a Melles Griot continuous-wave He laser (Model 05-LPH-925) with a wavelength of 632.8 nm (maximum power: 35 mW) through an objective lens. The laser beam was defocused by a separate lens so a large area of the sample (170 um in diameter) can be homogenously illuminated. Reflected Raman signal, after passing through the beam splitters, was filtered by a long-pass filter (to block the laser excitation) and refocused onto an intermediate image plane. The Raman signals were detected by a Photometrics CoolSnap camera for nano-imaging. The Raman signals can also be collected by a spectrometer (Horiba Jobin Yvon, model iHR550) and detected by another charged-coupled-device (CCD) camera (Horiba Jobin Yvon, model Synapse) for spectroscopic analysis. The spectrometer and CCD camera were optimized for the visible frequency with up to 95% quantum efficiency capable of single exosome measurement. The system was fully automated by a set of Labview computer programs which synchronize all optical measurements. Thus, the same area of exosome samples on the chamber slide can be simultaneously detected with dark field light scattering imaging, Raman imaging, and Raman spectroscopy.
Data collection.
Data analysis.
The targeted protein on an EXO was define as positive based on the cut off value from the IgG control. Three parameters were used define to measure the expression of a targeted protein p: fraction of the positive exosomes Fp, mean value of the protein level per EXO from the positive EXOs ζp, and mean value of the protein level per EXO from the total EXOs ζt. As reports an average value from all investigated EXOs, it is comparable to bulk measurement. In the example shown in
To account for the variations from batch-to-batch nanotags, the SERS spectrum of the nanotag solution (0.1 nM) before use was measured and the 1497 cm-1 peak was normalized to 2000 a.u., the typical value of a 0.1 nM nanotag solution. This gives a correction factor for each batch of nanotags. In the above data, the 1497 cm-1 for the 0.1 nM nanotag was 2010 a.u., therefore, correction was not needed in this study.
Profiling of HER 2 Expression on EXOs Derived from SKBR3 Cells
The use of SERS-SVT in single EXO profiling was further demonstrate by analyzing EXOs from a different origin, SKBR3 cells. SKBR3 cells are known to have high expression of HER2 cancer protein markers and low expression of CD44 and thus they represent another good model for technology validation.
The examples demonstrate that the SERS-SVT method can be used to quantitatively measure protein expressions on EXOs at single exosome level. Measurement of protein level on single exosomes can be used to diagnose cancer potentially at early stages and monitor cancer. Proteins may also be measured on other type of membrane vesicles and used for other type of diseases such as Alzheimer.
From the foregoing description, it will be apparent that variations and modifications may be made to the invention described herein to adopt it to various usages and conditions. Such embodiments are also within the scope of the following claims.
The recitation of a listing of elements in any definition of a variable herein includes definitions of that variable as any single element or combination (or subcombination) of listed elements. The recitation of an embodiment herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.
All patents and publications mentioned in this specification are herein incorporated by reference to the same extent as if each independent patent and publication was specifically and individually indicated to be incorporated by reference.
This application claims the benefit of and priority to the following U.S. Provisional Application Nos.: 62/607,133, filed Dec. 18, 2017, the entire contents of which are incorporated herein by reference.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US18/66251 | 12/18/2018 | WO | 00 |
Number | Date | Country | |
---|---|---|---|
62607133 | Dec 2017 | US |