WO2018129055A1 - Ligand binding assays using maldi-tof mass spectrometry - Google Patents

Ligand binding assays using maldi-tof mass spectrometry Download PDF

Info

Publication number
WO2018129055A1
WO2018129055A1 PCT/US2018/012199 US2018012199W WO2018129055A1 WO 2018129055 A1 WO2018129055 A1 WO 2018129055A1 US 2018012199 W US2018012199 W US 2018012199W WO 2018129055 A1 WO2018129055 A1 WO 2018129055A1
Authority
WO
WIPO (PCT)
Prior art keywords
beads
ligand binding
mass
sample plate
molecules
Prior art date
Application number
PCT/US2018/012199
Other languages
French (fr)
Inventor
Marvin L. Vestal
Original Assignee
Virgin Instruments Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Virgin Instruments Corporation filed Critical Virgin Instruments Corporation
Priority to EP18736271.0A priority Critical patent/EP3566052A4/en
Priority to CN201880005874.2A priority patent/CN110168369A/en
Publication of WO2018129055A1 publication Critical patent/WO2018129055A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5302Apparatus specially adapted for immunological test procedures
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6848Methods of protein analysis involving mass spectrometry
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6848Methods of protein analysis involving mass spectrometry
    • G01N33/6851Methods of protein analysis involving laser desorption ionisation mass spectrometry
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/84Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving inorganic compounds or pH
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2560/00Chemical aspects of mass spectrometric analysis of biological material

Definitions

  • ELISA Immunosorbent Assays
  • Ligand binding assays have been used to measure of the interactions that occur between two molecules, such as protein-bindings, as well as the degree of affinity for which the reactants bind together. More specifically, ligand binding assays are used to test for the presence of target molecules in a sample that is known to bind to the receptor. Various detection methods have been used to determine the presence and extent of the ligand-receptor complexes formed. Known methods include electrochemical detection through various fluorescence detection methods.
  • FIG. 1 illustrates a block diagram of an embodiment of a ligand binding assay apparatus according to the present teaching that produces and analyzes ligand binding assays.
  • FIG. 2 illustrates a flow diagram of an embodiment of a method of generating ligand binding assays by MALDI-TOF mass spectrometry according to the present teaching.
  • FIG. 3 A illustrates a layout of a portion of a microwell sample plate of the ligand binding assay apparatus according to the present teaching.
  • FIG. 3B illustrates a detailed view of one particular hexagon in the hexagonal array shown in FIG. 3 A.
  • FIGS. 4A illustrates a layout of an embodiment of a sample plate with division into regions according to the present teaching.
  • FIGS. 4B illustrates a layout of another embodiment of a sample plate with division into regions according to the present teaching.
  • FIG. 5 illustrates a commercially available biotin linker employed to covalently attach biotin to molecules of interest for use in the ligand binding assay method and apparatus of the present teaching.
  • One aspect of the present teaching relates to providing a highly multiplexed Mass
  • MSIA Spectrometric ImmunoAssay
  • ELISA Enzyme- Linked Immunosorbent Assays
  • the apparatus and method of the present teaching can be used to perform multiplexed detection and characterization of single cells.
  • One application of the method and apparatus of the present teaching relates to screening of a human protein library to discover new autoimmune and tumor antigens in patient sera for clinical diagnostics and immunotherapy. Another application of the method and apparatus of the present teaching relates to screening of a synthetic combinatorial library against specific protein targets to discover new drugs. Another application of the method and apparatus of the present teaching relates to developing drug compounds with high efficacy and minimal side-effects to assist in new drug discoveries.
  • some embodiments of the apparatus and method for ligand binding assays use beads prepared with a tag of predetermined mass and a bait molecule attached.
  • the beads comprise Streptavidin Sepharose® High Performance beads, which are commercially available from GE Healthcare Life Sciences.
  • One type of suitable bead is GE Healthcare Life Sciences Product Code: 17- 5113-01.
  • These Streptavidin Sepharose® beads are nominally 34 ⁇ in diameter.
  • the mass tag molecules and bait molecules are biotinylated and are bound to the beads by the streptavidin- biotin interaction.
  • FIG. 1 illustrates a block diagram 100 of an embodiment of a ligand binding assay apparatus according to the present teaching that produces and analyzes ligand binding assays.
  • the ligand binding assay apparatus 100 accepts beads 102 into an incubator 104 with a tag of predetermined mass and a bait molecule attached.
  • the beads 102 comprise Streptavidin Sepharose High Performance beads.
  • the mass tag molecules and bait molecules are biotinylated and are bound to the beads 102 by the streptavidin-biotin interaction.
  • the beads 102 are loaded into an incubator 104.
  • the incubator 104 incubates the bait molecule with samples of interest so that the bait molecules strongly bind to target molecules contained in the sample.
  • the ligand binding assay apparatus 100 further comprises a washer 106 that washes the incubated beads 102.
  • the washer 106 dispenses washing fluid on the beads and agitates the incubated beads 102 in the washing fluid.
  • the properties of the washing solution are chosen so that weakly bound molecules are cleaved from the beads 102 and strongly bound molecules remain attached to the beads 102.
  • the washed beads 102 are then transferred to a sample handler 108 that includes a sample plate loader 110 and matrix material applicator 112, such as a sprayer.
  • matrix material applicator 112 such as a sprayer.
  • matrix material applicators can be used to spray and/or directly apply matrix materials.
  • the incubated and washed beads are positioned on a sample plate suitable for MALDI-TOF analysis.
  • the surface of the sample plate comprises a plurality of microwells where the dimensions of the microwells are chosen so that only one bead can be contained within any given microwell. That is, the number of beads applied to the microwell sample plate is not greater than the number of microwells in each region of the microwell sample plate.
  • Microwell sample plates are described in more detail in connection with FIGS. 3A, 3B, 4A and 4B. In one embodiment, both the diameter and the depth of the microwells of the microtiter sample plate are on order of about 40 ⁇ .
  • the sample plate loader 110 applies a suspension of incubated and washed beads
  • the number of beads is not greater than the number of microwells in that region of the microwell sample plate.
  • the sample plate is divided into regions with a cover having apertures that separate the sample plate into predetermined portions with a gasket that prevents liquid from flowing between separate regions.
  • the sample plate loader 110 then agitates the sample plate with a vibrator causing beads to spread out in a monolayer on the sample plate. Also, in some embodiments comprising microwell plates, the sample plate loader 110 then agitates the sample plate with a vibrator causing each bead to settle into a microwell and eventually dry.
  • the sample handler 108 also includes a matrix material applicator 112 for depositing MALDI matrix on the surface of the sample plate.
  • the matrix material applicator 112 can included a sprayer or equivalent type of apparatus that produces small droplets of fluid with MALDI matrix material.
  • One suitable sprayer for spraying these very small droplets is the HTX TM-Sprayer, which is commercially available from HTX
  • the matrix solution sprayed on the microtiter sample plate is allowed to dry.
  • the drying process causes rupture of the streptavidin-biotin bonds and rupture of the non-covalent bonds between bait molecules and the strongly bound target molecules.
  • the rupture results in the released molecules being incorporated into matrix crystals.
  • the sample plate handler 108 also includes a transfer mechanism that transfers the incubated and washed beads 102 in the sample plate to the sample mass analyzer 114.
  • the sample mass analyzer 114 comprises a mass spectrometer 116 such as a Time-of-Flight (TOF) mass spectrometer like the matrix-assisted laser desorption/ionization (MALDI) TOF mass spectrometers manufactured by Virgin Instruments Corporation, the assignee of the present application.
  • TOF Time-of-Flight
  • MALDI matrix-assisted laser desorption/ionization
  • a laser ionization source generates a laser beam that scans over the surface of the sample plate producing and recording a mass spectrum from each location on the multi-well sample plate.
  • a translation stage can be used to move the sample plate in two dimensions to scan the laser in the MALDI-TOF mass spectrometer 116 to ionize samples over the entire two-dimensional surface of the sample plate so that the MALDI-TOF mass spectrometer 116 can produce and record a mass spectrum from each location on the plate.
  • a processor 118 such as a computer, interfacing with the MALDI TOF mass spectrometer 116 executes an algorithm using the spectra data generated by the MALDI TOF mass spectrometer 116 to detect the predetermined mass to produce a mass spectrum of the molecules that were originally bound to the beads by the streptavidin-biotin bonds.
  • the mass spectra in which the predetermined mass of the mass tag is detected are summed together to produce a mass spectrum of the molecules that were originally bound to the beads by the streptavidin-biotin bonds and the target molecules that were bound to the bait molecules.
  • the processor then uses the mass spectrum to produce a ligand binding assay.
  • FIG. 2 illustrates a flow diagram of a method 200 of generating ligand binding assays by MALDI-TOF mass spectrometry 100 according to the present teaching.
  • the beads 102 are supplied.
  • the beads include a tag of predetermined mass and a bait molecule attached.
  • the beads 102 comprise Streptavidin Sepharose High Performance beads which are commercially available from GE Healthcare Life Sciences.
  • the mass tag molecules and bait molecules are biotinylated and are bound to the beads by the streptavidin-biotin interaction.
  • ligand- binding assays are produced from biological samples, where small aliquots of affinity beads are added to a biological sample.
  • the biological sample can be human serum, diluted as appropriate.
  • Step two 204 of the method 200 includes incubating beads 102 with samples of interest.
  • the bait molecule strongly binds to a molecule contained in the sample.
  • incubating beads 102 with samples of interest comprises mixing a sample and the beads 102 in predetermined relative liquid volumes in a well in a microtiter plate. The mixture of a sample and beads 102 is agitated to bring beads 102 in contact with sample molecules. Then, after a predetermined time, the beads 102 are allowed to settle in the bottom of the well and supernatant liquid is withdrawn. In one specific embodiment, the beads 102 are incubated for 30 min under agitation to allow for analyte binding.
  • Step three 206 of the method 200 includes washing the incubated beads 102.
  • the beads 102 are collected by centrifugation in a microcentrifuge tube and washed multiple times in 20 mM Tris pH 7.3, 5 mM EDTA, 1 M NaCl and then washed multiple times in the same Tris pH 7.3 buffer in the absence of salt. After the final wash, the remaining beads are re-suspended in a minimum volume of buffer, also referred to as a carrier solvent.
  • step three 206 includes adding a washing solution to the wells in the microtiter plate and then agitating the microtiter plate so that the beads interact with the washing solution.
  • the washing solution is chosen such that the washing step causes weakly bound molecules to cleave from the beads while strongly bound molecules remain attached to the beads.
  • Step four 208 of the method 200 includes loading the incubated and washed beads
  • the sample plate comprises a microwell sample plate with a plurality of wells where each well has dimensions chosen so that only one bead is contained within the well. In one specific embodiment of the microwell sample plate, both the diameter of the wells and their depth is on order of about 40 ⁇ .
  • step four 208 includes transferring incubated beads to the surface of the sample plate so that each bead 102 is contained in a well in a predetermined region of the sample plate.
  • the step of loading the incubated and washed beads 102 comprises spreading the re-suspended remaining beads 102 in the carrier solvent over a well-defined area of the sample plate and drying the beads 102.
  • the sample plate is divided into regions by a cover with apertures that separate the plate into predetermined portions with a gasket that prevents liquid from flowing between separate regions.
  • a suspension of beads 102 in appropriate liquid is applied to each region with the number of beads 102 applied being limited to the number of microwells in that region of the sample plate.
  • the plate is then agitated causing each bead 102 to settle into a microwell of the sample plate.
  • the plate is then allowed to dry.
  • Step five 210 of the method 200 includes depositing a MALDI matrix on the surface of the sample plate.
  • the cover and gasket are removed from the sample plate and a solution of MALDI matrix is applied onto the surface of the sample plate with a spraying apparatus.
  • the spraying apparatus produces very small droplets.
  • the matrix solution on the sample plate is then allowed to dry. The drying process causes rupture of the streptavidin-biotin bonds and rupture of the non-covalent bonds between bait molecules and strongly bound target molecules. The rupture causes a release of molecules that are then incorporated into the matrix crystals.
  • the loaded sample plate with the MALDI matrix material applied is installed in a MALDI-TOF mass spectrometer.
  • a solution of MALDI matrix is deposited directly on the surface of each region of the sample plate and allowed to dry.
  • the drying process causes rupture of the streptavidin-biotin bonds and rupture of the non-covalent bonds between bait molecules and strongly bound target molecules.
  • the rupture causes a release of molecules that are then incorporated into the matrix crystals.
  • the cover and gasket are removed from the sample plate, and then the loaded sample plate with the MALDI matrix material applied is installed in a MALDI-TOF mass spectrometer.
  • Step six 212 of the method 200 includes performing MALDI-TOF mass spectroscopy to produce a mass spectra.
  • MALDI-TOF mass spectroscopy is performed by scanning a laser beam over the surface of the sample plate to produce mass spectra.
  • the sample plate is moved to cause the laser beam to scan over the surface of the sample plate producing a mass spectrum from each location or pixel on the sample plate.
  • a mass spectrum is recorded from each microwell location in the sample plate.
  • the detection limit for MALDI-TOF spectroscopy is not limited by chemical noise. In these embodiments, the detection limit for MALDI-TOF spectroscopy is about 1 femtomole/mm 2 .
  • the detection limit for MALDI-TOF spectroscopy corresponds to 1.3 attomoles/bead or equivalently 780,000 molecules/bead which is 0.04 pg for PSA. In some measurements, one bead may be sufficient to realize a desired detection limit for MALDI-TOF spectroscopy. In other measurements, multiple beads are used to produce lower detection limits, greater dynamic range, and better coefficient of variation CV, which is the ratio of the standard deviation of the peak intensity to the mean.
  • Step seven 214 of the method 200 includes analyzing the mass spectra to produce immunoassays.
  • the analyzing the mass spectra comprises executing a computer algorithm that sums the mass spectra obtained for the predetermined mass of a mass tag together to produce a mass spectrum of the molecules that were originally bound to the beads by the streptavidin-biotin bonds and the target molecules that were bound to the bait molecules.
  • a computer algorithm sums all of the mass spectra in which a predetermined mass tag is detected to produce a mass spectrum of the bound target molecules. Each given mass tag detected corresponds to a unique bait molecule.
  • the mass spectra in which the mass tag is detected also include the mass of captured molecules that are strongly bound to the bait molecule.
  • FIG. 3 A illustrates a layout of a portion of a microwell sample plate of the ligand binding assay apparatus according to the present teaching.
  • the microwells 302 are 40 ⁇ in diameter and are arranged in a regular hexagonal array 304 with individual hexagons 306 having a 50 ⁇ height.
  • FIG. 3A shows the size of a 25- ⁇ pixel 308 and a 10- ⁇ pixel 310 in relation to the size of an individual hexagon 306 and the regular hexagon array 304.
  • FIG. 3B shows a detailed view of one particular hexagon in the hexagonal array
  • the hexagon layout has two hexagonal dimensions, a 312 and b 314.
  • the laser beam in MALDI-TOF mass spectrometer is rastered over the surface of a microwell plate comprising the layout 300 illustrated in FIG. 3A.
  • the rastering is performed at intervals of 25 ⁇ with a 10- ⁇ diameter laser beam using laser repetition rate of 5 kHz, a scanning speed of 2.5 mm/s, and summing of 50 laser shots per pixel to produce 25 ⁇ long pixels.
  • the total pixels/cell ratio is about 3.5 with about half on the well and half with significant contribution from adjacent wells.
  • Total number of laser shots on each well is 100, with the laser irradiation time per cell equal to about 0.035s.
  • the rastering is performed over the surface of microwell plate comprising the layout 300 illustrated in FIG. 3A at intervals of 10 ⁇ with a 10- ⁇ diameter laser beam using laser repetition rate of 5 kHz, scanning speed of 1 mm/s, and summing of 50 laser shots per pixel to produce 10 ⁇ long pixels.
  • the total pixels/cell ratio is about 25 with about 58% on the well having no significant contribution from adjacent wells.
  • Total number of laser shots on the well is 600, and the laser irradiation time per cell is 0.25 seconds.
  • the rastering is performed over the surface of microwell plate comprising the layout 300 illustrated in FIG. 3A at intervals of 12.5 ⁇ with a 10- ⁇ diameter laser beam using laser repetition rate of 5 kHz, scanning speed of 1.25 mm/s, and summing of 50 laser shots per pixel to produce 12.5 ⁇ long pixels.
  • the total pixels/cell ratio is about 16 with about 58% on the well and with no significant contribution from adjacent wells.
  • Total number of laser shots on each well is 400 with the laser irradiation time equal to 0.16 seconds.
  • FIGS. 4A illustrates a layout 400 of an embodiment of a microwell sample plate
  • the microwell plate 402 has nominally a 25x75 mm outside dimensions and comprises approximately 866,250 microwells.
  • the microwell plate 402 is separated into regions or spots 404 by masks that are clamped to the plate with a gasket (not shown) that prevents liquid flow between the spots 404.
  • the plate 400 shown in FIG. 4A illustrates a layout 400 comprising twenty-four spots 404 arranged in a 3x8 array. Each spot 404 is 7x7 mm and there is a 1-mm-long space between spots 404. Each spot 404 comprises approximately 22,600 microwells. The twenty-four spots include a total of about 542,400 wells.
  • FIG. 4B illustrates a layout 450 of another embodiment of a microwell sample plate with division into regions according to the present teaching.
  • FIG. 4B illustrates a layout 450 that comprises ninety-six spots 406 arranged in an 8x20 array. Each spot 406 is 3x3 mm and there is a 1-mm-long space between spots 406. Each spot 406 comprises approximately 4100 microwells.
  • the plate comprises three-hundred eighty-four spots arranged in a 12x32 array with each spot being dimensioned 1.5x1.5 mm and with a 0.5-mm-long space between spots. Each spot comprises approximately 1030 microwells.
  • the plate comprises one-thousand five hundred and thirty-six spots arranged in a 24x64 array with each spot being dimensioned 0.5x0.5 mm and with 0.5 mm space between spots. In this layout, each spot comprises approximately 115 microwells.
  • any number of spots can be arranged as needed for a particular application.
  • Biotin is a relatively small molecule, which is easily bonded to peptides or aptamers with negligible impact on biological activity. Biotin / Streptavidin association occurs rapidly, and is relatively stable with
  • FIG. 5 illustrates a commercially available biotin linker 500 employed to covalently attach biotin to molecules of interest in an embodiment of the ligand binding assay apparatus and method of the present teaching.
  • the mass of biotinylated molecule is increased by 473.22 Da by this addition of the biotin.
  • the masses of biotinylated peptides used for mass tags must be sufficiently different to be distinguished by the mass spectrometer. Taking into account the isotopic envelopes, a difference of 2000 ppm is adequate.
  • a list of peptides can be generated with a mass range, m n -m 0j of 800-2400 Da and a resolution of 2000 ppm to approach this theoretical limit.
  • These suitable peptides are chosen with biotinylated N-terminus and C-terminal arginine (R) and up to 12 other amino acid in order to obtain a relative uniform response factors with MALDI-TOF MS.
  • One feature of the ligand-binding assay apparatus and method of the present teaching is the ability to utilize a number of known and commercially available beads.
  • purified Streptavidin Sepharose beads which are commercially available through GE Healthcare Life Sciences as Product Code 17-5113-01 are used. These beads are composed of a highly cross-linked, chemically stable, rigid agarose core of about 34 microns in diameter. These beads are well suited for placement in a microtiter samples plate, one bead per 40 ⁇ m-diameter well. In addition, these beads are specified to bind 6mg BSA/mL of beads.
  • 1.0 mL of beads comprises approximately, 2.5xl0 7 beads/mL.
  • the number of molecules can be determined from 6 mg BSA, which is equal to 90 nmoles or equivalently 5.4xl0 16 molecules. Then 5.4xl0 16 molecules/2.5xl0 7 beads is equal to 2.16xl0 9
  • biotinylated aptamers which is commercially available from SomaLogic Inc., Boulder, CO
  • biotinylated peptides which is commercially available from 21 st Century Biochemicals, Marlborough, MA
  • beads enable simple construction of a diverse array of targeted analyses.
  • biotinylated aptamer beads for example anti-Beta-2-microglobulin, which is commercially available from SomaLogic, Boulder, CO as product number B-3485- 28 2
  • the beads and a biotinylated mass tag are mixed the in ratio of 10: 1 (aptamer: mass tag) in 20 mM Tris pH 7.3 buffer, 5 mM EDTA (ethylenediaminetetraacetic acid), 1 M NaCl buffer solution. This mixture is allowed to incubate for 30 min. The incubated beads are then collected by centrifugation and washed three times in the same Tris pH 7.3 buffer in the absence of NaCl.
  • the aptamer-to-peptide ratio is chosen to mimic the relative sensitivity for the target analyte and peptide mass tag in the mass spectrometer.
  • the approximate number of binding sites on a single bead is estimated at 2x10 9 .
  • target analytes are sequestered from biological samples by using analyte specific aptamer reagents bound to beads.
  • the beads also contain a mass-tag for bead identification and to aid in analyte signal averaging. Both the analyte specific affinity aptamers and the bead specific mass tags are non-covalently bound to the bead by a biotin / streptavidin linkage.
  • Some methods according to the present teaching use a plurality of sets of beads in the microtiter sample plate where each set of beads includes a unique mass tag and a unique bait molecule.
  • the mass spectrometer can operate in a multiplex mode of operation where mass spectra measurements can be taken on a plurality of different sets of beads and a computer algorithm is used to process the spectra to produce a ligand binding assay for each of the plurality of sets of beads.
  • Various computer algorithms can be used to process the mass spectra to perform various types of analysis. Some algorithms sum together all of the mass spectra in which a predetermined mass tag is detected to produce a mass spectrum of the bound molecules. Each given mass tag detected corresponds to a unique bait molecule in these algorithms. Thus, the mass spectra in which the mass tag is detected also include the mass of captured prey molecules. The summed mass spectrum for each mass tag is processed to yield mass and intensity for each mass peak detected. These include mass and intensity of the tag molecule ion, mass and intensity of ions produced by captured prey molecules, and may include ions produced from bait molecules.
  • the ratio of the intensity of ions produced from prey molecules to the intensity of molecular ions from the mass tag may be used to determine the concentration of prey molecules in samples of interest. In some embodiments, this may require calibration of this ratio using molecular standards by techniques commonly used in quantitative applications of mass spectrometry.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Optics & Photonics (AREA)
  • Inorganic Chemistry (AREA)
  • Other Investigation Or Analysis Of Materials By Electrical Means (AREA)

Abstract

An apparatus for ligand binding assays includes an incubator that incubates a plurality of beads with a sample of interest wherein each of the plurality of beads comprises a tag of predetermined mass and a bait molecule. A washer washes the incubated beads so that weakly bound molecules are removed while strongly bound molecules are retained. A sample plate loader loads the washed beads into the sample plate such that respective ones of the plurality of beads are loaded into respective ones of the plurality of well. A sprayer deposits matrix assisted laser desorption ionization (MALDI) matrix material on the surface of the sample plate so that each of the plurality of beads is exposed to MALDI matrix material. A MALDI-TOF mass spectrometer receives the sample plate and performs mass spectrometry on samples in the plurality of wells. A computer executes an algorithm that analyzes the mass spectra.

Description

Ligand Binding Assays Using MALDI-TOF Mass Spectrometry
[0001] The section headings used herein are for organizational purposes only and should not to be construed as limiting the subject matter described in the present application in any way.
Cross Reference to Related Application
[0002] The present application is non-provisional application of U.S. Provisional Patent
Application Serial No. 62/442,512, entitled "Ligand Binding Assays Using MALDI-TOF Mass Spectrometry" filed on January 5, 2017. The entire contents of U.S. Provisional Patent
Application Serial No. 62/442,512 are herein incorporated by reference.
Introduction
[0003] Major research efforts are focusing on characterizing the millions of interactions of the human proteome with other molecules. These include proteins, nucleic acids, lipids, and metabolites. Immunoassays are very important tools that are used to perform this work. Factors such as: (1) the rising incidences of chronic and infectious diseases; (2) the rapidly expanding biotechnology and pharmaceutical industries; (3) the extensive use of immunoassays in oncology because of its cost-effectiveness and rapid action; (4) and the growing geriatric population are expected to propel the growth of the immunoassay market in the coming years. See, for example, Genetic and Engineering &Biotechnology News, September 15, 2016, p. 12. It is, however, highly desirable to have an alternative to the widely used Enzyme-Linked
Immunosorbent Assays (ELISA) that is significantly faster, more sensitive, and less expensive.
[0004] Ligand binding assays have been used to measure of the interactions that occur between two molecules, such as protein-bindings, as well as the degree of affinity for which the reactants bind together. More specifically, ligand binding assays are used to test for the presence of target molecules in a sample that is known to bind to the receptor. Various detection methods have been used to determine the presence and extent of the ligand-receptor complexes formed. Known methods include electrochemical detection through various fluorescence detection methods.
Brief Description of the Drawings
[0003] The present teaching, in accordance with preferred and exemplary embodiments, together with further advantages thereof, is more particularly described in the following detailed description, taken in conjunction with the accompanying drawings. The skilled person in the art will understand that the drawings, described below, are for illustration purposes only. The drawings are not necessarily to scale, emphasis instead generally being placed upon illustrating principles of the teaching. The drawings are not intended to limit the scope of the Applicant's teaching in any way.
[0004] FIG. 1 illustrates a block diagram of an embodiment of a ligand binding assay apparatus according to the present teaching that produces and analyzes ligand binding assays.
[0005] FIG. 2 illustrates a flow diagram of an embodiment of a method of generating ligand binding assays by MALDI-TOF mass spectrometry according to the present teaching.
[0006] FIG. 3 A illustrates a layout of a portion of a microwell sample plate of the ligand binding assay apparatus according to the present teaching.
[0007] FIG. 3B illustrates a detailed view of one particular hexagon in the hexagonal array shown in FIG. 3 A.
[0008] FIGS. 4A illustrates a layout of an embodiment of a sample plate with division into regions according to the present teaching.
[0009] FIGS. 4B illustrates a layout of another embodiment of a sample plate with division into regions according to the present teaching.
[0010] FIG. 5 illustrates a commercially available biotin linker employed to covalently attach biotin to molecules of interest for use in the ligand binding assay method and apparatus of the present teaching.
Description of Various Embodiments
[0010] The present teaching will now be described in more detail with reference to exemplary embodiments thereof as shown in the accompanying drawings. While the present teachings are described in conjunction with various embodiments and examples, it is not intended that the present teachings be limited to such embodiments. On the contrary, the present teachings encompass various alternatives, modifications and equivalents, as will be appreciated by those of skill in the art. Those of ordinary skill in the art having access to the teaching herein will recognize additional implementations, modifications, and embodiments, as well as other fields of use, which are within the scope of the present disclosure as described herein.
[0011] Reference in the specification to "one embodiment" or "an embodiment" means that a particular feature, structure, or characteristic described in connection with the embodiment is included in at least one embodiment of the teaching. The appearances of the phrase "in one embodiment" in various places in the specification are not necessarily all referring to the same embodiment.
[0012] It should be understood that the individual steps of the methods of the present teachings can be performed in any order and/or simultaneously as long as the teaching remains operable. Furthermore, it should be understood that the apparatus and methods of the present teachings can include any number or all of the described embodiments as long as the teaching remains operable.
[0013] One aspect of the present teaching relates to providing a highly multiplexed Mass
Spectrometric ImmunoAssay (MSIA) that can be used as an alternative to widely used Enzyme- Linked Immunosorbent Assays (ELISA). In particular, the apparatus and method of the present teaching can be used to perform multiplexed detection and characterization of single cells.
Using the highly multiplexed Mass Spectrometric ImmunoAssay of the present teaching can provide much faster results with higher sensitivity and higher dynamic range that are less expensive to produce compared with the widely used Enzyme-Linked Immunosorbent Assays. Apparatus of the present teaching can be fully automated to achieve very high throughput at relatively low cost. One feature of these apparatus is that they can be label-free since fluorescent tags are not required.
[0014] One application of the method and apparatus of the present teaching relates to screening of a human protein library to discover new autoimmune and tumor antigens in patient sera for clinical diagnostics and immunotherapy. Another application of the method and apparatus of the present teaching relates to screening of a synthetic combinatorial library against specific protein targets to discover new drugs. Another application of the method and apparatus of the present teaching relates to developing drug compounds with high efficacy and minimal side-effects to assist in new drug discoveries.
[0015] More specifically, some embodiments of the apparatus and method for ligand binding assays according to the present teaching use beads prepared with a tag of predetermined mass and a bait molecule attached. In one embodiment, the beads comprise Streptavidin Sepharose® High Performance beads, which are commercially available from GE Healthcare Life Sciences. One type of suitable bead is GE Healthcare Life Sciences Product Code: 17- 5113-01. These Streptavidin Sepharose® beads are nominally 34 μπι in diameter. The mass tag molecules and bait molecules are biotinylated and are bound to the beads by the streptavidin- biotin interaction. Biotinylation is a process of covalently attaching biotin to a protein, nucleic acid or other molecule. Biotinylation is both a rapid and a specific process. Furthermore, biotinylation is performed without a significant perturbation to the natural function of the molecule due to the small size of biotin (MW = 244.3 Da).
[0016] FIG. 1 illustrates a block diagram 100 of an embodiment of a ligand binding assay apparatus according to the present teaching that produces and analyzes ligand binding assays. The ligand binding assay apparatus 100 accepts beads 102 into an incubator 104 with a tag of predetermined mass and a bait molecule attached. In one embodiment, the beads 102 comprise Streptavidin Sepharose High Performance beads. In this embodiment, the mass tag molecules and bait molecules are biotinylated and are bound to the beads 102 by the streptavidin-biotin interaction. The beads 102 are loaded into an incubator 104. The incubator 104 incubates the bait molecule with samples of interest so that the bait molecules strongly bind to target molecules contained in the sample. [0017] The ligand binding assay apparatus 100 further comprises a washer 106 that washes the incubated beads 102. The washer 106 dispenses washing fluid on the beads and agitates the incubated beads 102 in the washing fluid. In some embodiments, the properties of the washing solution are chosen so that weakly bound molecules are cleaved from the beads 102 and strongly bound molecules remain attached to the beads 102.
[0018] The washed beads 102 are then transferred to a sample handler 108 that includes a sample plate loader 110 and matrix material applicator 112, such as a sprayer. One skilled in the art will appreciate that numerous types of matrix material applicators can be used to spray and/or directly apply matrix materials. In some embodiments, the incubated and washed beads are positioned on a sample plate suitable for MALDI-TOF analysis. In one embodiment the surface of the sample plate comprises a plurality of microwells where the dimensions of the microwells are chosen so that only one bead can be contained within any given microwell. That is, the number of beads applied to the microwell sample plate is not greater than the number of microwells in each region of the microwell sample plate. Microwell sample plates are described in more detail in connection with FIGS. 3A, 3B, 4A and 4B. In one embodiment, both the diameter and the depth of the microwells of the microtiter sample plate are on order of about 40 μιη.
[0019] The sample plate loader 110 applies a suspension of incubated and washed beads
102 in appropriate liquid to each desired region of the sample plate where the number of beads
102 applied is not greater than the number of beads that can be spread in a monolayer in the desired region of the sample plate. In some embodiments comprising microwell plates, the number of beads is not greater than the number of microwells in that region of the microwell sample plate. In one embodiment, the sample plate is divided into regions with a cover having apertures that separate the sample plate into predetermined portions with a gasket that prevents liquid from flowing between separate regions. The sample plate loader 110 then agitates the sample plate with a vibrator causing beads to spread out in a monolayer on the sample plate. Also, in some embodiments comprising microwell plates, the sample plate loader 110 then agitates the sample plate with a vibrator causing each bead to settle into a microwell and eventually dry.
[0020] The sample handler 108 also includes a matrix material applicator 112 for depositing MALDI matrix on the surface of the sample plate. In some apparatus, the matrix material applicator 112 can included a sprayer or equivalent type of apparatus that produces small droplets of fluid with MALDI matrix material. One suitable sprayer for spraying these very small droplets is the HTX TM-Sprayer, which is commercially available from HTX
Technologies in Chapel Hill, NC. The matrix solution sprayed on the microtiter sample plate is allowed to dry. The drying process causes rupture of the streptavidin-biotin bonds and rupture of the non-covalent bonds between bait molecules and the strongly bound target molecules. The rupture results in the released molecules being incorporated into matrix crystals.
[0021] The sample plate handler 108 also includes a transfer mechanism that transfers the incubated and washed beads 102 in the sample plate to the sample mass analyzer 114. The sample mass analyzer 114 comprises a mass spectrometer 116 such as a Time-of-Flight (TOF) mass spectrometer like the matrix-assisted laser desorption/ionization (MALDI) TOF mass spectrometers manufactured by Virgin Instruments Corporation, the assignee of the present application. The operation of MALDI TOF mass spectrometers is well known in the art. A laser ionization source generates a laser beam that scans over the surface of the sample plate producing and recording a mass spectrum from each location on the multi-well sample plate. [0022] One feature of the present teaching is that a translation stage can be used to move the sample plate in two dimensions to scan the laser in the MALDI-TOF mass spectrometer 116 to ionize samples over the entire two-dimensional surface of the sample plate so that the MALDI-TOF mass spectrometer 116 can produce and record a mass spectrum from each location on the plate.
[0023] A processor 118, such as a computer, interfacing with the MALDI TOF mass spectrometer 116 executes an algorithm using the spectra data generated by the MALDI TOF mass spectrometer 116 to detect the predetermined mass to produce a mass spectrum of the molecules that were originally bound to the beads by the streptavidin-biotin bonds. In one method according to the present teaching, the mass spectra in which the predetermined mass of the mass tag is detected are summed together to produce a mass spectrum of the molecules that were originally bound to the beads by the streptavidin-biotin bonds and the target molecules that were bound to the bait molecules. The processor then uses the mass spectrum to produce a ligand binding assay.
[0024] FIG. 2 illustrates a flow diagram of a method 200 of generating ligand binding assays by MALDI-TOF mass spectrometry 100 according to the present teaching. Referring to both the block diagram of the embodiment of a ligand binding assay apparatus 100 described in connection with FIG. 1 and the flow diagram of the method 200, in step one 202, the beads 102 are supplied. In some embodiments, the beads include a tag of predetermined mass and a bait molecule attached. In one specific embodiment, the beads 102 comprise Streptavidin Sepharose High Performance beads which are commercially available from GE Healthcare Life Sciences. In some embodiments, the mass tag molecules and bait molecules are biotinylated and are bound to the beads by the streptavidin-biotin interaction. In another specific embodiment, ligand- binding assays are produced from biological samples, where small aliquots of affinity beads are added to a biological sample. For example, the biological sample can be human serum, diluted as appropriate.
[0025] Step two 204 of the method 200 includes incubating beads 102 with samples of interest. In the incubation step two 204, the bait molecule strongly binds to a molecule contained in the sample. In one embodiment, incubating beads 102 with samples of interest comprises mixing a sample and the beads 102 in predetermined relative liquid volumes in a well in a microtiter plate. The mixture of a sample and beads 102 is agitated to bring beads 102 in contact with sample molecules. Then, after a predetermined time, the beads 102 are allowed to settle in the bottom of the well and supernatant liquid is withdrawn. In one specific embodiment, the beads 102 are incubated for 30 min under agitation to allow for analyte binding.
[0026] Step three 206 of the method 200 includes washing the incubated beads 102. In one specific embodiment, the beads 102 are collected by centrifugation in a microcentrifuge tube and washed multiple times in 20 mM Tris pH 7.3, 5 mM EDTA, 1 M NaCl and then washed multiple times in the same Tris pH 7.3 buffer in the absence of salt. After the final wash, the remaining beads are re-suspended in a minimum volume of buffer, also referred to as a carrier solvent.
[0027] During the washing procedure, weakly bound molecules are removed while strongly bound molecules are retained. In one method according to the present teaching, step three 206 includes adding a washing solution to the wells in the microtiter plate and then agitating the microtiter plate so that the beads interact with the washing solution. In some methods according to the present teaching, the washing solution is chosen such that the washing step causes weakly bound molecules to cleave from the beads while strongly bound molecules remain attached to the beads.
[0028] Step four 208 of the method 200 includes loading the incubated and washed beads
102 in a sample plate. In some methods, the sample plate comprises a microwell sample plate with a plurality of wells where each well has dimensions chosen so that only one bead is contained within the well. In one specific embodiment of the microwell sample plate, both the diameter of the wells and their depth is on order of about 40 μπι. In some methods, step four 208 includes transferring incubated beads to the surface of the sample plate so that each bead 102 is contained in a well in a predetermined region of the sample plate. In some methods according to the present teaching, the step of loading the incubated and washed beads 102 comprises spreading the re-suspended remaining beads 102 in the carrier solvent over a well-defined area of the sample plate and drying the beads 102.
[0029] In one specific embodiment of the sample plate, the sample plate is divided into regions by a cover with apertures that separate the plate into predetermined portions with a gasket that prevents liquid from flowing between separate regions. With these sample plates, a suspension of beads 102 in appropriate liquid is applied to each region with the number of beads 102 applied being limited to the number of microwells in that region of the sample plate. The plate is then agitated causing each bead 102 to settle into a microwell of the sample plate. The plate is then allowed to dry.
[0030] Step five 210 of the method 200 includes depositing a MALDI matrix on the surface of the sample plate. In some methods of depositing a MALDI matrix, the cover and gasket are removed from the sample plate and a solution of MALDI matrix is applied onto the surface of the sample plate with a spraying apparatus. In some methods, the spraying apparatus produces very small droplets. After the MALDI matrix solution is sprayed on the surface of the sample plate, the matrix solution on the sample plate is then allowed to dry. The drying process causes rupture of the streptavidin-biotin bonds and rupture of the non-covalent bonds between bait molecules and strongly bound target molecules. The rupture causes a release of molecules that are then incorporated into the matrix crystals. The loaded sample plate with the MALDI matrix material applied is installed in a MALDI-TOF mass spectrometer.
[0031] In some methods of depositing a MALDI matrix, a solution of MALDI matrix is deposited directly on the surface of each region of the sample plate and allowed to dry. The drying process causes rupture of the streptavidin-biotin bonds and rupture of the non-covalent bonds between bait molecules and strongly bound target molecules. The rupture causes a release of molecules that are then incorporated into the matrix crystals. The cover and gasket are removed from the sample plate, and then the loaded sample plate with the MALDI matrix material applied is installed in a MALDI-TOF mass spectrometer.
[0032] Step six 212 of the method 200 includes performing MALDI-TOF mass spectroscopy to produce a mass spectra. MALDI-TOF mass spectroscopy is performed by scanning a laser beam over the surface of the sample plate to produce mass spectra. In some methods, the sample plate is moved to cause the laser beam to scan over the surface of the sample plate producing a mass spectrum from each location or pixel on the sample plate. In some embodiments, a mass spectrum is recorded from each microwell location in the sample plate. [0033] In some methods, the detection limit for MALDI-TOF spectroscopy is not limited by chemical noise. In these embodiments, the detection limit for MALDI-TOF spectroscopy is about 1 femtomole/mm2. The detection limit for MALDI-TOF spectroscopy corresponds to 1.3 attomoles/bead or equivalently 780,000 molecules/bead which is 0.04 pg for PSA. In some measurements, one bead may be sufficient to realize a desired detection limit for MALDI-TOF spectroscopy. In other measurements, multiple beads are used to produce lower detection limits, greater dynamic range, and better coefficient of variation CV, which is the ratio of the standard deviation of the peak intensity to the mean.
[0034] Step seven 214 of the method 200 includes analyzing the mass spectra to produce immunoassays. In some methods according to the present teaching, the analyzing the mass spectra comprises executing a computer algorithm that sums the mass spectra obtained for the predetermined mass of a mass tag together to produce a mass spectrum of the molecules that were originally bound to the beads by the streptavidin-biotin bonds and the target molecules that were bound to the bait molecules. In one specific method, a computer algorithm sums all of the mass spectra in which a predetermined mass tag is detected to produce a mass spectrum of the bound target molecules. Each given mass tag detected corresponds to a unique bait molecule. Thus, the mass spectra in which the mass tag is detected also include the mass of captured molecules that are strongly bound to the bait molecule.
[0035] FIG. 3 A illustrates a layout of a portion of a microwell sample plate of the ligand binding assay apparatus according to the present teaching. In the embodiment shown in FIG. 3A, the microwells 302 are 40 μπι in diameter and are arranged in a regular hexagonal array 304 with individual hexagons 306 having a 50 μπι height. FIG. 3A shows the size of a 25-μπι pixel 308 and a 10-μιη pixel 310 in relation to the size of an individual hexagon 306 and the regular hexagon array 304. In the example layout 300 shown in FIG. 3A, there are nominally 462 cells/mm2.
[0036] FIG. 3B shows a detailed view of one particular hexagon in the hexagonal array
304 shown in FIG. 3 A. The hexagon layout has two hexagonal dimensions, a 312 and b 314. In one particular example, the hexagon has a half-height of 25 μπι, which corresponds to a= 25 tan(30°), and b=50 cos(30°)-a, so that b+2a=57.72 μιη.
[0037] In some embodiments of the ligand binding assay apparatus according to the present teaching, the laser beam in MALDI-TOF mass spectrometer is rastered over the surface of a microwell plate comprising the layout 300 illustrated in FIG. 3A. In one particular method, the rastering is performed at intervals of 25 μιη with a 10-μιη diameter laser beam using laser repetition rate of 5 kHz, a scanning speed of 2.5 mm/s, and summing of 50 laser shots per pixel to produce 25 μιη long pixels. In this particular method, the total pixels/cell ratio is about 3.5 with about half on the well and half with significant contribution from adjacent wells. Total number of laser shots on each well is 100, with the laser irradiation time per cell equal to about 0.035s.
[0038] In another specific method, the rastering is performed over the surface of microwell plate comprising the layout 300 illustrated in FIG. 3A at intervals of 10 μπι with a 10- μπι diameter laser beam using laser repetition rate of 5 kHz, scanning speed of 1 mm/s, and summing of 50 laser shots per pixel to produce 10 μπι long pixels. In this particular method, the total pixels/cell ratio is about 25 with about 58% on the well having no significant contribution from adjacent wells. Total number of laser shots on the well is 600, and the laser irradiation time per cell is 0.25 seconds.
[0039] In another specific method, the rastering is performed over the surface of microwell plate comprising the layout 300 illustrated in FIG. 3A at intervals of 12.5 μπι with a 10-μπι diameter laser beam using laser repetition rate of 5 kHz, scanning speed of 1.25 mm/s, and summing of 50 laser shots per pixel to produce 12.5 μπι long pixels. In this particular method, the total pixels/cell ratio is about 16 with about 58% on the well and with no significant contribution from adjacent wells. Total number of laser shots on each well is 400 with the laser irradiation time equal to 0.16 seconds.
[0040] FIGS. 4A illustrates a layout 400 of an embodiment of a microwell sample plate
402 with division into regions according to the present teaching. In one particular plate according to the present teaching, the microwell plate 402 has nominally a 25x75 mm outside dimensions and comprises approximately 866,250 microwells. The microwell plate 402 is separated into regions or spots 404 by masks that are clamped to the plate with a gasket (not shown) that prevents liquid flow between the spots 404. The plate 400 shown in FIG. 4A illustrates a layout 400 comprising twenty-four spots 404 arranged in a 3x8 array. Each spot 404 is 7x7 mm and there is a 1-mm-long space between spots 404. Each spot 404 comprises approximately 22,600 microwells. The twenty-four spots include a total of about 542,400 wells. With a MALDI-TOF processing time of one hour per spot, the MALDI-TOF process completes in 24 hours per sample plate. A bar code 408 is attached to the plate at one end in a region 6x25 mm to identify the plate. [0041] FIG. 4B illustrates a layout 450 of another embodiment of a microwell sample plate with division into regions according to the present teaching. FIG. 4B illustrates a layout 450 that comprises ninety-six spots 406 arranged in an 8x20 array. Each spot 406 is 3x3 mm and there is a 1-mm-long space between spots 406. Each spot 406 comprises approximately 4100 microwells.
[0042] In another particular layout (not shown), the plate comprises three-hundred eighty-four spots arranged in a 12x32 array with each spot being dimensioned 1.5x1.5 mm and with a 0.5-mm-long space between spots. Each spot comprises approximately 1030 microwells. In yet another particular layout (not shown), the plate comprises one-thousand five hundred and thirty-six spots arranged in a 24x64 array with each spot being dimensioned 0.5x0.5 mm and with 0.5 mm space between spots. In this layout, each spot comprises approximately 115 microwells. Those skilled in the art will recognize that any number of spots can be arranged as needed for a particular application.
[0043] The methods of the present teaching are particularly well suited to producing a ligand binding assay from biotin. Biotin (MW = 244.3 Da), is a vitamin involved in several biological processes. These biological processes include, for example, cell growth and the citric acid cycle that is present, in low quantity, in all living cells. Streptavidin (MW ~ 52K), is a tetrameric protein purified from the bacterium Streptomyces Avidinii that is capable of binding 4 biotin molecules with extremely high affinity (Kd = 10-14M). Biotin is a relatively small molecule, which is easily bonded to peptides or aptamers with negligible impact on biological activity. Biotin / Streptavidin association occurs rapidly, and is relatively stable with
temperature changes, with organic solvents, with many denaturing agents, and is stable across pH range of about 4-9. These features of biotin and streptavidin chemistry make it a particularly useful tool for designing biological assay systems.
[0044] FIG. 5 illustrates a commercially available biotin linker 500 employed to covalently attach biotin to molecules of interest in an embodiment of the ligand binding assay apparatus and method of the present teaching. As illustrated in FIG. 5, the mass of biotinylated molecule is increased by 473.22 Da by this addition of the biotin. The masses of biotinylated peptides used for mass tags must be sufficiently different to be distinguished by the mass spectrometer. Taking into account the isotopic envelopes, a difference of 2000 ppm is adequate. The total number of theoretically distinguishable peaks, n, is 550 with a mass range, mn-m0; of 800-2400 Da and a resolution of 2000 ppm. This is calculated as follows: mn=m0(l+0.002)n n=log(3)/log(1.002)=550.
A list of peptides can be generated with a mass range, mn-m0j of 800-2400 Da and a resolution of 2000 ppm to approach this theoretical limit. These suitable peptides are chosen with biotinylated N-terminus and C-terminal arginine (R) and up to 12 other amino acid in order to obtain a relative uniform response factors with MALDI-TOF MS.
[0045] One feature of the ligand-binding assay apparatus and method of the present teaching is the ability to utilize a number of known and commercially available beads. For example, in some embodiments, purified Streptavidin Sepharose beads, which are commercially available through GE Healthcare Life Sciences as Product Code 17-5113-01 are used. These beads are composed of a highly cross-linked, chemically stable, rigid agarose core of about 34 microns in diameter. These beads are well suited for placement in a microtiter samples plate, one bead per 40^m-diameter well. In addition, these beads are specified to bind 6mg BSA/mL of beads.
[0046] Assuming that the volume of the beads in suspension is approximately
(0.0034)3cm3, then 1.0 mL of beads comprises approximately, 2.5xl07 beads/mL. The number of molecules can be determined from 6 mg BSA, which is equal to 90 nmoles or equivalently 5.4xl016 molecules. Then 5.4xl016 molecules/2.5xl07 beads is equal to 2.16xl09
molecules/bead. If one-tenth of the sites are occupied by mass tags, then ca. 2xl09 sites available ead. This results in 3.3 femtomoles/bead or equivalently 2.6 picomole/mm2 where the area irradiated is taken as the area of a 40^m-diameter well.
[0047] In other methods according to the present teaching, a biotinylated aptamers, which is commercially available from SomaLogic Inc., Boulder, CO, and biotinylated peptides, which is commercially available from 21st Century Biochemicals, Marlborough, MA, beads are used. These beads enable simple construction of a diverse array of targeted analyses. In embodiments comprising biotinylated aptamer beads, for example anti-Beta-2-microglobulin, which is commercially available from SomaLogic, Boulder, CO as product number B-3485- 28 2, the beads and a biotinylated mass tag are mixed the in ratio of 10: 1 (aptamer: mass tag) in 20 mM Tris pH 7.3 buffer, 5 mM EDTA (ethylenediaminetetraacetic acid), 1 M NaCl buffer solution. This mixture is allowed to incubate for 30 min. The incubated beads are then collected by centrifugation and washed three times in the same Tris pH 7.3 buffer in the absence of NaCl. The aptamer-to-peptide ratio is chosen to mimic the relative sensitivity for the target analyte and peptide mass tag in the mass spectrometer. The approximate number of binding sites on a single bead is estimated at 2x109.
[0048] In one method according to the present teaching, target analytes are sequestered from biological samples by using analyte specific aptamer reagents bound to beads. The beads also contain a mass-tag for bead identification and to aid in analyte signal averaging. Both the analyte specific affinity aptamers and the bead specific mass tags are non-covalently bound to the bead by a biotin / streptavidin linkage.
[0049] Some methods according to the present teaching use a plurality of sets of beads in the microtiter sample plate where each set of beads includes a unique mass tag and a unique bait molecule. In these methods, the mass spectrometer can operate in a multiplex mode of operation where mass spectra measurements can be taken on a plurality of different sets of beads and a computer algorithm is used to process the spectra to produce a ligand binding assay for each of the plurality of sets of beads.
[0050] Various computer algorithms can be used to process the mass spectra to perform various types of analysis. Some algorithms sum together all of the mass spectra in which a predetermined mass tag is detected to produce a mass spectrum of the bound molecules. Each given mass tag detected corresponds to a unique bait molecule in these algorithms. Thus, the mass spectra in which the mass tag is detected also include the mass of captured prey molecules. The summed mass spectrum for each mass tag is processed to yield mass and intensity for each mass peak detected. These include mass and intensity of the tag molecule ion, mass and intensity of ions produced by captured prey molecules, and may include ions produced from bait molecules. The ratio of the intensity of ions produced from prey molecules to the intensity of molecular ions from the mass tag may be used to determine the concentration of prey molecules in samples of interest. In some embodiments, this may require calibration of this ratio using molecular standards by techniques commonly used in quantitative applications of mass spectrometry.
Equivalents
[0051] While the Applicant's teaching are described in conjunction with various embodiments, it is not intended that the applicant's teaching be limited to such embodiments. On the contrary, the Applicant's teaching encompass various alternatives, modifications, and equivalents, as will be appreciated by those of skill in the art, which may be made therein without departing from the spirit and scope of the teaching.

Claims

What is claimed is: Claims
1. An apparatus for ligand binding assay, the apparatus comprising: a) an incubator that incubates a plurality of beads with a sample of interest, wherein each of the plurality of beads comprises a tag of predetermined mass and a bait molecule, the incubation binding the bait molecule to a sample molecule contained in the sample of interest; b) a washer that washes the incubated beads so that weakly bound sample molecules are removed while strongly bound sample molecules are retained; c) a sample plate configured for loading into a MALDI-TOF mass spectrometer; d) a sample plate loader that loads the incubated and washed beads into the sample plate so that a monolayer of beads is formed on the sample plate; e) a matrix material applicator that deposits matrix assisted laser desorption ionization (MALDI) matrix material onto a surface of the sample plate so that at least some of the plurality of beads are exposed to the MALDI matrix material; f) a matrix assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometer that receives the loaded sample plate and that performs time-of- flight mass spectrometry on the beads positioned on the sample plate, thereby generating mass spectra; and g) a computer that executes an algorithm using the mass spectra generated by the MALDI-TOF mass spectrometer to produce a ligand binding assay.
The apparatus for ligand binding assay of claim 1 wherein the sample plate defines a plurality of wells, each of the plurality of wells being dimensioned so that only one bead can be positioned in each well.
The apparatus for ligand binding assay of claim 1 wherein the plurality of beads comprises at least two sets of a plurality of beads, wherein each of the at least two sets comprises a mass tag and a bait molecule that are unique to that set.
The apparatus for ligand binding assay of claim 1 wherein each of the plurality of beads comprises a sepharose bead with immobilized streptavidin.
The apparatus for ligand binding assay of claim 4 wherein the mass tag molecules and the bait molecules are biotinylated and are bound to streptavidin immobilized on the sepharose beads.
The apparatus for ligand binding assay of claim 1 wherein the mass tag molecules and the bait molecules covalently attach biotin to at least one of a peptide, protein or a nucleic acid.
The apparatus for ligand binding assay of claim 1 wherein at least one of the plurality of beads is nominally 34 μπι in diameter.
The apparatus for ligand binding assay of claim 1 wherein at least one of the plurality of beads comprises biotinylated aptamers.
9. The apparatus for ligand binding assay of claim 1 wherein at least one of the plurality of beads comprises a biotinylated peptide.
10. The apparatus for ligand binding assay of claim 1 wherein the sample plate comprises a microwell sample plate.
11. The apparatus for ligand binding assay of claim 1 wherein the sample plate loader
comprises a vibrator.
12. The apparatus for ligand binding assay of claim 1 wherein the washer comprises a
microcentrifuge.
13. The apparatus for ligand binding assay of claim 1 wherein the matrix material applicator comprises a sprayer.
14. A method for producing a ligand binding assays, the method comprising: a) providing a plurality of beads, where each of the plurality of beads comprises a tag of predetermined mass and a bait molecule; b) incubating the plurality of beads with a sample of interest, the incubation binding the bait molecule to a sample molecule contained in the sample of interest; c) washing the incubated beads so that weakly bound sample molecules are removed while strongly bound sample molecules are retained; d) loading a sample plate comprising a plurality of wells with the incubated and washed beads such that respective ones of the plurality of beads are loaded into respective ones of the plurality of wells; e) adding matrix assisted laser desorption ionization (MALDI) matrix material on a surface of the loaded sample plate so that at least some of the plurality of beads are exposed to the MALDI matrix material; f) performing matrix assisted laser desorption ionization time-of-flight mass
spectrometry on at least some of the plurality of beads, thereby generating mass spectra; and g) processing the generated mass spectra to generate the ligand binding assays.
15. The method for producing a ligand binding assays of claim 14 wherein the providing the plurality of beads comprises providing at least two sets of plurality of beads, wherein each of the at least two sets comprises a mass tag and a bait molecule that are unique to that set.
16. The method for producing a ligand binding assays of claim 14 wherein the providing the plurality of beads comprises providing a sepharose bead with immobilized streptavidin.
17. The method for producing a ligand binding assays of claim 16 wherein the providing the plurality of beads comprises biotinylating the mass tag molecules and bait molecules bound to streptavidin immobilized on the sepharose beads.
18. The method for producing a ligand binding assays of claim 14 wherein the mass tag molecules and bait molecules covalently attach biotin to at least one of a peptide, protein or a nucleic acid.
19. The method for producing a ligand binding assays of claim 14 wherein the providing the plurality of beads comprises providing at least some beads that are nominally 34 μιη in diameter.
20. The method for producing a ligand binding assays of claim 14 wherein the providing the plurality of beads comprises providing at least some beads comprising biotinylated aptamers.
21. The method for producing a ligand binding assays of claim 14 wherein the providing the plurality of beads comprises providing at least some beads comprising biotinylated peptides.
22. The method for producing a ligand binding assays of claim 14 wherein the washing
comprises washing with a Tris pH 7.3 buffer solution.
23. The method for producing a ligand binding assays of claim 14 wherein the performing matrix assisted laser desorption ionization time-of-flight mass spectrometry comprises moving the loaded sample plate while rastering an ionizing laser beam.
24. The method for producing a ligand binding assays of claim 14 wherein the performing matrix assisted laser desorption ionization time-of-flight mass spectrometry comprises performing matrix assisted laser desorption ionization time-of-flight mass spectrometry in a multiplexed mode.
25. The method for producing a ligand binding assays of claim 14 wherein the processing the generated mass spectra comprises summing the generated mass spectra.
26. The method for producing a ligand binding assays of claim 14 further comprising drying the MALDI matrix material deposited on the surface of the sample plate to unbind at least one of the mass tag molecules, the prey molecules, or the bait molecules.
27. The method for producing a ligand binding assays of claim 14 further comprising
selecting a mass-tag for bead identification and to aid in analyte signal averaging.
PCT/US2018/012199 2017-01-05 2018-01-03 Ligand binding assays using maldi-tof mass spectrometry WO2018129055A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP18736271.0A EP3566052A4 (en) 2017-01-05 2018-01-03 Ligand binding assays using maldi-tof mass spectrometry
CN201880005874.2A CN110168369A (en) 2017-01-05 2018-01-03 It is measured using the ligand binding of MALDI-TOF mass spectrography

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762442512P 2017-01-05 2017-01-05
US62/442,512 2017-01-05

Publications (1)

Publication Number Publication Date
WO2018129055A1 true WO2018129055A1 (en) 2018-07-12

Family

ID=62711569

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/012199 WO2018129055A1 (en) 2017-01-05 2018-01-03 Ligand binding assays using maldi-tof mass spectrometry

Country Status (4)

Country Link
US (1) US11105797B2 (en)
EP (1) EP3566052A4 (en)
CN (1) CN110168369A (en)
WO (1) WO2018129055A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020256695A1 (en) * 2019-06-18 2020-12-24 Virgin Instruments Corporation Method and apparatus employing magnetic beads for ligand binding assays of biological samples
CN112799086B (en) * 2021-03-30 2023-07-14 上海思岚科技有限公司 Method, device and equipment for laser ranging

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7282707B1 (en) * 2005-06-30 2007-10-16 Thermo Finnigan Llc Method and apparatus for handling a sample plate for use in mass analysis
US20080090295A1 (en) * 2006-10-11 2008-04-17 Isabel Feuerstein Method and device for preparing an analyte for analysis by mass spectrometry
US20140274771A1 (en) * 2013-03-12 2014-09-18 Asociación Centro De Investigación Cooperative En Bioma Teriales Methods for making microarrays and their uses
US20160225596A1 (en) * 2015-02-01 2016-08-04 Virgin Instruments Corporation Method And Apparatus For Transporting Samples In A Mass Spectrometer

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19963536C2 (en) 1999-12-20 2003-04-10 Epigenomics Ag Procedure for the analysis of nucleic acid sequences
EP1358483A1 (en) * 2001-01-31 2003-11-05 Novozymes A/S Method of analysing granular composition by fluorescence analysis
US20040018515A1 (en) * 2002-04-03 2004-01-29 Diener John L. Compositions selective for adenosine diphosphate and methods of using same
WO2005123959A2 (en) * 2004-06-10 2005-12-29 Perkinelmer Las, Inc. Multiplexing assays for analyte detection
GB0809488D0 (en) * 2008-05-23 2008-07-02 Electrophoretics Ltd Mass spectrometric analysis
US9523680B2 (en) * 2010-06-30 2016-12-20 Ambergen, Inc. Global Proteomic screening of random bead arrays using mass spectrometry imaging
US20120202709A1 (en) 2011-02-09 2012-08-09 Adeptrix Corp. Devices and Methods for Producing and Analyzing Microarrays
WO2014176435A2 (en) 2013-04-25 2014-10-30 Bergo Vladislav B Microarray compositions and methods of their use

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7282707B1 (en) * 2005-06-30 2007-10-16 Thermo Finnigan Llc Method and apparatus for handling a sample plate for use in mass analysis
US20080090295A1 (en) * 2006-10-11 2008-04-17 Isabel Feuerstein Method and device for preparing an analyte for analysis by mass spectrometry
US20140274771A1 (en) * 2013-03-12 2014-09-18 Asociación Centro De Investigación Cooperative En Bioma Teriales Methods for making microarrays and their uses
US20160225596A1 (en) * 2015-02-01 2016-08-04 Virgin Instruments Corporation Method And Apparatus For Transporting Samples In A Mass Spectrometer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
See also references of EP3566052A4 *
ZHOU, YING ET AL.: "Proteome-wide drug screening using mass spectrometric imaging of bead-arrays", SCIENTIFIC REPORTS, vol. 6, no. 1, 19 May 2016 (2016-05-19), pages 1 - 8, XP055511841 *

Also Published As

Publication number Publication date
US20180188241A1 (en) 2018-07-05
CN110168369A (en) 2019-08-23
US11105797B2 (en) 2021-08-31
EP3566052A1 (en) 2019-11-13
EP3566052A4 (en) 2020-08-19

Similar Documents

Publication Publication Date Title
US11131674B2 (en) Microarray compositions and methods of their use
US20160008785A1 (en) Devices and methods for producing and analyzing microarrays
Poetz et al. Protein microarrays: catching the proteome
JP4587954B2 (en) Protein interaction difference mapping
Oh et al. Calixarene derivative as a tool for highly sensitive detection and oriented immobilization of proteins in a microarray format through noncovalent molecular interaction
Loo Mass spectrometry in the combinatorial chemistry revolution
US11105797B2 (en) Ligand binding assays using MALDI-TOF mass spectrometry
CA2682543A1 (en) Immunoassays and characterization of biomolecular interactions using self- assembled monolayers
US20070249060A1 (en) Determination of Proteins and/or Other Molecules Using Mass Spectroscopy
WO2001084146A2 (en) A sensitive, multiplexed mass-spectrometry-based biosensor based on immuno-pcr
Ha et al. Rapid characterization of protein chips using microwave-assisted protein tryptic digestion and MALDI mass spectrometry
Spera et al. Label free detection of NAPPA via mass spectrometry
US20060199279A1 (en) Methods for identifying post-translationally modified polypeptides
US20060223194A1 (en) Methods of screening for post-translationally modified proteins
Joos Protein microarray technology
US20190271707A1 (en) Method and Apparatus Employing Magnetic Beads for Ligand Binding Assays of Biological Samples
JP4543860B2 (en) Sample analysis method
US20020068367A1 (en) Linker and method for solid phase combinatorial synthesis
WO2020256695A1 (en) Method and apparatus employing magnetic beads for ligand binding assays of biological samples
JP2005283351A (en) Protein immobilizing method
US20130345093A1 (en) Method for Detection and Quantification of Target Biomolecules
PRESTIGIACOMO et al. Detection of autoantibodies using protein arrays
JP2004510134A (en) Method for producing and determining an appropriate effector of a target molecule using a substance library
Morris INTRODUCTION TO PROTEIN CHIP TECHNOLOGIES
JP2004012176A (en) Protein detection method

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18736271

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018736271

Country of ref document: EP

Effective date: 20190805