EP3867943A1 - Funktionalisierung eines probennahmeelements zur verwendung mit einem massenspektrometriesystem - Google Patents

Funktionalisierung eines probennahmeelements zur verwendung mit einem massenspektrometriesystem

Info

Publication number
EP3867943A1
EP3867943A1 EP19790858.5A EP19790858A EP3867943A1 EP 3867943 A1 EP3867943 A1 EP 3867943A1 EP 19790858 A EP19790858 A EP 19790858A EP 3867943 A1 EP3867943 A1 EP 3867943A1
Authority
EP
European Patent Office
Prior art keywords
silicate
sampling element
antibody
elongate
elongate sampling
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
EP19790858.5A
Other languages
English (en)
French (fr)
Inventor
Thomas R. Covey
Chang Liu
Subhasish Purkayastha
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
DH Technologies Development Pte Ltd
Original Assignee
DH Technologies Development Pte Ltd
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 DH Technologies Development Pte Ltd filed Critical DH Technologies Development Pte Ltd
Publication of EP3867943A1 publication Critical patent/EP3867943A1/de
Pending legal-status Critical Current

Links

Classifications

    • HELECTRICITY
    • H01ELECTRIC ELEMENTS
    • H01JELECTRIC DISCHARGE TUBES OR DISCHARGE LAMPS
    • H01J49/00Particle spectrometers or separator tubes
    • H01J49/02Details
    • H01J49/04Arrangements for introducing or extracting samples to be analysed, e.g. vacuum locks; Arrangements for external adjustment of electron- or ion-optical components
    • 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

Definitions

  • the present disclosure relates to mass spectrometry, and more particularly to systems, apparatus, and methods useful for sampling analytes in mass spectrometry systems.
  • Mass spectrometry is an analytical technique for determining the elemental composition of test substances with both qualitative and quantitative applications.
  • mass spectrometry systems can be used to identify unknown compounds, to determine the isotopic composition of elements in a molecule, and to determine the structure of a particular compound by observing its pattern of fragmentation, as well as to quantify the amount of a particular compound in the sample.
  • sample molecules are generally introduced by a process known as ionization.
  • the sample is converted into ions using an ion source.
  • Ions are typically generated upstream at atmospheric pressure (e.g., by chemical ionization, electrospray) before they pass through an inlet orifice and enter an ion guide disposed in a vacuum chamber.
  • the ions are then separated and detected downstream by one or more mass analyzers.
  • the present disclosure encompasses a recognition that there is a need in mass spectrometry systems for methods and apparatus for simply and discretely preparing and introducing biologic based samples into mass spectrometry systems for detection and analysis with high sensitivity and specificity.
  • the present disclosure provides methods of sampling analytes in mass spectrometry systems.
  • methods include functionalizing an outer surface of a sampling element with a polypeptide that preferentially and/or selectively binds to at least one analyte; exposing the sampling element to a sample; and inserting the sampling element to a sampling interface of the mass spectrometry system, such that if that analyte is present in the sample and the analyte comes in contact with the polypeptide, then at least a portion of the analyte can preferentially and/or specifically bind to the polypeptide and be sampled by the mass spectrometry system.
  • methods can include a step of providing an elongate sampling element, which can include an outer surface extending from a first end to a second end that terminates at a distal surface.
  • the second end can be configured to be inserted within a sampling interface of the mass spectrometry system.
  • the outer surface of the elongate sampling element can be functionalized, for example, with at least one polypeptide, where the polypeptide preferentially binds to an analyte of interest.
  • methods can include functionalizing at least a portion of an outer surface of an elongate sampling element.
  • methods can include functionalizing the outer surface of the sampling element, without limitation, for example, with at least one polypeptide, at least one antibody or fragment thereof, with at least one oligopeptide or fragment thereof, with at least one peptide or fragment thereof, with at least one protein or fragment thereof, with at least one antigen or fragment thereof.
  • methods can include functionalizing at least a portion of an outer surface of an elongate sampling element, for example, with at least one polypeptide can specifically include the step of functionalizing the at least a portion of the elongate sampling element with at least one antibody or fragment thereof.
  • At least a part of the outer surface of the elongate sampling element can be activated, for example via exposure to one or more reagents prior to bonding a polypeptide thereto. In some embodiments, at least a part of the outer surface of the elongate sampling element can be activated, for example, via exposure to at least one reagent prior to bonding an antibody thereto. In some embodiments, the step of functionalizing the at least a portion of the elongate sampling element with such an antibody or fragment thereof can include a step of applying an aminosilane reagent to at least a portion of the outer surface of the elongate sampling element.
  • the aminosilane reagent can be (3- aminopropyl)triethoxysilane (APTES).
  • the step of functionalizing at least a portion of the elongate sampling element can further include a step of reacting the outer surface amine of the elongate sampling element with glutaraldehyde.
  • the step of functionalizing at least a portion of the elongate sampling element can further include a step of immobilizing the at least one antibody or fragment thereof to the glutaraldehyde, thereby functionalizing at least a portion of the outer surface of the elongate sampling element.
  • the step of functionalizing at least a portion of the elongate sampling element with such an antibody or fragment thereof can include a step of applying an aminosilane reagent to at least a portion of the outer surface of the elongate sampling element.
  • the aminosilane reagent can be (3-aminopropyl)triethoxysilane (APTES).
  • the step of functionalizing can further include a step of reacting the outer surface amine of the elongate sampling element with N-hydroxysuccinimide (NHS) or sulfo- NHS.
  • the step of functionalizing can further include a step of
  • the step of functionalizing at least a portion of the elongate sampling element with such an antibody or fragment thereof can include a step of applying an aminosilane reagent to at least a portion of the outer surface of the elongate sampling element.
  • the aminosilane reagent can be (3-aminopropyl)triethoxysilane (APTES).
  • the step of functionalizing the at least a portion of the elongate sampling element can further include a step of reacting the outer surface amine of the elongate sampling element with maleimide.
  • the step of functionalizing at least a portion of the elongate sampling element with the at least one antibody or fragment thereof can further include a step of immobilizing the at least one antibody or fragment thereof to the maleimide, thereby functionalizing at least a portion of the outer surface of the elongate sampling element.
  • the present disclosure further teaches methods of exposing at least a portion of the polypeptide-functionalized elongate sampling element to a sample, such as a bodily fluid sample.
  • the step of exposing can include a step of mixing, for example, agitating and/or stirring, the elongate sampling element in a bodily fluid sample.
  • the step of exposing the polypeptide-functionalized elongate sampling element can include exposing the sampling element to a sample of blood, blood product, saliva, vomit, urine, tear, sweat, bile, milk, cerebrospinal fluid, feces, body secretion, pus, mucus, lymph, gastric acid juice, earwax, blister, humoral fluid, intracellular fluid, extracellular fluid, human fluid, animal fluid, plant fluid, rinsate of solid, rinsate of surface, fluid extract, or any combination thereof.
  • an analyte in a sample can include a polypeptide, a protein, in some embodiments, the protein can be an antibody or fragment thereof.
  • the analyte if it is present in the bodily fluid sample, preferentially and/or specifically binds to the at least one polypeptide functionalized to the sample element.
  • the polypeptide functionalized on the outer surface of the elongate sampling element is characterized in that it preferentially binds to at least one analyte, such that if the analyte is present in the bodily fluid sample and the analyte comes in contact with the polypeptide, then the analyte can bind to the polypeptide.
  • the analyte, if present in the bodily fluid sample and/or the biologic sample can, without limitation, for example, be an antibody or fragment thereof, a peptide or fragment thereof, a polypeptide, and/or a protein or fragment thereof.
  • methods as provided herein can include a step of inserting at least a portion of the elongate sampling element into the sampling interface of the mass spectrometry system.
  • the outer surface of the inserted sampling element is coated with a polypeptide-bound analyte.
  • methods of sampling can further include a step of inserting the sample-exposed portion of the elongate sampling element into a sampling interface of the mass spectrometry system, so that the sample-exposed portion is positioned to contact an extraction solvent flowing through the sampling interface to deliver at least a portion of the analyte to an ion source of the mass spectrometry system.
  • methods further include a step of contacting at least a portion of the elongate sampling element with an extraction solvent.
  • methods can include a step of flowing the extraction solvent through the sampling interface so as to extract at least a portion of the polypeptide-bound analyte and introduce the extracted analyte to an ion source of the mass spectrometry system.
  • an extraction solvent contacts at least a portion of the polypeptide-bound analyte that is coated on the outer surface of the elongate sampling element.
  • the present disclosure provides methods of carrying, delivering, and/or transmitting ions of the extracted analytes to one or more downstream components of the mass spectrometry system, including for example an ion source.
  • the present disclosure provides methods of transmitting ions of the extracted analytes to one or more downstream components of the mass spectrometry system, including a mass analyzer for detection thereof.
  • methods disclosed herein further include a step of performing a mass spectrometric analysis on the extracted analyte.
  • sampling methods according to the present teachings can exhibit a sensitivity sufficient to detect a biologic, for example, testosterone, from serum/plasma, at a concentration as low as 0.1 pg/mL.
  • the present disclosure further provides an elongate sampling element, which is configured for insertion in a sampling interface for use with a mass spectrometry system.
  • the elongate sampling element includes an outer surface extending from a first end to a second end.
  • the outer surface of the elongate sampling element for example, its second end, can include a coating disposed on a least a portion thereof.
  • the coating is a functionalized coating.
  • the functionalized coating includes at least one polypeptide immobilized on the outer surface of the second end of the elongate sampling element.
  • the at least one polypeptide is characterized in that it can preferentially bind to at least one analyte.
  • the elongate sampling element can be formed from or can include, for example, alumina silicate, antimony silicate, arsenic silicate, barium silicate, bismuth silicate, boron silicate, cadmium silicate, gallium silicate, germanium silicate, glass, gold silicate, lead silicate, lime silicate, lithium silicate, magnesium silicate, nickel, nitrogen silicate, platinum silicate, silica, sodium silicate, phosphorus silicate, potassium silicate, tin silicate, indium silicate, silver silicate, zinc silicate, or any combination thereof.
  • the elongate sampling element can exhibit properties, for example, including magnetic properties.
  • the elongate sampling element can be heated, cooled, and/or have a field applied thereto.
  • the second end of the elongate sampling element can include one or more protrusions or patterns of protrusions that project from at least a portion of the outer surface of the second end of the elongate sampling element.
  • the protrusions can include bead or bead-like structures.
  • such beads can increase or enhance surface area of the outer surface thereby enhancing the capability of the elongate sampling element to capture analytes of interest from the sample.
  • the distal surface of the sampling element can have a variety of cross-section shapes.
  • the cross-sectional shape of the distal surface can be that of a square, a diamond, a star having 5 points, a star having 6 points, a star having 7 points, a star having 8 points, a star having 9 points, a star having 10 points, or a star having any number of points that are bent or angled.
  • apparatus, systems and methods of the present disclosure provide enhanced sensitivity and specificity for introduction of biologic based samples to mass spectrometry systems as provided herein.
  • functionalizing an outer surface of an elongate sampling element with a polypeptide that preferentially binds to at least one analyte can result in an enhanced sensitivity.
  • exposing the elongate sampling element and extracting the analyte from the elongate sampling element as disclosed herein can result in an enhanced selectivity.
  • FIG. 1 in a schematic diagram, illustrates a mass spectrometry system having sample introduction apparatus in accordance with one aspect of various embodiments of the present disclosure
  • FIG. 2 in a schematic diagram, depicts elongate sampling elements in accordance with one aspect of various embodiments of the present disclosure.
  • FIG. 2 at panel (A) shows a non-roughened elongate sampling element.
  • FIG. 2 at panel (B) shows a roughened elongate sampling element;
  • FIG. 3 in a schematic diagram, depicts elongate sampling elements in accordance with one aspect of various embodiments of the present disclosure
  • FIG. 4 in a schematic diagram, illustrates an end cross-sectioned view of a distal surface of an elongate sampling element in accordance with one aspect of various embodiments of the present disclosure
  • FIG. 5 in a schematic diagram, illustrates another end cross-sectioned view of a distal surface of an elongate sampling element in accordance with one aspect of various embodiments of the present disclosure
  • FIG. 7 illustrates steps of a method sampling an analyte in accordance with one aspect of various embodiments of the present disclosure
  • FIG. 8 in a schematic diagram, illustrates a sampling interface of a mass spectrometry system in accordance with one aspect of various embodiments of the present disclosure
  • FIG. 9 illustrates steps of a method for functionalizing an outer surface of an elongate sampling element in accordance with one aspect of various embodiments of the present disclosure
  • FIG. 10 in a schematic diagram, illustrates steps for immobilizing an antibody or fragment thereof on an outer surface of an elongate sampling element and binding an analyte thereto in accordance with one aspect of various embodiments of the present disclosure
  • FIG. 11 in a schematic diagram, illustrates another set of steps for immobilizing an antibody or fragment thereof on an outer surface of an elongate sampling element and binding an analyte thereto in accordance with one aspect of various embodiments of the present disclosure.
  • the term“approximately” or“about” refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
  • the term“a” may be understood to mean“at least one.”
  • the term“or” may be understood to mean“and/or.”
  • the terms“comprising” and“including” may be understood to encompass itemized components or steps whether presented by themselves or together with one or more additional components or steps.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic hydrocarbon radical, or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di- and multivalent radicals, having the number of carbon atoms designated (i.e. Ci -6 means one to six carbons).
  • saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, homologs and isomers of, for example, n-pentyl, n-hexyl, and the like.
  • An unsaturated alkyl group is one having one or more double bonds or triple bonds.
  • alkyl groups examples include, but are not limited to, vinyl, 2-propenyl, 2-isopentenyl, 2-(butadienyl), 2,4- pentadienyl, 3-(l,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • alkyl unless otherwise noted, is also meant to include those derivatives of alkyl defined in more detail below, such as“heteroalkyl.”
  • the definition of each expression, e.g. alkyl, m, n, and the like, when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
  • alkoxyl refers to an alkyl group, as defined herein, having an oxygen radical attached thereto.
  • alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like.
  • the alkyl portion of an alkoxy group is sized like the alkyl groups, and can be substituted by the same groups that are suitable as substituents on alkyl groups, to the extent permitted by the available valences.
  • amino acid in its broadest sense, refers to any compound and/or substance that can be incorporated into a polypeptide chain, e.g., through formation of one or more peptide bonds.
  • an amino acid has the general structure H2N-C(H)(R)-COOH.
  • an amino acid is a naturally- occurring amino acid.
  • an amino acid is a synthetic amino acid; in some
  • an amino acid is a D-amino acid; in some embodiments, an amino acid is an L- amino acid.
  • Standard amino acid refers to any of the twenty standard L-amino acids commonly found in naturally occurring peptides.
  • Nonstandard amino acid refers to any amino acid, other than the standard amino acids, regardless of whether it is prepared synthetically or obtained from a natural source.
  • an amino acid, including a carboxy - and/or amino-terminal amino acid in a polypeptide can contain a structural modification as compared with the general structure herein.
  • an amino acid may be modified by methylation, amidation, acetylation, and/or substitution as compared with the general structure.
  • such modification may, for example, alter the circulating half-life of a polypeptide containing the modified amino acid as compared with one containing an otherwise identical unmodified amino acid. In some embodiments, such modification does not significantly alter a relevant activity of a polypeptide containing the modified amino acid, as compared with one containing an otherwise identical unmodified amino acid.
  • the term“amino acid” is used to refer to a free amino acid; in some embodiments, it is used to refer to an amino acid residue of a polypeptide.
  • the term“antibody” refers to an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, steroid, etc., through at least one antigen recognition site, located in the variable domain of the immunoglobulin molecule.
  • a target such as a carbohydrate, polynucleotide, lipid, polypeptide, steroid, etc.
  • the term encompasses not only intact polyclonal or monoclonal antibodies, but also, unless otherwise specified, any antigen-binding portion thereof that competes with the intact antibody for specific binding, fusion proteins comprising an antigen-binding portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site.
  • Antigen-binding portions include, for example, Fab, Fab', F(ab')2, Fd, Fv, domain antibodies (dAbs, e.g., shark and camelid antibodies), portions including complementarity determining regions (CDRs), single chain variable fragment antibodies (scFv), maxibodies, minibodies, intrabodies, diabodies, triabodies, tetrabodies, v- NAR and bis-scFv, and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide.
  • immunoglobulins can be assigned to different classes.
  • immunoglobulins There are five major classes (i.e., isotypes) of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (subtypes), e.g., IgG-i , lgG 2 , lgG3, lgG 4 , IgAi and lgA 2 .
  • the heavy-chain constant regions that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • the term“antigen (Ag)” refers to the molecular entity used for immunization of an immunocompetent vertebrate to produce the antibody (Ab) that recognizes the Ag or to screen an expression library (e.g., phage, yeast or ribosome display library, among others).
  • Ag is termed more broadly and is generally intended to include target molecules that are specifically recognized by the antibody or fragment thereof, thus including portions or mimics of the molecule used in an immunization process for raising the antibody or fragment thereof or in library screening for selecting the antibody or fragment thereof.
  • full-length IL-2 from mammalian species e.g., human, monkey, mouse and rat IL-2
  • monomers and multimers such as dimers, trimers, etc. thereof, as well as truncated and other variants of IL-2
  • an antigen e.g., human, monkey, mouse and rat IL-2
  • the terms“antigen-binding portion” or“antigen-binding fragment” of an antibody refers to one or more portions of an antibody that retain the ability to specifically bind to an antigen (e.g., IL-2). It has been shown that the antigen-binding function of an antibody can be performed by portions of a full-length antibody.
  • binding portions encompassed within the term“antigen-binding portion” of an antibody include (i) a Fab portion, a monovalent portion consisting of the VL, VH, CL and CH-i domains; (ii) a F(ab')2 portion, a bivalent portion comprising two Fab portions linked by a disulfide bridge at the hinge region; (iii) a Fd portion consisting of the VH and CHi domains; (iv) a Fv portion consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb portion (Ward et al, (1989) Nature 341 : 544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR), disulfide-linked Fvs (dsFv), and anti-idiotypic (anti-id) antibodies and intrabodies.
  • CDR complementarity determining region
  • dsFv disulfide-linked
  • the two domains of the Fv portion, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv)); see e.g., Bird et al. Science 242:423-426 (1988) and Huston et al. Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988)).
  • scFv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term“antigen-binding portion” of an antibody.
  • Other forms of single chain antibodies, such as diabodies are also encompassed.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger ef a/. Proc. Natl. Acad.
  • aryl means, unless otherwise stated, a substituted or unsubstituted polyunsaturated, aromatic, hydrocarbon substituent which can be a single ring or multiple rings (preferably from 1 to 3 rings) having 3 to 10 or alternatively 3 to 7 members which are fused together or linked covalently.
  • binding affinity is herein used as a measure of the strength of a non-covalent interaction between two molecules, e.g., an antibody or portion thereof and an antigen.
  • the term“binding affinity” is used to describe monovalent interactions (intrinsic activity). Binding affinity between two molecules may be quantified by determination of the dissociation constant (KD). In turn, KD can be determined by measurement of the kinetics of complex formation and dissociation using, e.g., the surface plasmon resonance (SPR) method (Biacore).
  • SPR surface plasmon resonance
  • the rate constants corresponding to the association and the dissociation of a monovalent complex are referred to as the association rate constants ka (or kon) and dissociation rate constant kd (or koff), respectively.
  • the value of the dissociation constant can be determined directly by well-known methods, and can be computed even for complex mixtures by methods such as those set forth in Caceci et al. (1984, Byte 9: 340-362).
  • the KD may be established using a double- filter nitrocellulose filter binding assay such as that disclosed by Wong & Lohman (1993, Proc. Natl. Acad. Sci. USA 90: 5428-5432).
  • chimeric antibody is intended to refer to antibodies in which the variable domain sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable domain sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody or vice versa.
  • the term also encompasses an antibody comprising a V region from one individual from one species (e.g., a first mouse) and a constant region from another individual from the same species (e.g., a second mouse).
  • the term“contact residue” as used herein with respect to an antibody or the antigen specifically bound thereby refers to an amino acid residue present on an antibody/antigen comprising at least one heavy atom (i.e., not hydrogen) that is within 4 A or less of a heavy atom of an amino acid residue present on the cognate antibody/antigen.
  • a“constant region” of an antibody refers to the constant region of the antibody light chain or the constant region of the antibody heavy chain, either alone or in combination.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and at least one heteroatom selected from the group consisting of O, N, Si and S, and wherein the nitrogen, carbon and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule. Examples include, but are not limited to, -CH2 -CH2 -O-CH3 , -CH2 -CH2 -NH-CH3
  • heteroalkyl encompass poly( ethylene glycol) and its derivatives.
  • heteroaryl refers to aryl groups that contain from one to four heteroatoms selected from N, O, and S, wherein the nitrogen, carbon and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • Non-limiting examples of aryl and heteroaryl groups include phenyl, 1 -naphthyl, 2-naphthyl, 4-biphenyl, l-pyrrolyl, 2- pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4- thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3 -thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2- pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-ind
  • aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below.
  • “Aryl” and“heteroaryl” also encompass ring systems in which one or more non-aromatic ring systems are fused, or otherwise bound, to an aryl or heteroaryl system, such as a benzodioxolyl (e.g., l,3-benzodioxol-5-yl), benzofuran, isobenzofuran, indole, isoindole, indoxazine, indazole, benzoxazole, and anthranil.
  • the heteroaryl is a thiophene, isoxazole, tetrahydrofuran, pyridyl, benzofuran, or furanopyridine.
  • R', R", R'" and R" each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, e.g., aryl substituted with 1 -3 halogens, substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups.
  • each of the R groups is independently selected as are each R', R", R'" and R"" groups when more than one of these groups is present.
  • alkyl is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g.,— CF 3 and— CH 2 CF 3 ).
  • the term“alkyl” will also include groups including acyl (e.g.,— C(0)CH 3 ,— C(0)CF 3 ,— C(0)CH 2 OCH3, and the like).
  • acyl e.g.,— C(0)CH 3 ,— C(0)CF 3 ,— C(0)CH 2 OCH3, and the like.
  • heteroatom includes oxygen (O), nitrogen (N), sulfur (S) and silicon (Si).
  • halo or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom.
  • the neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
  • solvate as used herein, is a molecular or ionic complex of molecules or ions of a solvent with molecules or ions. When water is the solvent, the molecule is referred to as a“hydrate”.
  • stereoisomers refers to compounds whose molecules have the same number and kind of atoms and the same atomic arrangement, but differ in their spatial arrangement. Where stereochemistry is not specifically indicated, all stereoisomers of the compounds provided herein are included within the scope of this disclosure, as pure isomers as well as mixtures thereof. Unless otherwise indicated, individual enantiomers, diastereomers, geometrical isomers, and combinations and mixtures thereof are all encompassed by the present disclosure.
  • human antibody is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human and/or has been made using any of the techniques for making human antibodies as disclosed herein. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen binding residues.
  • the term“isolated molecule” (where the molecule is, for example, a polypeptide, a polynucleotide, or an antibody or portion thereof) is a molecule that by virtue of its origin or source of derivation (1 ) is not associated with naturally associated components that accompany it in its native state, (2) is substantially free of other molecules from the same species (3) is expressed by a cell from a different species, or (4) does not occur in nature.
  • a molecule that is chemically synthesized, or expressed in a cellular system different from the cell from which it naturally originates will be“isolated” from its naturally associated components.
  • a molecule also may be rendered substantially free of naturally associated components by isolation, using purification techniques well known in the art.
  • Molecule purity or homogeneity may be assayed by a number of means well known in the art.
  • the purity of a polypeptide sample may be assayed using polyacrylamide gel electrophoresis and staining of the gel to visualize the polypeptide using techniques well known in the art.
  • higher resolution may be provided by using HPLC or other means well known in the art for purification.
  • the term“epitope” refers to the area or region of an antigen to which an antibody specifically binds, i.e., an area or region in physical contact with the antibody.
  • the term“epitope” refers to that portion of a molecule capable of being recognized by and bound by an antibody at one or more of the antibody's antigen-binding regions.
  • an epitope is defined in the context of a molecular interaction between an“antibody, or antigen binding portion thereof (Ab), and its corresponding antigen.
  • Epitopes often consist of a surface grouping of molecules such as amino acids or sugar side chains and have specific three- dimensional structural characteristics as well as specific charge characteristics.
  • the epitope can be a protein epitope. Protein epitopes can be linear or
  • A“nonlinear epitope” or“conformational epitope” comprises noncontiguous polypeptides (or amino acids) within the antigenic protein to which an antibody specific to the epitope binds.
  • the term“antigenic epitope” as used herein, is defined as a portion of an antigen to which an antibody can specifically bind as determined by any method well known in the art, for example, by conventional immunoassays. Alternatively, during the discovery process, the generation and characterization of antibodies may elucidate information about desirable epitopes.
  • the term“monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally- occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present disclosure may be made by the hybridoma method first described by Kohler and Milstein, 1975, Nature 256:495, or may be made by recombinant DNA methods such as described in U.S. Pat. No. 4,816,567.
  • the monoclonal antibodies may also be isolated from phage libraries generated using the techniques described in McCafferty et al., 1990, Nature 348:552-554, for example.
  • humanized antibody refers to forms of non-human (e.g., murine) antibodies that are chimeric immunoglobulins, immunoglobulin chains, or portions thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a CDR of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity.
  • the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance.
  • polypeptide refers to a polymer of at least three amino acids, linked to one another by peptide bonds.
  • the term is used to refer to specific functional classes of polypeptides.
  • the present specification provides several examples of amino acid sequences of known exemplary polypeptides within the class; in some embodiments, such known polypeptides are reference polypeptides for the class.
  • polypeptide refers to any member of the class that shows significant sequence homology or identity with a relevant reference polypeptide. In many embodiments, such member also shares significant activity with the reference polypeptide.
  • such member also shares a particular characteristic sequence element with the reference polypeptide (and/or with other polypeptides within the class; in some embodiments, with all polypeptides within the class).
  • a member polypeptide shows an overall degree of sequence homology or identity with a reference polypeptide that is at least about 30-40%, and is often greater than about 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more and/or includes at least one region (i.e., a conserved region that may in some embodiments, may be or comprise a characteristic sequence element) that shows very high sequence identity, often greater than 90% or even 95%, 96%, 97%, 98%, or 99%.
  • Such a conserved region usually encompasses at least 3- 4 and often up to 20 or more amino acids; in some embodiments, a conserved region encompasses at least one stretch of at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more contiguous amino acids.
  • a useful polypeptide may comprise or consist of a fragment of a parent polypeptide.
  • a useful polypeptide as may comprise or consist of a plurality of fragments, each of which is found in the same parent polypeptide in a different spatial arrangement relative to one another than is found in the polypeptide of interest (e.g., fragments that are directly linked in the parent may be spatially separated in the polypeptide of interest or vice versa, and/or fragments may be present in a different order in the polypeptide of interest than in the parent), so that the polypeptide of interest is a derivative of its parent polypeptide.
  • a polypeptide may comprise natural amino acids, non-natural amino acids, or both.
  • a polypeptide may comprise only natural amino acids or only non-natural amino acids.
  • a polypeptide may comprise D-amino acids, L-amino acids, or both. In some embodiments, a polypeptide may comprise only D-amino acids. In some embodiments, a polypeptide may comprise only L-amino acids. In some embodiments, a polypeptide may include one or more pendant groups, e.g., modifying or attached to one or more amino acid side chains, and/or at the polypeptide's N- terminus, the polypeptide's C-terminus, or both. In some embodiments, a polypeptide may be cyclic. In some embodiments, a polypeptide is not cyclic. In some embodiments, a polypeptide is linear.
  • an antibody that“preferentially binds” or“specifically binds” is an antibody that“preferentially binds” or“specifically binds”
  • a molecule is said to exhibit“specific binding” or“preferential binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances.
  • an antibody that specifically or preferentially binds to an IL-2 epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other IL-2 epitopes or non-IL-2 epitopes.
  • an antibody which specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target.
  • “specific binding” or“preferential binding” does not necessarily require (although it can include) exclusive binding.
  • the term“protein” refers to a polypeptide (i.e., a string of at least two amino acids linked to one another by peptide bonds). Proteins may include moieties other than amino acids (e.g., may be glycoproteins, proteoglycans, etc.) and/or may be otherwise processed or modified. Those of ordinary skill in the art will appreciate that a“protein” can be a complete polypeptide chain as produced by a cell (with or without a signal sequence), or can be a characteristic portion thereof. Those of ordinary skill will appreciate that a protein can sometimes include more than one polypeptide chain, for example linked by one or more disulfide bonds or associated by other means.
  • Polypeptides may contain L-amino acids, D- amino acids, or both and may contain any of a variety of amino acid modifications or analogs known in the art. Useful modifications include, e.g., terminal acetylation, amidation, methylation, etc.
  • proteins may comprise natural amino acids, non-natural amino acids, synthetic amino acids, and combinations thereof.
  • the term“peptide” is generally used to refer to a polypeptide having a length of less than about 100 amino acids, less than about 50 amino acids, less than 20 amino acids, or less than 10 amino acids.
  • proteins are antibodies, antibody fragments, biologically active portions thereof, and/or characteristic portions thereof.
  • variable domain of an antibody refers to the variable domain of the antibody light chain (VL) or the variable domain of the antibody heavy chain (VH), either alone or in combination.
  • VL variable domain of the antibody light chain
  • VH variable domain of the antibody heavy chain
  • the variable domains of the heavy and light chains each consist of four framework regions (FRs) connected by three complementarity determining regions (CDRs) also known as hypervariable regions, and contribute to the formation of the antigen-binding site of antibodies.
  • FRs framework regions
  • CDRs complementarity determining regions
  • variants of a subject variable domain are desired, particularly with substitution in amino acid residues outside a CDR (i.e., in the framework region)
  • appropriate amino acid substitution in some embodiments, conservative amino acid substitution, can be identified by comparing the subject variable domain to the variable domains of other antibodies which contain CDR1 and CDR2 sequences in the same canonical class as the subject variable domain (see, e.g., Chothia and Lesk, J. Mol. Biol. 196(4): 901 -917, 1987).
  • definitive delineation of a CDR and identification of residues comprising the binding site of an antibody is accomplished by solving the structure of the antibody and/or solving the structure of the antibody-ligand complex.
  • various methods of analysis can be employed to identify or approximate the CDRs.
  • various methods of analysis can be employed to identify or approximate the CDRs. Examples of such methods include, but are not limited to, the Kabat definition, the Chothia definition, the AbM definition, the contact definition, the conformational definition and the IMGT definition.
  • the Kabat definition is a standard for numbering the residues in an antibody and is typically used to identify CDR regions. See, e.g., Johnson & Wu, 2000, Nucleic Acids Res., 28: 214-8.
  • the Chothia definition is similar to the Kabat definition, but the Chothia definition takes into account positions of certain structural loop regions. See, e.g., Chothia et ai, 1986, J. Mol. Biol., 196: 901 -17; Chothia et ai, 1989, Nature, 342: 877-83.
  • the AbM definition uses an integrated suite of computer programs produced by Oxford Molecular Group that model antibody structure. See, e.g., Martin et al, 1989, Proc Natl Acad Sci (USA), 86:9268-9272;“AbMTM, A Computer Program for Modeling
  • the AbM definition models the tertiary structure of an antibody from primary sequence using a combination of knowledge databases and antibody or fragment thereof initio methods, such as those described by Samudrala et al., 1999,“Ab Initio Protein Structure Prediction Using a Combined
  • the contact definition is based on an analysis of the available complex crystal structures. See, e.g., MacCallum et al, 1996, J. Mol. Biol., 5:732-45.
  • the positions of the CDRs may be identified as the residues that make enthalpic contributions to antigen binding. See, e.g., Makabe et al, 2008, Journal of Biological Chemistry, 283: 1 156-1 166.
  • CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues do not significantly impact antigen binding.
  • a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches. The methods used herein may utilize CDRs defined according to any of these approaches. For any given embodiment containing more than one CDR, the CDRs may be defined in accordance with any of Kabat, Chothia, extended, AbM, contact, and/or conformational definitions.
  • the extended CDR refers to all of the amino acid residues identified by the Kabat and Chothia methods.
  • the terms“wild-type amino acid,”“wild-type IgG,”“wild-type antibody,” or“wild-type mAh,” refer to a sequence of amino or nucleic acids that occurs naturally within a certain population (e.g., human, mouse, rats, cell, etc.).
  • the term“substantially” refers to a qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest.
  • One of ordinary skill in the art will understand that electrical properties rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. Substantially is therefore used herein to capture a potential lack of completeness inherent therein. Values may differ in a range of values within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than). For example, values may differ by 5%.
  • the term“substantially free of’ when used to describe a material or compound, means that the material or compound lacks a significant or detectable amount of a designated substance.
  • the designated substance is present at a level not more than about 1%, 2%, 3%, 4% or 5% (w/w or v/v) of the material or compound.
  • a preparation of a particular stereoisomer is“substantially free of’ other stereoisomers if it contains less than about 20%, 15%, 10%, 5%, 3%, 2%, 1%, 0.5% (w/w or v/v) of the other stereoisomers other than the particular stereoisomer designated.
  • the term“substantially pure” means an object species is the predominant species present (i.e., on a molar basis it is more abundant than any other individual species in the composition), and in some embodiments, a substantially purified fraction is a composition wherein the object species (e.g., a glycoprotein, including an antibody or receptor) comprises at least about 50 percent (on a molar basis) of all macromolecular species present.
  • a substantially pure composition will comprise more than about 80 percent of all macromolecular species present in the composition, in some embodiments, more than about 85%, 90%, 95%, and 99%.
  • the object species is purified to essential homogeneity (contaminant species cannot be detected in the composition by conventional detection methods) wherein the composition consists essentially of a single macromolecular species.
  • a substantially pure material is at least 50% pure (i.e., free from contaminants), in some embodiments, at least 90% pure, in some embodiments, at least 95% pure, yet in some embodiments, at least 98% pure, and in some embodiments, at least 99% pure. These amounts are not meant to be limiting, and increments between the recited percentages are specifically envisioned as part of the disclosure.
  • the term“substituted” refers to a chemical group, such as alkyl, cycloalkyl aryl, and the like, wherein at least one hydrogen is replaced with a substituent as described herein, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, -CF 3 , -CN, or the like.
  • the term“substituted” is also contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, for example, those described herein above.
  • the permissible substituents may be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This disclosure is not intended to be limited in any manner by the permissible substituents of organic compounds.
  • any single substituent has fewer than the 100 total atoms. In many embodiments, however, any single substituent has fewer than the 10 total atoms. It will be understood that“substitution” or“substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, or other reaction.
  • Traditional methods for introducing gas and/or liquid samples that are present in high concentration, at high sample levels, or from samples having a high vapor pressure are highly efficient. It can be relatively easy to ionize such samples at atmospheric pressure (e.g., by chemical ionization, electrospray) thereby creating ions of analytes of interests, as well as interfering/contaminating ions and neutral molecules, in high abundance.
  • These traditional sample preparation and introduction methods are however less suitable for the unique sampling circumstances presented for biological fluid and tissue samples as contemplated by the present disclosure.
  • Challenges relate to the capability to target a particular analyte of a group of analytes, while limiting or eliminating interference. Moreover, it can be challenging to do so with sensitivity, simplicity, selectivity, speed, and throughput.
  • the present disclosure encompasses a recognition that in mass spectrometry systems, it is desirable to introduce biological samples, for example, a sample that is present in a bodily fluid or tissue where that sample is present in very low levels.
  • the present disclosure encompasses a recognition that there is a need for a discrete, simple, apparatus and a workflow to directly immobilize polypeptides, such as anti-bodies to sample extraction devices for capturing biological analytes for analysis.
  • methods disclosed herein include sampling biologies by a mass spectrometry system, including functionalizing at least a portion of an outer surface of an elongate sampling element with at least one polypeptide, wherein the at least one polypeptide is characterized in that it can preferentially bind to at least one analyte, where the elongate sampling element can include an outer surface that extends from a first end to a second end that terminates at a distal surface, where the second end could be configured to be inserted within a sampling interface of the mass spectrometry system.
  • teachings of the present disclosure can provide enhanced sensitivity through a more uniform sample extraction area.
  • teachings of the present disclosure can generally provide enhanced surface area relative to known sampling devices, which can increase the amount of extracted analyte for analysis.
  • enhancement to uniformity, surface area, and extraction efficiency can correspond with increased sensitivity.
  • sensitivity can be improved from sub-ng/mL of a solid-phase microextraction fiber to single-digit pg/mL.
  • apparatus, systems and methods of the present disclosure can sample biologic analytes at what is typically a low-level without introducing isomeric interference, that is with both sensitivity and specificity.
  • implementations of the present disclosure are useful in the preparation of biologic samples and/or the preparation of apparatus for the introduction of biologic samples to mass spectrometry systems.
  • sample preparation techniques for mass spectrometry systems of the present disclosure in various aspects can be fast, reliable, reproducible, inexpensive, and amenable to automation.
  • the present disclosure provides methods for sampling in a mass spectrometry system including steps of functionalizing at least a portion of an outer surface of an elongate sampling element with at least one polypeptide.
  • the at least one polypeptide is characterized in that it preferentially binds to at least one analyte.
  • the step of functionalizing includes steps of coating the outer surface of the elongate sampling element and immobilizing the at least one polypeptide on the coated surface.
  • methods further include a step of exposing at least a portion of the polypeptide-functionalized elongate sampling element to a bodily fluid sample, so that the analyte, if present in the bodily fluid sample, binds the polypeptide.
  • methods further include a step of inserting at least a portion of the elongate sampling element into a sampling interface of the mass spectrometry system such that an extraction solvent flowing through the sampling interface extracts at least a portion of the polypeptide-bound analyte and transmits the extracted analyte downstream to an ion source and onto a mass analyzer of the mass spectrometry system.
  • methods further include a step of performing a mass spectrometric analysis on the extracted analyte.
  • the present disclosure provides apparatus, for example, an elongate sampling element, which is at least partially coated with a polypeptide.
  • the polypeptide-coated surface of the elongate sampling element is configured such that it will bind to an analyte when they come in contact with one another. That is, in some embodiments, the at least one polypeptide is characterized in that it preferentially binds to the at least one analyte.
  • the present disclosure further encompasses a recognition that workflows to directly immobilize antigen or fragment thereof to sample extraction devices for capturing biological analytes, specifically, antibody or fragment thereof.
  • the outer surface of the elongate sampling element can be functionalized, with at least one antigen or fragment thereof, where the antigen or fragment thereof preferentially binds to an analyte of interest.
  • At least one analyte can be contained in a biologic sample.
  • the at least one analye can include, without limitation, an antibody or fragment thereof, a ligand, a nucleic acid, a peptide, a polypeptide, or a protein.
  • mass spectrometry systems and mass spectrometry-based analytical systems and methods are provided herein.
  • an extraction solvent can be utilized for introducing at least one analyte to the mass spectrometry system.
  • the extraction solvent can contact an elongate sampling element having at least one analyte bound to its surface.
  • the extraction solvent desorbs and/or extracts the at least one analyte from the surface of the elongate sampling element.
  • the extraction solvent can be configured to transmit and/or carry the at least one analyte downstream to an ion source for subsequent ionization and ultimately detection via the mass spectrometry system.
  • the present disclosure provides a sampling interface and ion source with both sensitivity and specificity as provided herein but without having a need for a liquid chromatography column located between the sampling interface and the ion source.
  • the mass spectrometry system 100 includes a substrate sampling interface 130 (e.g., an open port probe).
  • the substrate sampling interface 130 can be in fluid communication with an ion source 140.
  • the ion source 140 can be used for discharging a liquid containing at least one analyte into an ionization chamber 120.
  • the ionization chamber 120 is in fluid communication with a mass analyzer 160 positioned downstream from the ionization chamber 120.
  • the mass analyzer 160 can detect and/or process ions generated by the ion source 140.
  • the substrate sampling interface 130 can be configured to receive at least a portion of an elongate sampling element 125 (e.g., solid-phase microextraction substrate).
  • the elongate sampling element can include an outer surface extending from a first end 124 to a second end 126 that terminates at a distal surface (not shown), wherein the second end 126 is configured to be inserted within the substrate sampling interface 130.
  • the elongate sampling element 125 is placed in a fluid pathway of the substrate sampling interface 130 extending between an extraction solvent source 131 and the ion source probe (e.g., electrospray electrode) 144.
  • analytes desorbed from the outer surface of the elongate sampling element 125 by the extraction solvent flow directly into the ion source 140 within the extraction solvent for ionization. That is, the surface of the elongate sampling element 125 can be brought into contact with an extraction solvent, which can extract at least one analyte from the sampling element and carry the extracted analyte to the ion source 140 and the mass analyzer 160.
  • the extraction solvents can be or include any of the following methanol, ethanol, isopropanol, acetonitrile, acetone, chloroform, dichloromethane, water, or combinations thereof.
  • the ion source 140 can have a variety of configurations.
  • the ion source 140 can be configured to ionize one analyte contained within a liquid (e.g., the extraction solvent) that is received, for example, from the substrate sampling interface 130.
  • a liquid e.g., the extraction solvent
  • an electrospray electrode 144 which can comprise a capillary fluidly coupled to the elongate sampling element 125, terminates in an outlet end that at least partially extends into the ionization chamber 120 and discharges the extraction solvent therein.
  • the outlet end of the electrospray electrode 144 can atomize, aerosolize, nebulize, or otherwise discharge (e.g., spray with a nozzle) the extraction solvent into the ionization chamber 120 to form a sample plume 150.
  • the sample plume 150 includes micro-droplets that can be generally directed toward the curtain plate aperture 1 l4b and the vacuum chamber sampling orifice 116b.
  • the at least one analyte that was extracted and contained within the micro-droplets can be ionized (i.e., charged) by the ion source 140.
  • the at least one analyte is ionized as the sample plume 150 is generated.
  • the outlet end of the electrospray electrode 144 can be formed from and/or fabricated out of a conductive material and electrically coupled to a pole of a voltage source (not shown), while the other pole of the voltage source can be grounded.
  • micro droplets contained within the sample plume 150 can be charged by the voltage applied to the outlet end of the electrospray electrode 144 such that as the extraction solvent within the droplets evaporates during desolvation in the ionization chamber 120, bare charged analyte ions are released and drawn toward and through the apertures 1 l4b, 116b and focused (e.g., via one or more ion lens) into the mass analyzer 160.
  • the ion source probe can be an electrospray electrode 144.
  • the ion source 140 can be an electrospray ionization device, a nebulizer assisted electrospray device, a chemical ionization device, a nebulizer assisted atomization device, a photoionization device, a laser ionization device, a thermospray ionization device, or a sonic spray ionization device.
  • the ionization chamber 120 can be evacuated to a pressure lower than atmospheric pressure. In some embodiments, the ionization chamber 120 can be maintained at an atmospheric pressure. In some embodiments, the ionization chamber 120 can be separated from a gas curtain chamber 114 by a plate 1 l4a having a curtain plate aperture 1 l4b. As shown in FIG. 1, a vacuum chamber 116, which can house the mass analyzer 160, can be separated from the curtain chamber 114 by a plate 1 l6a having a vacuum chamber sampling orifice 116b.
  • the curtain chamber 114 and vacuum chamber 116 can be maintained at a selected pressure(s) (e.g., the same or different sub-atmospheric pressures, a pressure lower than the ionization chamber) by evacuation through one or more vacuum pump ports 118.
  • the mass spectrometry system 100 can include a source of pressurized gas 170 (e.g. nitrogen, air, or noble gas).
  • the source of pressurized gas 170 supplies a high velocity nebulizing gas flow which surrounds the outlet end of the electrospray electrode 144 and interacts with the fluid discharged therefrom to enhance the formation of the sample plume 150.
  • the mass analyzer 160 can assume one of a variety of configurations.
  • the mass analyzer 160 can be configured to process (e.g., filter, sort, dissociate, detect, etc.) sample ions generated by the ion source 140.
  • the mass analyzer 160 can be a triple quadrupole mass spectrometry system.
  • the mass analyzer 160 can be any mass analyzer known in the art or modified in accordance with the teachings herein.
  • any number of additional elements can be included in the mass spectrometry system including, for example, an ion mobility spectrometer (e.g., a differential mobility spectrometer) that is configured to separate ions based on their mobility through a drift gas rather than their mass-to-charge ratio.
  • the mass analyzer 160 can include a detector that can detect ions which pass through the mass analyzer 160 and can, for example, supply a signal indicative of the number of ions per second that are detected.
  • the present disclosure provides an elongate sampling element having a surface configured to be functionalized such that at least one analyte in a sample, for example, a bodily fluid sample preferentially bind to such an elongate sampling element.
  • the present disclosure provides an elongate sampling element having at least one polypeptide immobilized to its surface, where the at least one polypeptide is characterized in that it preferentially binds to at least one analyte in a sample, for example, in a bodily fluid sample.
  • an elongate sampling element can be configured for use with a substrate sampling interface, for example, an open port probe of a mass spectrometry system.
  • the elongate sampling element can have an outer surface that extends from a first end (124 of FIG. 1) to a second end (126 of FIG. 1).
  • the first end (124 of FIG. 1) of the elongate sampling element can be useful as a handle of machine manipulation.
  • the elongate sampling element which terminates at a distal surface and can be coated with a polypeptide- bound analyte, can be inserted within the substrate sampling interface of the mass spectrometry system, so as to extract the analyte for introduction into a downstream ion source.
  • the elongate sampling element can be formed from or can include, for example, alumina silicate, antimony silicate, arsenic silicate, barium silicate, bismuth silicate, boron silicate, cadmium silicate, gallium silicate, germanium silicate, glass, gold silicate, lead silicate, lime silicate, lithium silicate, magnesium silicate, nickel, nitrogen silicate, platinum silicate, silica, sodium silicate, phosphorus silicate, potassium silicate, tin silicate, indium silicate, silver silicate, zinc silicate, or any combination thereof.
  • the elongate sampling element can be formed from or can be fabricated from a metal.
  • the elongate sampling element can be formed from or can be fabricated from an elemental metal or an alloy.
  • an elongate sampling element is a unified structure.
  • an elongate sampling element can include at least two layers.
  • the distal surface of the elongate sampling element can be a solid surface such as, for example and without limitation, a carbon surface, a quartz surface, a glass surface, a gold surface, a silver surface, a copper surface, an iron oxide surface, an alloy surface, a composite surface, a polymer surface, or any combination thereof.
  • a surface can be independently porous, non-porous, or combination thereof.
  • the elongate sampling element can be formed from or can be fabricated from an organic material, an inorganic material, a biological material, or any combinations thereof.
  • the surface can have one or more layers comprising proteins such as whole proteins, fractional proteins, natural proteins, synthetic protein, functionalized protein, or any combinations thereof.
  • the proteins can be from animal, plant, microbes, or any combination thereof.
  • the elongate sampling element can be formed from or can be fabricated from a material having magnetic properties and/or exhibit properties, for example, including magnetic properties.
  • the elongate sampling element can be heated, cooled, and/or have a field applied thereto.
  • the elongate sampling element has a coating on its surface such that the coating material of the elongate sampling element forms the outer surface of the elongate sampling element.
  • the coating of the elongate sampling element can be formed from or can be fabricated from alumina silicate, antimony silicate, arsenic silicate, barium silicate, bismuth silicate, boron silicate, cadmium silicate, gallium silicate, germanium silicate, glass, gold silicate, lead silicate, lime silicate, lithium silicate, magnesium silicate, nickel, nitrogen silicate, platinum silicate, silica, sodium silicate, phosphorus silicate, potassium silicate, tin silicate, indium silicate, silver silicate, zinc silicate, or any combination thereof
  • the elongate sampling elements can have a length of about 1 cm, 2 cm, 3 cm, 4 cm, 5 cm, 6 cm, or more. In some embodiments, the elongate sampling element has a surface that is roughened to increase its surface area for more effective
  • FIG. 2 in a schematic diagram shows an exemplary elongate sampling elements 200.
  • FIG. 2 at panel (A) shows a first end 210 and a second end 220 that terminates at a distal surface.
  • FIG. 2 at panel (B) shows a first end 230 and a second end 240 that terminates at a distal surface having a roughened end.
  • the functionalized distal surface of the second end (126 of FIG. 1) of the elongate sampling element can have a variety of shapes.
  • an increase in the surface area of the distal surface of the second end (126 of FIG. 1) of the elongate sampling element can increase the amount of sample exposed to and thereby desorbed by the extraction solvent.
  • the distal surface of the second end (126 of FIG. 1) of the elongate sampling element can have a cross-sectional shape, for example, a square, a diamond, a star having 5 points, a star having 6 points, a star having 7 points, a star having 8 points, a star having 9 points, or a star having 10 points.
  • the distal surface of the second end (126 of FIG. 1) of the elongate sampling element can have a variety shapes that protrude and/or project from a center of the elongate sampling element to at least a portion of the outer surface of the second end (126 of FIG. 1) of the elongate sampling element. In some embodiments, these protruding or projecting shapes can be angled, bent, turned, twisted, etc. [0095] In some embodiments, for example, with reference to FIG. 3, the elongate sample elements as shown are fabricated from silica. In this embodiment, the distal surface shows a star pattern. In this embodiments, with each half turn or full turn, the surface area of the second end (126 of FIG. 1) increases. With reference to FIG.
  • 310 shows a second end with a distal surface having no turns
  • 320 shows a second end with a distal surface having 1/2 turn
  • 330 shows a second end with a distal surface having one turn
  • 340 shows a second end with a distal surface having two turns
  • 350 shows a second end with a distal surface having three turns
  • 360 shows a second end with a distal surface having four turns.
  • the second end (126 of FIG. 1) is inserted into the substrate sampling interface such that an increase in the surface area of the distal surface of the second end (126 of FIG. 1) of the elongate sampling element can increase the amount of sample exposed to and thereby desorbed by the extraction solvent.
  • FIG. 4 An exemplary shape of a distal surface 320 of a second end (126 of FIG. 1) of an elongate sampling element 410 is shown in FIG. 4.
  • the 7-point star 420 includes a coated surface area that provides an increased surface area relative to known substrates, thereby increasing the binding capacity of the surface.
  • the seven points extend radially-outward from the minimum diameter of the outer surface 420.
  • outer and inner surfaces can include variations between the maximum and minimum cross-sectional diameters about the perimeter of the cross-sectional shapes such that each surface includes a plurality of protrusions so as to further increase the surface area that can be exposed to an extraction solvent.
  • the elongate sampling element 510 has an outer surface 520 and an inner surface 530.
  • a coating can be bound to an outer surface of an elongate sampling element.
  • the coated elongate sampling element can include one or more moieties that can couple to a surface of the sampling element, for example, chemically, physically, or via combination thereof to bind to at least one antibody or fragment thereof.
  • such moieties include, for example and without limitation, amine groups (e.g., amine groups from lysine residues and the N-terminus of each polypeptide chain), thiol/sulfhydryl groups (e.g., thiol/sulfhydryl groups on cysteine residues and the ones from disulfide bonds that stabilize the antibody or fragment thereof molecular structure), and carbohydrate (sugar) groups (e.g., carbohydrate (sugar) groups from Fc regions of Ab).
  • amine groups e.g., amine groups from lysine residues and the N-terminus of each polypeptide chain
  • thiol/sulfhydryl groups e.g., thiol/sulfhydryl groups on cysteine residues and the ones from disulfide bonds that stabilize the antibody or fragment thereof molecular structure
  • carbohydrate (sugar) groups e.g., carbohydrate (sugar) groups from Fc regions
  • the surface of elongate sampling elements can contain silica or the like.
  • the silica can contain hydroxyl groups.
  • the hydroxyl groups can be formed on a silica surface.
  • hydroxyl groups on silica surface can be formed by various different methods. For example, hydroxyl groups on silica surface can be formed by reacting a silica surface with a solution containing one or more oxidant acids, such as a solution containing sulfuric acid, nitric acid, hydrogen peroxide, or any combinations thereof.
  • the presence of hydroxyl groups on the silica surface provides sites for the attachment of different groups, such as a silane group.
  • a heterobifunctional or homobifunctional cross-linker with different reactive groups on each end can be coupled to the silane on one end, while the other free end can form an amide bond with the terminal amino groups of an antibody or fragment thereof and thus immobilize the antibody or fragment thereof to the surface.
  • the surface of elongate sampling elements can be modified using one or more amine-terminal silanes, such as aminopropyltriethoxysilane (APTES), 3- isocyanatopropyl triethoxysilane, and 3-aminopropyltrimethoxysilane (APTMS); one or more thiol-terminal silanes, such as 3-mercaptopropyltrimethoxysilane (MPTMS) and
  • MDS mercaptomethyldimethylethoxysilane
  • a cross linker with two active groups can also be used to covalently immobilize antibody or fragment thereof by conjugation between a silane layer on a surface and primary amines in the antibody or fragment thereof.
  • the cross-linker can be, for example and without limitation, glutaraldehyde (GA), A-succinimidyl-4-maleimidobutyrate (GMBS), or N- succinimidyl-4-(/V-maleimido-methyl)-cyclohexane- 1 -carboxylate.
  • a polypeptide for example, an antibody
  • the surface of the elongate sampling element has enough active groups and/or moieties such that the polypeptide can be attached.
  • the outer surface of the elongate sampling element may be activated and/or may need to be activated before a polypeptide can be bound thereto.
  • the surface of the elongate sampling element does not have enough active groups and/or moieties so that the antibody or fragment thereof cannot be attached to the surface.
  • the solid surface when the surface does not have enough active groups for an antibody or fragment thereof to attach, can be activated or functionalized with active groups that can react with one or more moieties from the antibody or fragment thereof (e.g., the Fab region of the antibody or fragment thereof, the Fc region of the antibody or fragment thereof, etc.) to facilitate immobilization.
  • the functionalization of the surface to facilitate the immobilization of antibody or fragment thereof can be performed in a number of different ways, such as, for example and without limitation, by physical adsorption to the surface of at least one active group, covalent attachment to the surface of at least one functional group, non-covalent attachment to the surface of at least one functional group, or any combinations thereof.
  • an antibody or fragment thereof can be immobilized to a coated elongate sampling element.
  • the present disclosure provides elongate sampling elements having an antibody immobilized on an outer surface thereof.
  • an antibody or fragment thereof is immobilized on the outer surface of the elongate sampling element through covalent binding, non-covalent binding, physical binding, or any combinations thereof.
  • antibody immobilization can occur for example via an amine, amide, or amino bond at the outer surface of the elongate sampling element.
  • antibody immobilization can occur via a cross-linking reaction at the outer surface of the elongate sampling element.
  • an antibody or fragment thereof can be immobilized to the outer surface of the elongate sampling element, for example, via an amine/glutaraldehyde moiety.
  • reactive ends of an exposed amine group on the outer surface of the elongate sample element can react with an amino group from one or more amino acids residues.
  • the amino group is located, for example, on an antibody or fragment thereof, such as a lysine residue.
  • such a bond can be utilized as an anchoring point to immobilize the antibody or fragment thereof on the outer surface of the elongate sampling element.
  • the exposed amine group could be from a reactive end, for example, from a glutaraldehyde molecule that was reacted with and/or coated to the outer surface of the elongate sampling element.
  • an antibody or fragment thereof can be immobilized to the outer surface of the elongate sampling element, for example, via an amine/N- hydroxysuccinimide moiety.
  • N-hydroxysuccinimide esters react with the amine.
  • the antibody or fragment thereof can be immobilized by a labile Schiff s base formation between its one or more amine group and the aldehyde group of N- hydroxysuccinimide.
  • an N-hydroxysuccinimide or sulfa-N-hydroxysuccinimide ester can be introduced for antibody immobilization.
  • N-hydroxysuccinimide ester reacts with the amine on the protein and yields a stable amide bond while releasing the NHS leaving groups.
  • an antibody or fragment thereof can be immobilized on the outer surface of the elongate sampling element, for example, via an amine/Maleimide moiety.
  • the Maleimide moiety and the imide functional group is readily available for crosslinking of sulfhydryl groups of cysteine residues, or sulfhydrylized antibodies.
  • a thiol group from one or more amino acid residues located on an antibody or fragment thereof e.g., a cysteine residue
  • a thiol group from the one or more amino acid residues is located on the exterior of the antibody or fragment thereof to facilitate binding with the surface.
  • the thiol group from the antibody or fragment thereof can be covalently attached to a polymerizable lipid with a terminal linker group.
  • the antibody or fragment thereof can be thiolated using 2- iminothiolane hydrochloride and attached to a gold surface array by means of thiol-Au linkage.
  • an antibody or fragment thereof can be immobilized on the outer surface of the elongate sampling element, for example, via one or more sugar residues located on an antibody or fragment thereof.
  • the one or more sugar residues are located on the exterior of the antibody or fragment thereof to facilitate binding with the surface.
  • an antibody or fragment thereof can be immobilized on the outer surface of an elongate sampling device having been previously modified by an aminosilane, such as (3-aminopropyl)triethoxysilane, (APTES) by reactions between amines in the saline and aldehydes, which can be produced by the sodium peroxidase oxidation of sugar residues at the C- terminal of the antibody or fragment thereof.
  • an aminosilane such as (3-aminopropyl)triethoxysilane, (APTES)
  • an antibody or fragment thereof can be immobilized on the outer surface of the elongate sampling element, for example, through pretreatment of the outer surface with plasma.
  • the outer surface can be treated with a microwave- induced FbO/Ar plasma to obtain silicon hydroxyls and active available bond sites.
  • these offer bonding sites for surface modifiers, such as (3- aminopropyl)triethoxysilane, (APTES) and thus can increase the density of cross-linkers, such as glutaraldehyde, that can couple with antibody or fragment thereof.
  • APTES (3- aminopropyl)triethoxysilane
  • cross-linkers such as glutaraldehyde
  • antibody or fragment thereof can be coupled to a plasma-treated polymer, such as poly methyl methacrylate (PMMA).
  • PMMA poly methyl methacrylate
  • oxygen plasma pretreatment of a polymer deposited on a surface yields surface polar groups on the polymer that can be used to immobilize the antibody or fragment thereof.
  • a polyethyleneimine (PEI) layer can be deposited onto oxygen plasma-activated PMMA foil and further cross-linked with GA to form an amine-reactive aldehyde surface (PEI-GA).
  • the antibody or fragment thereof can be deposited on the PEI-GA surface using different techniques, such as overprinting.
  • functional groups can be introduced onto the surface by plasma pretreatment to facilitate the immobilization of antibody or fragment thereof. This process can depend on the plasma parameters such as the power, used gases, treatment time, and pressure.
  • amino groups can be introduced onto a surface by using Ar/0 2 plasma treatment and ammonia plasma treatment, respectively.
  • an antibody or fragment thereof can be immobilized on the outer surface of the elongate sampling element, for example, via intermolecular forces such as ionic interactions, hydrophobic interactions, hydrophilic interactions, polar interactions, van der Waal interactions, and electrostatic interactions can be used to immobilize antibody or fragment thereof onto a surface.
  • the outer surface of the elongate sampling element can include avidin and/or other biotin-binding proteins, which can biotinylated antibody binding to the surface.
  • biotinylated antibody or fragment thereof, antibody or fragment thereof labeled with biotin i.e., also known as vitamin H, vitamin B7, or coenzyme R
  • avidin and other biotin-binding proteins including streptavidin, neutravidin, tamavidin, and captavidin to generate a biocompatible layer on a surface.
  • Biotin comprises an ureido unit that binds avidin and a thiophene unit with a carboxyl group at the tip of a valeric acid side chain wherein the carboxyl group can be derivatized to conjugate antibody or fragment thereof.
  • the surface needs to be activated before attaching biotin or biotinylated molecules.
  • the silanized glass can be treated with acrylamide or 4-aminophenylmercuric acetate to produce free amino groups that react with NHS biotin.
  • the antibody or fragment thereof can be immobilized on glass including a biotinylated polyethylene glycol layer, streptavidin layer, and a protein L-biotin layer.
  • the biotin or biotinylated molecules can be attached to the surface by APTES to generate free amine terminals that covalently bind to the NHS -ester of biotin by an azide group.
  • the avidin can be used to form an avidin-biotin complex.
  • the two biotin-binding sites of avidin face the surface of a coupled biotinylated antibody.
  • functional groups on the immobilized antibody or fragment thereof can be added, activated, and/or tailored for specific interactions with at least one analyte using chemical treatment(s) and/or physical treatment(s), which can lead to the transformation of the surface of the antibody or fragment thereof into a more reactive form.
  • the binding can be done, for example and without limitation, by the accessible functional groups of exposed amino acids, which can lead to the reversible or non-reversible binding of the antibody or fragment thereof with the outer surface of the elongate sampling element and which can give different degrees of surface coverage.
  • the degree of surface coverage from the antibody or fragment thereof can be about 1% of the outer surface of the elongate sampling element, about 5% of the outer surface of the elongate sampling element, about 10% of the outer surface of the elongate sampling element, about 20% of the outer surface of the elongate sampling element, about 30% of the outer surface of the elongate sampling element, about 40% of the outer surface of the elongate sampling element, about 50% of the outer surface of the elongate sampling element, about 60% of the outer surface of the elongate sampling element, about 70% of the outer surface of the elongate sampling element, about 80% of the outer surface of the elongate sampling element, about 90% of the outer surface of the elongate sampling element, or about 100% of the outer surface of the elongate sampling element.
  • an antibody-bound surface can be configured to preferentially and/or specifically bind to an analyte.
  • an analyte preferentially (and/or selectively) binds to and/or is preferentially (and/or selectively) bound to the antibody or fragment thereof.
  • the analyte is from a bodily fluid.
  • the bodily fluid can be any of blood, blood product, saliva, vomit, urine, tear, sweat, bile, milk, cerebrospinal fluid, feces, body secretion, pus, mucus, lymph, gastric acid juice, earwax, blister, humoral fluid, intracellular fluid, extracellular fluid, human fluid, animal fluid, plant fluid, rinsate of solid, rinsate of surface, fluid extract, or any combination thereof.
  • methods of mixing and/or exposing an elongate sampling element that is functionalized with an immobilized antibody on the outer surface thereof to an analyte are disclosed.
  • the analyte is present in a bodily fluid sample. It is noted that the analyte is (or it is presumed that it its) present in a concentration substantial and sufficient for binding, extraction, detection, etc.
  • method steps can include providing a mixing kit 610 that includes: a biological sample vial 660 including a biological sample 650 and an elongate sampling element 620.
  • methods can include inserting a second end 640 of an elongate sampling element 620 into the biological sample 650.
  • the biological sample could be blood, blood product, saliva, vomit, urine, tear, sweat, bile, milk, cerebrospinal fluid, feces, body secretion, pus, mucus, lymph, gastric acid juice, earwax, blister, humoral fluid, intracellular fluid, extracellular fluid, human fluid, animal fluid, plant fluid, rinsate of solid, rinsate of surface, fluid extract, and any combination thereof.
  • methods further include a step of mixing with the handle and/or mechanical mixer 630.
  • At least a portion of the outer surface of the elongate sampling element can be functionalized with a polypeptide and then bound to a sample.
  • methods of the present disclosure can further include steps of performing a mass spectrometric analysis of that sample.
  • the present method can include steps of delivering the sample to the mass spectrometry system.
  • method steps include inserting at least a portion of the elongate sampling element into a substrate sampling interface of the mass spectrometry system.
  • methods can further include a step of flowing the extraction solvent over the second end of the elongate sampling element, such that it can contact at least a portion of the bound analyte and carries the extracted analyte or at least a portion thereof to an ion source of the mass spectrometry system.
  • methods can include a step of inserting at least a portion of the analye bound elongate sampling element into a substrate sampling interface of the mass spectrometry system, whereby an extraction solvent can contact at least a portion of the analyte, such that the extraction solvent can extract and carry the extracted analyte to an ion source of the mass spectrometry system.
  • methods can further include a step of performing a mass spectrometric analysis on the extracted analyte.
  • FIG. 8 shows such an apparatus and/or system for extracting analyte from an elongate sampling element and carrying the extracted analyte for performing a mass
  • an exemplary substrate sampling interface 810 (e.g., an open port probe) for extracting at least one analyte from the elongate sampling element 820 and suitable for use in the system of FIG. 1 is schematically depicted.
  • the elongate sampling element 820 is shown with a first end 812 and a second end 814 having a distal surface.
  • the substrate sampling interface 810 includes an outer tube 870 (e.g., outer capillary tube). In some embodiments, the outer tube 870 extends from a proximal end 870p to a distal end 870d.
  • an inner tube 840 (e.g., inner capillary tube) is disposed co-axially within the outer capillary tube. As shown, the inner capillary tube 840 also extends from a proximal end 840p to a distal end 840d.
  • the inner capillary tube 840 comprises an axial bore providing a fluid channel therethrough and defines a sampling conduit 850 having a distal end 850d through which liquid can be transmitted from the substrate sampling probe 860 to the ion source (140 of FIG. 1 (i.e., the sampling conduit 850 is fluidly coupled to inner bore of the electrospray electrode 144 of FIG. 1)).
  • the annular space between the inner surface of the outer capillary tube 870 and the outer surface of the inner capillary tube 840 can define an extraction solvent conduit 890 having a distal end 890d extending from an inlet end coupled to the extraction solvent source 860 (e.g., via conduit 865) to an outlet end (adjacent the distal end 840d of the inner capillary tube 840).
  • the proximal end 840p of the inner capillary tube 840 can be recessed relative to the proximal end 870p of the outer capillary tube 870 (e.g., by a distance h) so as to define a proximal fluid chamber 835 of the substrate sampling interface 810 that extends between and is defined by the proximal end 840p of the inner capillary 840 and the proximal end 870p of the outer capillary tube 870.
  • the proximal fluid chamber 835 represents the space adapted to contain fluid between the open proximal end of the substrate sampling interface 810 and the proximal end 840p of the inner capillary tube 840.
  • the extraction solvent conduit 890 is in fluid communication with the sampling capillary 850 via this proximal fluid chamber 835.
  • fluid that is delivered to the proximal fluid chamber 835 through the extraction solvent conduit 890 can enter the inlet end of the sampling conduit 850 for transmission to its outlet end and subsequently to the ion source.
  • the conduit defined by the inner capillary tube 840 can instead be coupled to the extraction solvent source 860 (so as to define the extraction solvent conduit) and the annular space defined between the inner and outer capillaries 840, 870 can be coupled to the ion source so as to define the sampling conduit.
  • methods using the apparatus illustrated in FIG. 8 includes a step of fluidly coupling the extraction solvent source 860 via the supply conduit 865 through which extraction solvent can be delivered at a selected volumetric rate (e.g., via one or more pumping mechanisms including reciprocating pumps, positive displacement pumps such as rotary, gear, plunger, piston, peristaltic, diaphragm pump, and other pumps such as gravity, impulse and centrifugal pumps can be used to pump liquid sample), all by way of non-limiting example.
  • Any extraction solvent effective to extract analytes from the elongate sampling element and amenable to the ionization process are suitable for use in the present teachings.
  • one or more pumping mechanisms can be provided for controlling the volumetric flow rate through the sampling conduit 850 and/or the electrospray electrode (not shown), these volumetric flow rates selected to be the same or different from one another and the volumetric flow rate of the extraction solvent through the extraction solvent conduit 890.
  • controlling these different volumetric flow rates through the various channels of the substrate sampling interface 810 and/or the electrospray electrode 144 (as shown in FIG. 1) can be by adjusting the flow rate so as to control the movement of fluid throughout the system.
  • steps can include inserting the analyte-bound coated elongate sampling element 825 through the open end of the substrate sampling interface 810 such that at least some of the analyte coated on the outer surface of the elongate sampling element is extracted and/or adsorbed by the extraction solvent (e.g., the extraction solvent within the proximal fluid chamber 835). That is, when the coated surface of the sampling element 825 is inserted into the proximal fluid chamber 835, the step of flowing extraction solvent can be effective to desorb at least a portion of the at least one analyte adsorbed on the coated surface such that any extracted analytes flow with the extraction solvent into the inlet of the sampling conduit 850.
  • the extraction solvent e.g., the extraction solvent within the proximal fluid chamber 835
  • methods disclosed herein further include a step of performing a mass spectrometric analysis on the extracted analyte.
  • sampling methods according to the present teachings can exhibit a sensitivity sufficient to detect a biologic, for example, testosterone, from serum/plasma, at a concentration as low as 0.1 pg/mL.
  • an antigen-bound surface can be configured to
  • analyte preferentially and/or specifically bind to an analyte, for example preferentially and/or specifically bind to an antibody or fragment thereof.
  • the analyte is from the bodily fluid.
  • the present example discloses methods of functionalizing an outer surface of an elongate sampling element.
  • FIG. 9 A general workflow for functionalizing an outer surface of an elongate sampling element with an antibody or fragment thereof is shown in FIG. 9.
  • Method steps include providing an elongate sampling element.
  • the elongate sampling element as provided herein can be formed from and/or fabricated, for example, from glass.
  • the elongate sampling element has an outer surface extending from a first end to a second end.
  • the elongate sampling element terminates at a distal surface, wherein the second end is configured to be inserted within a substrate sampling interface of a mass spectrometry system.
  • Methods further include a step of applying an aminosilane reagent to at least a portion of the outer surface of the elongate sampling element.
  • the aminosilane reagent is (3-aminopropyl)triethoxysilane, (APTES), which can activate the glass surface with the amine.
  • APTES (3-aminopropyl)triethoxysilane
  • Methods further include a step of immobilizing at least one antibody or fragment thereof to the glutaraldehyde, thereby functionalizing the outer surface of the elongate sampling element with the at least one antibody or fragment thereof.
  • the elongate sampling element can be glass.
  • the (3- aminopropyl)triethoxysilane, (APTES) can be applied to at least a portion of the outer surface of the elongate sampling element which can activate the glass surface with the amine.
  • the amine present on the outer surface of the elongate sampling element can then be reacted with glutaraldehyde.
  • the first end of the glutaraldehyde can reacts with the amine on the activated glass.
  • a second end of the glutaraldehyde can react the amino groups of lysine of an antibody to immobilize the antibody to the glutaraldehyde.
  • Methods further include a step of introducing and immobilizing the antibody to the glutaraldehyde.
  • FIG. 10 illustrates a workflow in accordance with some embodiments in various aspect of the present disclosure.
  • the workflow generally follows the methods of example 1.
  • FIG. 10 at panel (A) shows a bare elongate sampling element.
  • FIG. 10 at panel (B) shows the elongate sampling element following an activating step, that is, following applying, for example, an aminosilane reagent to at least a portion of the outer surface of the elongate sampling element as disclosed herein to activate the glass surface with amine groups.
  • FIG. 10 at panel (C) shows the elongate sampling element following a step of reacting with glutaraldehyde.
  • FIG. 10 at panel (A) shows a bare elongate sampling element.
  • FIG. 10 at panel (B) shows the elongate sampling element following an activating step, that is, following applying, for example, an aminosilane reagent to at least a portion of the outer surface of the elongate sampling element as disclosed herein to activate the glass surface with
  • FIG. 10 at panel (D) shows the elongate sampling element following a step of immobilizing an antibody or fragment thereof to the activated surface of the elongate sampling element.
  • the antibody or fragment thereof preferentially and/or selectively binds to at least one analyte.
  • FIG. 10 at panel (E) shows the elongate sampling element following a step of exposing the functionalized sampling element to a sample, for example, a bodily sample so that an analyte of interest if present in the sample would bind to the antibody or fragment thereof that is immobilized on the outer surface of the elongate sampling element.
  • Example 2 [0135] The present example discloses methods in accordance with the present teachings.
  • the present example discloses another method of functionalizing an outer surface of an elongate sampling element with at least one antibody or fragment thereof.
  • the amine present on the outer surface of the elongate sampling element can react with N-hydroxysuccinimide (NHS) or sulfo-NHS.
  • NHS N-hydroxysuccinimide
  • the NHS ester can react with the amine on protein to yield a stable amide bond while releasing NHS leaving groups. After activating the glass surface with the aminosilane reagents, the NHS or sulfa-NHS ester can be introduced for antibody immobilization.
  • Methods further include a step of immobilizing at least one antibody or fragment thereof to the N-hydroxysuccinimide (NHS) or sulfo-NHS, thereby functionalizing the at least one antibody or fragment thereof to the at least the portion of the outer surface of the elongate sampling element.
  • Methods further include a step of introducing and immobilizing the antibody to the elongate sampling element.
  • the present example discloses methods in accordance with the present teachings.
  • the present example discloses another method of functionalizing an outer surface of an elongate sampling element with at least one antibody or fragment thereof.
  • the amine present on the outer surface of the elongate sampling element can be reacted with Maleimide. After activating the glass surface with aminosilane reagents, the elongate sampling element can be further maleimide-activated, which is then ready for crosslinking of sulfhydryl groups of cysteine residues, or sulfhydrylized antibodies (with Traut's reagents or SATA reagents).
  • Methods can further include a step of immobilizing at least one antibody or fragment thereof to the Maleimide, thereby functionalizing the at least one antibody or fragment thereof to at least a portion of the outer surface of the elongate sampling element. Methods further include a step of introducing and immobilizing the antibody to the surface of the elongate sampling element.
  • FIG. 11 illustrates the general workflow of the methods of example 3.
  • FIG. 11 at panel (A) shows a bare elongate sampling element.
  • FIG. 11 at panel (B) shows the elongate sampling element following an activating step, that is, following, for example, applying an aminosilane reagent to at least a portion of the outer surface of the elongate sampling element as disclosed herein to activate the glass surface with amine groups.
  • FIG. 11 at panel (C) shows the elongate sampling element following a step of reacting with Maleimide and immobilizing an antibody to the activated surface.
  • FIG. 11 at panel (D) shows the elongate sampling element following a step of mixing the elongate sampling element functionalized with an antibody or fragment thereof immobilized to the outer surface with a bodily sample having an analyte.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Analytical Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Other Investigation Or Analysis Of Materials By Electrical Means (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
EP19790858.5A 2018-10-18 2019-10-17 Funktionalisierung eines probennahmeelements zur verwendung mit einem massenspektrometriesystem Pending EP3867943A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862747480P 2018-10-18 2018-10-18
PCT/IB2019/058876 WO2020079647A1 (en) 2018-10-18 2019-10-17 Functionalizing a sampling element for use with a mass spectrometry system

Publications (1)

Publication Number Publication Date
EP3867943A1 true EP3867943A1 (de) 2021-08-25

Family

ID=68296604

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19790858.5A Pending EP3867943A1 (de) 2018-10-18 2019-10-17 Funktionalisierung eines probennahmeelements zur verwendung mit einem massenspektrometriesystem

Country Status (4)

Country Link
EP (1) EP3867943A1 (de)
JP (1) JP2022505162A (de)
CN (1) CN112868086A (de)
WO (1) WO2020079647A1 (de)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4154013A1 (de) * 2020-05-22 2023-03-29 DH Technologies Development Pte. Ltd. Festphasenaffinitätsauswahl durch massenspektrometrie
CN116615649A (zh) * 2020-12-22 2023-08-18 豪夫迈·罗氏有限公司 自动化临床诊断系统和方法
JP2024507179A (ja) * 2021-02-25 2024-02-16 ディーエイチ テクノロジーズ デベロップメント プライベート リミテッド サンプリングのためのシステムおよび方法

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US6207370B1 (en) * 1997-09-02 2001-03-27 Sequenom, Inc. Diagnostics based on mass spectrometric detection of translated target polypeptides
DE60045816D1 (de) * 1999-04-27 2011-05-19 Bio Rad Laboratories Probenhalter für ein gasphaseionenspektrometer
US20090026122A1 (en) * 2002-03-11 2009-01-29 Janusz Biocompatible solid-phase microextraction coatings and methods for their preparation
CA2988172C (en) * 2014-06-13 2018-10-02 Jp Scientific Limited A probe for extraction of molecules of interest from a sample
US10103015B2 (en) * 2016-04-29 2018-10-16 Dh Technologies Development Pte. Ltd. Sampling interface for mass spectrometry systems and methods
EP3526811A4 (de) * 2016-10-14 2020-06-17 DH Technologies Development Pte. Ltd. Verfahren und systeme zur erhöhung der empfindlichkeit von direktabtastungsschnittstellen für massenspektrometrische analyse

Also Published As

Publication number Publication date
WO2020079647A1 (en) 2020-04-23
JP2022505162A (ja) 2022-01-14
CN112868086A (zh) 2021-05-28

Similar Documents

Publication Publication Date Title
JP6997271B2 (ja) タンパク質が結合した作用物質化合物のインタクト質量の測定
WO2020079647A1 (en) Functionalizing a sampling element for use with a mass spectrometry system
Atmanene et al. Extending mass spectrometry contribution to therapeutic monoclonal antibody lead optimization: characterization of immune complexes using noncovalent ESI-MS
AU2010270163B2 (en) TLR3 binding agents
DK3023777T3 (en) Method for Preparation of Peptide Fragments, Kits for Preparation of Peptide Fragments for Use Therein, and Method of Analysis
CN112513640B (zh) 糖基化肽的检测和定量
CN113301974A (zh) 在线色谱法和电喷雾电离质谱仪
Liu et al. Development of a monoclonal antibody based-ELISA for the detection of chloramphenicol in shrimp, feed and milk samples and validation by LC-MS/MS coupled with immunoaffinity clean-up
JP2022518254A (ja) 合剤中の二量体の定量化及び同定
US20120308581A1 (en) Compositions and methods for detecting tlr3
JP2022519246A (ja) 抗体電荷不均一性についてのネイティブマイクロ流体ce-ms解析法
US20080187461A1 (en) Protein, method for immobilizing protein, structure, biosensor, nucleic acid, vector and kit for detecting target substance
CN115078569B (zh) 一种生物传感集成uplc-ms技术的止咳关键质量属性辨识方法
EP3269821A1 (de) Verfahren zur gewinnung eines peptidfragments aus einem antikörper mittels proteasezersetzungsreaktion mit beschränktem reaktionsfeld
TW202314247A (zh) 量化抗cd40抗體之方法
KR101993563B1 (ko) 당단백질의 당쇄를 조제하는 방법, 키트 및 장치
Atmanene et al. Noncovalent mass spectrometry for the characterization of antibody/antigen complexes
Ikegawa et al. Immunoaffinity extraction for liquid chromatographic determination of equilin and its metabolites in plasma
JP2023542858A (ja) 水素交換質量分析を使用した、結合部位の同定のための方法
JP2022548116A (ja) 抗体アイソタイピングおよび定量化のためのlc-ms方法
Engin Development of chromatographic and electrochemical methods for trastuzumab determination
WO2021258114A1 (en) Process for direct readout of immunoglobulins
Amundsen Use of non-specific and specific interactions in the analysis of testosterone and related compounds by capillary electromigration techniques
Stumbaum Development and bioanalytical application of affinity-mass spectrometry for identification and structural characterisation of protein-ligand interactions

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20210430

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230601