METHODS FOR CHARACTERIZING COMPOSITIONS COMPRISING PEANUT ANTIGENS

Information

  • Patent Application
  • 20220170942
  • Publication Number
    20220170942
  • Date Filed
    November 10, 2021
    2 years ago
  • Date Published
    June 02, 2022
    2 years ago
Abstract
Methods for determining an in vitro release profile of peanut allergens in a sample are provided. Methods for determining one or more signatures of peanut allergens in a sample are provided.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Nov. 10, 2021, is named 724086_SA9-180DVCON_ST25.txt and is 59,489 bytes in size.


FIELD OF THE INVENTION

The present invention relates to methods for characterizing e.g., determining release profiles, allergen signatures and the like, of therapeutic compositions comprising peanut allergens for use in treating, alleviating or otherwise reducing peanut allergy in a subject.


BACKGROUND

Peanut allergy develops when the body's immune system has an abnormal hypersensitivity response to one or more peanut allergens. Peanut allergy is one of the most common food allergies in both children and adults. It receives particular attention because it is relatively common, typically lifelong, and can cause severe allergic reactions. Peanut allergy is the leading cause of anaphylaxis and death due to food allergy. It can lead to significant burden on patients and their families. Peanut is a common food ingredient making strict avoidance difficult. Therefore, there is a relatively high rate of accidental peanut ingestion for those trying to avoid peanuts. For these reasons, peanut allergy has become an important public health issue.


Research is currently underway focused on the development of compositions for the treatment of peanut allergy. Methods are needed for determining in vitro peanut allergen release data of known and newly developed compositions, both for quality control and to predict in vivo release profiles.


SUMMARY OF THE INVENTION

The present invention is based in part on the discovery of highly sensitive methods to determine release profiles of allergens and/or to determine allergenic signatures of compositions comprising allergens. The methods described herein provide the sensitivity and accuracy needed to profile peanut allergens present in a sample, such as a protein extract, a therapeutic composition, or a dissolution or release medium and to allow measurement of low level, relevant allergens present in the sample. In exemplary embodiments, the methods include the step of detecting allergen digest products that exist in one or more isoforms of one or more allergens in the sample. The identification of allergen digest products found in multiple isoforms of a peanut allergen provides qualitative and quantitative information about a sample, which can be indicators of batch-to-batch consistency, and can provide a release profile of allergens stored on or in a substrate, such as, e.g., a nanoparticle.


In one aspect, the invention features a highly sensitive method for determining a signature of peanut allergens in a composition. The method includes the step of digesting peanut allergens present in a composition (e.g., medium or other sample) from the composition to generate allergen digest products, fragmenting the allergen digest products to generate peptide fragments, detecting and identifying the allergen digest products by mass spectrometry, and determining the signature of peanut allergens in the composition. In particular embodiments, the methods are used to determine a signature of one or more peanut allergens that are present in low concentrations in a composition. The signature includes the type and quantity (e.g., relative quantity) of allergens in the composition.


In certain embodiments, the composition is an aqueous medium, such as an aqueous pharmaceutical composition, an analytical sample, a dissolution medium or a release medium. In some embodiments, the total amount of peanut allergens in the composition is very low. For example, the amount of a particular peanut allergen (e.g., Ara h1, Ara h2, Ara h3 or Ara h6), may be less than about 2 μg/ml, less than about 1.5 μg/ml, less than about 1 μg/ml or less than about 0.5 μg/ml, e.g., about 0.2 μg/ml.


In some embodiments, the method further comprises the step of comparing a peanut allergen signature to a signature standard. The signature standard can be a peptide profile set by a regulatory authority, such as the FDA (Food and Drug Administration) or EMA (European Medicines Association), a peptide profile established by industry standards, or a profile set by expectations as determined by repeated experimentation. The signature standard can specify the types and relative amounts of peanut allergens that are expected to be found in a sample.


In certain exemplary embodiments, the allergen digest products are between about 4 amino acids and about 50 amino acids in length, between about 6 amino acids and about 30 amino acids in length, or between about 15 amino acids and about 20 amino acids in length, or are about 15, 16, 17, 18, 19 or 20 amino acids in length


In certain exemplary embodiments, the steps of fragmenting the allergen digest products and detecting the peptide fragments are performed by a method that includes tandem mass spectrometry, such as Liquid Chromatography-tandem Mass Spectroscopy (LC-MS-MS), nano tandem Mass Spectroscopy (nanoLC-MS-MS) or nano High Performance Liquid Chromatography-tandem Mass Spectroscopy (nanoHPLC-MS-MS).


In certain exemplary embodiments, the signature (e.g., peanut allergen signature and/or signature standard) comprises one or more Ara h1 allergen digest products having an amino acid sequence selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:177, SEQ ID NO:155, SEQ ID NO:93 and SEQ ID NO:40. In some embodiments, the signature comprises allergen digest products from Ara h1, Ara h2 and Ara h6. In other embodiments, the signature comprises allergen digest products from Ara h1, Ara h2, Ara h3 and Ara h6. In some embodiments, the signature includes one or more allergen digest products having amino acid sequences selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:177, SEQ ID NO:197, SEQ ID NO:198, SEQ ID NO:199, SEQ ID NO:200, SEQ ID NO:201, SEQ ID NO:236 and SEQ ID NO:237. In some embodiments, the signature includes one or more allergen digest products having amino acid sequences selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:197, SEQ ID NO:198, SEQ ID NO:199, SEQ ID NO:200, SEQ ID NO:201, SEQ ID NO:236 and SEQ ID NO:237.


In some embodiments, allergen digest products suitable for use in establishing a peanut allergen profile are present in more than one isoform of a peanut allergen, such as in 2, 3, 4, 5, 6, 7, 8, 10, 12, 14, 16 or more isoforms of a particular allergen (e.g., an Ara h protein or polypeptide). For example, an allergen digest product suitable for use in a peanut allergen signature profile can be present in one or any combination of in 2, 3, 4, 5, 6, 7, 8 or more of isoforms of Ara h1, in 2, 3, 4, 5, 6, 7, 8 or more of isoforms of Ara h2, in 2, 3, 4, 5, 6, 7, 8 or more of isoforms of Ara h3, and/or in 2, 3, 4, 5, 6, 7, 8 or more of isoforms of Ara h6. In some embodiments, an allergen digest product suitable for use in a peanut allergen signature profile is present in all isoforms of one or more peanut allergens.


In certain exemplary embodiments, peanut allergens are digested with one or more proteases. For example, peanut allergens may be digested with one or more proteases selected from the group consisting of trypsin, endoproteinase Lys-C and endoproteinase Arg-C.


In certain exemplary embodiments, a composition comprising peanut allergens further comprises an internal standard. For example, in some embodiments, the internal standard comprises one or more heavy isotopes, such as 13C and/or 15N. In certain embodiments, the internal standard is not a full-length allergen, but is instead a fragment of the peptide. The fragment is typically less than 50 amino acids long (such as, e.g., between about 4 and about 50 amino acids long, between about 6 and about 30 amino acids long, or between about 10 and about 20 amino acids long). In some embodiments, the fragment has the sequence of an expected or predicted allergen digest product. In one embodiment, the internal standard is comprised of multiple peanut allergen peptides, each labeled at the C-terminus with a heavy isotope. For example, an exemplary internal standard can comprise any combination of two or more isotope-labeled fragments of an Ara h1 peptide, two or more isotope-labeled fragments of an Ara h2 peptide, two or more isotope-labeled fragments of an Ara h3 peptide and two or more isotope-labeled fragments of an Ara h6 peptide. In one embodiment, a composition comprising peanut antigens is digested with trypsin, a standard mix comprising 13C and/or 15N-labeled peanut allergen peptides is added to the digested mix, and then the composition is assayed by fragmentation, e.g., by mass spectrometry, such as e.g., LC-MS, or LC-MS-MS.


In certain exemplary embodiments, a signature comprises allergen digest products that do not contain missed proteolytic cleavage sites.


In another aspect, the invention features a method for determining a release profile, e.g., an in vitro release profile, of a composition comprising one or more peanut allergens. In one embodiment, the method includes obtaining one or more samples from the composition at each of a plurality of time points, digesting the peanut allergens present in the one or more samples to generate allergen digest products, fragmenting the allergen digest products to generate peptide fragments, and detecting the peptide fragments for at least two of the plurality of time points to determine the release profile of the peanut allergens. The composition can be, for example, a peanut extract, a therapeutic composition comprising peanut allergens, a dissolution medium or an analytical sample. In some embodiments the composition is an aqueous medium. In some embodiments, the amount of peanut allergens in the composition is very low. For example, the amount of a particular peanut allergen (e.g., Ara h1, Ara h2, Ara h3 or Ara h6), may be less than about 2 μg/ml, less than about 1.5 μg/ml, less than about 1 μg/ml or less than about 0.5 μg/ml, e.g., about 0.2 μg/ml, or are about 15, 16, 17, 18, 19 or 20 amino acids in length. In some embodiments, allergens in a sample taken from a composition are digested to produce allergen digest products between about 4 amino acids and about 50 amino acids in length, between about 6 amino acids and about 30 amino acids in length, or between about 15 amino acids and about 20 amino acids in length. The pattern of allergen digest products obtained after digestion creates a peptide signature for the sample.


In some embodiments, the steps of fragmenting the allergen digest products and detecting the peptide fragments include one or more of a separation method and a peptide detection method. For example, in some embodiments, the steps of detecting and identifying the peptide fragments include performing methods such as LC-MS-MS, nano-LC-MS-MS, HPLC-MS-MS, or nanoHPLC-MS-MS. In some embodiments, the peptide signature includes a collection of fragments from one or more of peanut proteins Ara h1, Ara h2, Ara h3, Ara h4, Ara h5, Ara h6, Ara h7, Ara h8, Ara h9, Ara h10, Ara h11, Ara h12 and Ara h13.


In some embodiments, a peptide signature includes one or more Ara h1 allergen digest products having amino acid sequences selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:177, SEQ ID NO:155, SEQ ID NO:93 and SEQ ID NO:40.


In some embodiments, the peptide signature comprises one or more allergen digest products from any combination of Ara h1, Ara h2 and Ara h6, or one or more allergen digest products from any combination of Ara h1, Ara h2, Ara h3 and Ara h6.


In some embodiments, the signature comprises one or more allergen digest products having amino acid sequences selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:177, SEQ ID NO:197, SEQ ID NO:198, SEQ ID NO:199, SEQ ID NO:200, SEQ ID NO:201, SEQ ID NO:236 and SEQ ID NO:237.


In certain embodiments, allergen digest products are present in more than one isoform of a peanut allergen, such as in one, two, three, four or all isoforms of a peanut allergen, such as in one, two, three, four or all isoforms of Ara h1. In one embodiment, the allergen digest products are in more than one peanut allergen isoform.


In certain exemplary embodiments, the peanut allergens are digested with one or more proteases, such as one or more proteases selected from the group consisting of trypsin, endoproteinase Lys-C and endoproteinase Arg-C.


In certain embodiments, a sample, e.g., a sample taken from an extract or pharmaceutical composition, further comprises an internal standard. An internal standard is typically a peptide having the sequence of an expected or predicted allergen digest product, and is typically less than about 50 amino acids long (such as between about 4 and about 50 amino acids long, between about 6 and about 30 amino acids long, or between about 10 and about 20 amino acids long or about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19 or about 20 amino acids long). The internal standard can be added to the composition prior to sampling, and/or added to the sample prior to the enzymatic digestion step. In certain exemplary embodiments, the internal standard comprises one or more heavy isotopes, such as, e.g., 13C and/or 15N.


In some embodiments, the signature comprises allergen digest products that do not contain missed (uncleaved) proteolytic cleavage sites.


In some embodiments, the composition comprises a particle that encases the allergens and/or has allergens bound to the surface. The particle can be, for example, a nanoparticle, a microparticle, a film, a capsule or a hydrogel.


In some embodiments, the release profile is obtained over a period of time, such as, e.g., over a period of hours, e.g., a three-hour period of time, a six-hour period of time, a twelve-hour period of time, or a twenty-four hour period of time.





BRIEF DESCRIPTION OF THE DRAWINGS

The foregoing and other features and advantages of the present invention will be more fully understood from the following detailed description of illustrative embodiments taken in conjunction with the accompanying drawings.



FIGS. 1A-1C depict a high quality Ara h1 peptide (SEQ ID NO:70) identified by tryptic digestion followed by MS-MS. (A) depicts the sequence, the number of uncleaved trypsin cleavage sites (“missed”), the percentage of False Discovery Rate (FDR), the number of post-translational modification sites (“PTM”), confirmation in BLAST (Basic Local Alignment Tool) that the identified sequence is unique to the target protein (“BLAST”), the database searched, and the number of isoforms in the database that included the identified sequence. (B) and (C) depict MS-MS data in Table form and graphical form, respectively.





DETAILED DESCRIPTION

The present invention is based in part on the development of sensitive analytical methods for measuring and profiling peanut allergens present in a composition, such as an extract or a therapeutic composition, and for determining a release profile, such as an in vitro release profile, from formulated compositions comprising peanut allergens. The methods include the detection of peptides that exist in one or more isoforms of one or more allergens in a sample. The identification of peptides found in multiple isoforms of a peanut allergen provides qualitative and quantitative information about a sample, which can be indicators of batch-to-batch consistency, and can provide a release profile of antigens stored on or in a substrate, such as a nanoparticle. The methods are particularly useful for profiling allergen content in compositions containing a very low amount of peanut allergens.


As used herein, the terms “peanut,” “groundnut” and “Arachis hypogea” are used interchangeably, and refer to a legume belonging to the Leguminosae family and the Papillionacea subfamily. Over 17 different peanut allergens have been identified. Peanut protein allergens include Ara h1, Ara h2, Ara h3, Ara H4, Ara h5, Ara h6, Ara h7, Ara h8, Ara h9, Ara h10, Ara h11, Ara h12, Ara h13, Ara h14, Ara h15, Ara h16 and Ara h17. GenBank Accession Numbers for the cDNA sequences of exemplary allergens include L34402.1 (Ara h1), AY007229.1 (Ara h2.0101), AY158467.1 (Ara h2.0201), AF093541.1 (Ara h3.0101), AF086821.1 (Ara h3.0201), AF059616 (Ara h5), AF092846.1 (Ara h6), AF091737.1 (Ara h7), EU046325.1 (AraH7.0201), AY328088.1 (Ara h8.0101), EF436550.1 (Ara h8.0201), EU159429.1 (Ara h9.0101), and EU161278.1 (Ara h9.0201), AY722694.2 (Ara h10.0101), AV72269.5.1 (Ara h10.0201), DQ097716.1 (Ara h11), EY396089.1 (Ara h12), EY396019.1 (AraH13), AAK13449 (Ara h14.0101), AAK13450 (Ara h14.0102), AAT11925 (Ara h14.0103), AAU21501 (Ara h15.0101), respectively (see, e.g., Leon et al., The peanut allergy epidemic: allergen molecular characterization and prospects for specific therapy. Expert Rev. Mol. Med. Vol. 9, Issue 1, January 2007; see also Arkwright et al., IgE Sensitization to the Nonspecific Lipid-Transfer Protein Ara h 9 and Peanut-Associated Bronchospasm, BioMed Research International, vol. 2013, Article ID 746507; see url address allergen.org/search.php?allergensource=Arachis+hypogaea).


In an exemplary embodiment, a signature of allergen digest products in a sample is obtained by digesting peanut allergens present in the sample to generate allergen digest products, fragmenting the allergen digest products to generate peptide fragments, and detecting and identifying the peptide fragments to obtain the signature of allergen digest products. As used herein, an “allergen digest product” refers to a peanut allergen present in an extract or sample that is digested, e.g., using an enzyme (e.g., trypsin, endoproteinase Lys-C, endoproteinase Arg-C and the like) to generate a product that is smaller than the intact allergen. The term “allergen digest product” also refers to the amino acid sequence of a peptide that would be produced if the peanut allergen were enzymatically digested.


As used herein, an “allergen” refers to a subset of antigens (e.g., peanut peptide antigens) which elicit the production of IgE in addition to other isotypes of antibodies. The terms “allergen,” “natural allergen,” and “wild-type allergen” may be used interchangeably.


As used herein, an “antigen” refers to a molecule (e.g., a peanut peptide) that elicits production of an antibody response (i.e., a humoral response) and/or an antigen-specific reaction with T-cells (i.e., a cellular response) in an animal.


Non-limiting examples of enzymes, specifically proteases, that are suitable to digest peanut allergens to generate allergen digest products include, but are not limited to, trypsin, endoproteinase Glu-C, endoproteinase Asp-N, chymotrypsin, endoproteinase Lys-C, and endoproteinase Arg-C, pepsin, papain, thermolysin, subtilisin, protease K, bromelain, sulfhydryl-specific protease (ficin) and the like.


As used herein, a “peptide fragment,” or “gas phase peptide fragment,” refers to any part or portion of the allergen that is smaller than the intact natural allergen that is generated by a fragmentation method that does not utilize an enzyme. Typically, fragmentation conditions are introduced in the gas phase, e.g., in a mass spectrometer step. In certain exemplary embodiments, a peptide fragment is less than 50 amino acids long, e.g., between about 2 and about 50 amino acids in length, between about 6 and about 30 amino acids in length, or between about 15 and about 20 amino acids in length or any values or sub-ranges within these ranges. In certain exemplary embodiments, a peptide fragment is about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, or about 49 amino acids in length.


In certain exemplary embodiments, a peptide fragment is generated by fragmenting an allergen digest product, and then using a separation process and a peptide identification method, such as Liquid Chromatography-tandem Mass Spectroscopy (LC-MS-MS), nano-LC-MS-MS, High Performance Liquid Chromatography-tandem MS (HPLC-MS-MS), nanoHPLC-MS-MS, UltraPerformance-tandem MS (UPLC-MS-MS), nanoUPLC-MS-MS, and Ultra High Performance-tandem MS (UHPLC-MS-MS), nanoUHPLC-MS-MS, or the like.


According to certain exemplary embodiments, a method described herein further comprises mass analyzing allergen digest products and/or peptide fragments using a mass analyzer. The mass analyzer typically comprises a triple quadrupole mass analyzer. According to other embodiments the mass analyzer may comprise a mass analyzer selected from the group consisting of: (i) triple quadrupole mass spectrometer, (ii) an orbitrap, such as a Fourier transform orbitrap, such as an Orbitrap ELITE™ (Thermo Scientific); (iii) a Fourier Transform (“FT”) mass analyzer; (ii) a Fourier Transform Ion Cyclotron Resonance (“FTICR”) mass analyzer; (iii) a Time of Flight (“TOF”) mass analyzer; (iv) an orthogonal acceleration Time of Flight (“oaTOF”) mass analyzer; (v) an axial acceleration time of flight mass analyzer; (vi) a magnetic sector mass spectrometer; (vii) a Paul or 3D quadrupole mass analyzer; (viii) a 2D or linear quadrupole mass analyzer; (ix) a Penning trap mass analyzer; (x) an ion trap mass analyzer; and (xiii) an electrostatic Fourier transform mass spectrometer.


In certain exemplary embodiments, the methods described herein utilize a separation process such as a chromatography method, e.g., liquid chromatography. According to an embodiment, fragmenting the allergen digest products and/or detecting and identifying the peptide fragments is performed by: (i) High Performance Liquid Chromatography (“HPLC”), (ii) anion exchange, (iii) anion exchange chromatography; (iv) cation exchange; (v) cation exchange chromatography; (vi) ion pair reversed-phase chromatography; (vii) chromatography; (viii) single dimensional electrophoresis; (ix) multi-dimensional electrophoresis; (x) size exclusion; (xi) affinity; (xii) reverse phase chromatography; (xiii) Capillary Electrophoresis Chromatography (“CEC”); (xiv) electrophoresis; (xv) ion mobility separation; (xvi) Field Asymmetric Ion Mobility Separation or Spectrometry (“FAIMS”); (xvii) capillary electrophoresis; and (xviii) supercritical fluid chromatography.


According to certain exemplary embodiments, the method further comprises ionizing allergen digest products and/or peptide fragments in a sample to be analyzed. The ion source may comprise a continuous ion source. According to an embodiment, the ion source may be selected from the group consisting of: (i) an Electrospray ionization (“ESI”) ion source; (ii) a Matrix Assisted Laser Desorption Ionization (“MALDI”) ion source; (iii) a Desorption Ionization on Silicon (“DIOS”) ion source; and (iv) a Desorption Electrospray Ionization (“DESI”) ion source.


The peanut allergen signatures described herein are generally determined by measurement of multiple reaction monitoring (MRM) transitions consisting of the peptide precursor ion, one or more fragment ions and a retention time. This measurement is performed, for example, on a triple quadrupole instrument. The signature can also be obtained by a combination of retention time and accurate high resolution mass spectrometric analysis of the intact peptide. These quantitation methods typically require a labeled internal standard and an external synthetic peptide calibration curve. In one embodiment, a signature is obtained by a nano-LC/MS/MS (nLC-MS-MS) analysis where as many peptides as possible are fragmented and identified by database analysis and nLC/MS/MS datasets consisting of retention time and mass/charge values and intensity are measured and compared to determine global proteome changes.


In certain exemplary embodiments, internal standards are used that include a combination of two or more allergen digest products from protein allergens, such as from one or more of Ara h1, Ara h2, Ara h3, Ara h4, Ara h5, Ara h6, Ara h7, Ara h8, Ara h9, Ara h10, Ara h11, Ara h12, Ara h13, Ara h14, Ara h15, Ara h16 and Ara h17. The internal standards typically have a known peptide sequence and are provided in a known quantity. In some embodiments, the internal standards include a combination of allergen digest products from the peanut allergens Ara h1, Ara h2 and Ara h6. In other embodiments, internal standards are used that include a combination of allergen digest products from Ara h1, Ara h2, Ara h3 and Ara h6. In certain exemplary embodiments, a sample can include one or more internal standards that comprise one or any combination of allergen digest products listed in Tables 19-23. In some embodiments, the standards are labeled, such with one or more heavy isotopes, e.g., 13C or 15N.


In certain embodiments, the allergen digest products chosen to characterize the composition uniquely represent the peanut allergen or peanut allergen isoform family and are present in the majority or in all isoforms of the allergen it is derived from, enabling its usage for specific quantitation of the allergen in question, e.g., peanut allergens. Accordingly, in certain exemplary embodiments, a signature of allergen digest products is generated. As used herein, a “signature,” or “allergenic signature” refers to the presence of a particular combination and amount of specific peanut allergen digest products (e.g., Ara h1, Ara h2, Ara h6 and/or Ara h3 allergen digest products). In certain exemplary embodiments, a signature comprises 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more distinct allergen digest products, each distinct product having a unique sequence.


The selection of peptides for use in a peanut allergen signature can be based on the identification of isoforms that include the peptides identified by the steps of digestion and fragmentation. A variety of sequence databases can be used to identify the sequences of peanut allergen isoforms, including UniProt (online at uniprot.org, and which includes the Swiss-Prot and TrEMBL databases), the database at Allergen Nomenclature (online at allergen.org), GenBank (online at ncbi.nlm.nih.gov), GeneCards (online at genecards.org), Ensembl (online at ensemble.org), and the like.


Any sequence alignment algorithm can be used to identify the isoforms that include the peptides identified following digestion and fragmentation of a sample comprising peanut allergens. Exemplary sequence alignment algorithms include BLAST (online at blast.ncbi.nlm.nih.gov/Blast.cgi), Clustal Omega (online at ebi.ac.uk/Tools/msa/clustalo), and the like.


The peanut allergen profiling methods can be useful for a variety of purposes. For example, the methods described herein are useful for performing batch-to-batch reproducibility assessments in peanut-containing compositions. The methods described herein are also useful for measuring the release or leakage of peanut allergens from the surface or interior of a vessel or substrate, such as from a bead or a particle (e.g., a nanoparticle or microparticle), a capsule, a film or a strip (such as, e.g., for sublingual administration of the composition), or a gel (such as a hydrogel).


In some embodiments, the peptide signature includes peptides that include one or more immunogenic epitopes, or one or more immunodominant epitopes of one or more peanut allergens. Peptide signatures that comprise immunogenic epitopes or immunodominant epitopes can provide a standard for the immunogenicity of a composition comprising peanut allergens.


As used herein, a “sample” refers to any composition containing peanut allergens. Exemplary samples include, but are not limited to, peanut-containing extracts, peanut-containing powders, analytical samples comprising one or more peanut allergens, pharmaceutical compositions (e.g., therapeutic vaccines), dissolution or release media and the like. In typical embodiments, a sample will be aqueous.


A sample for use in the allergen profiling methods featured in the invention can be a peanut extract, such as an extract made from whole roasted or raw peanuts, or from peanut flour. The extract can be for use in a pharmaceutical composition, such as for the treatment or prevention of peanut allergy. The extract can be used in a composition for oral immunotherapy (OIT), or sublingual immunotherapy (SLIT), or in a composition for use in a nanoparticle composition, with or without an adjuvant. By assessing the peanut allergen signature of the peanut extract, batch-to-batch consistencies can be monitored. The peanut allergen signature can also be used as a factor to deduce the immunogenicity of the extract, as extracts with similar signatures are expected to have similar immunogenic properties.


In some embodiments, the amount of peanut allergens in the composition is very low. For example, the amount of a particular peanut allergen (e.g., Ara h1, Ara h2, Ara h3 and/or Ara h6), may be less than about 2 μg/ml, less than about 1.5 μg/ml, less than about 1 μg/ml or less than about 0.5 μg/ml, e.g., about 0.2 μg/ml.


Peanut protein extracts can be made by methods known in the art including defatting and/or filtration methods to produce peanut allergen preparations.


A sample for use in the allergen profiling methods described herein can be a therapeutic composition, such as a liquid formulation for administration orally, sublingually, mucosally, intradermally, subcutaneously, intravenously, intramuscularly, parenterally or by inhalation.


In some embodiments, a therapeutic composition will be in the form of a film or a strip. A sample can include a piece of the film dissolved in a buffer prior to analysis by the allergen profiling methods described herein.


In other embodiments, the sample will include a substrate, such as a nanoparticle, a capsule, a film or tablet, or a gel, such as a hydrogel. The allergen profiling methods described herein are useful to assay release of peanut allergens from the substrate, such as, e.g., from a nanoparticle or capsule, or from a film or a hydrogel. The release can be from the interior of the substrate, e.g., a particle, or from the exterior (e.g., a surface) of a substrate. In one embodiment, the release profile is assayed by performing a complete release of peanut allergens and then assaying for a controlled release, such as over a period of time or in different culture or solution conditions (e.g., at different temperatures, pH or the like). The amount of allergen released in the controlled release assay is typically reported as a fraction or percentage as compared to the amount of allergen released under the complete release conditions.


In certain exemplary embodiments, two or more signatures obtained at specific points in time can be used to determine an in vitro release profile of peanut allergens in a sample containing a substrate (e.g., an aqueous sample, e.g., a dissolution or release medium). An in vitro release profile can be determined over a period of hours, days or weeks. In certain exemplary embodiments, a release profile is obtained over a period of time of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or about 24 hours. In certain exemplary embodiments, a release profile is obtained over a period of time of about 1, 2, 3, 4, 5, 6 or about 7 days. In certain exemplary embodiments, a release profile is obtained over a period of time of about one week, about two weeks, about three weeks, about four weeks, about five weeks, about six weeks, about seven weeks, or about eight weeks. In certain exemplary embodiments, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50 or more signatures are obtained over a particular period of time. For example, the release burst of peanut allergens can be assessed within the first 24 hours in evenly spaced time points with sufficient data to determine the burst kinetics followed by the release of the remaining amount of the same allergens from the sample to help assess overall product composition performance. The release profile can include types of allergens released from the substrate and/or the relative amounts of allergen released.


In one embodiment, the methods described herein indicate release of allergen from the surface of a substrate, e.g., a nanoparticle, by detecting a burst of peptides in a dissolution or release medium near the start of the assay. If allergen is present in the interior of the substrate, the detection of allergen over time will be more gradual according to the rate of release. Allergen that is both present on the surface of a substrate and encapsulated within the substrate may be identified by an initial burst of peptide detection in the dissolution or release medium near the start of the assay, as antigen from the surface is released from the substrate, followed by a more gradual increase in peptide detection in the dissolution or release media as allergen is released (e.g., leaked) from the interior of the substrate.


As used herein, “dissolution medium,” “dissolution media,” “release medium” and “release media” refer to a composition that is used to provide in vitro drug release information. Dissolution or release media is useful, for example, for quality control testing of a sample for determining the release and/or stability of allergen in the sample. In choosing a suitable dissolution or release medium, it is useful to determine the analytical target profile of the allergen (e.g., delayed release, constant release, extended release and the like) and/or the allergen solubility profile. For a review of dissolution media selection, see Martin and Gray (Summer 2011) Journal of Validation Technology.


As used herein, “release rate” refers to the rate that an entrapped peanut allergen agent flows from a composition and into a surrounding medium in an in vitro release test. In one exemplary embodiment, the composition is first prepared for release testing by placing the composition into the appropriate in vitro release medium. This is generally performed by exchanging the buffer after centrifugation to pellet the substrate (e.g., a synthetic nanocarrier), and reconstituting of the substrate using mild conditions. In certain embodiments, the assay is started by placing the sample at 37° C. in an appropriate temperature-controlled apparatus. A sample is typically removed at various time points.


As used herein, a “release profile” refers to the types of proteins and/or peptides, e.g., peanut allergens, that are released from a substrate or vessel over time, and may also include the rate at which each particular protein, peptide and/or allergen in or on the substrate or vessel is released.


“Exhibits a pH-sensitive dissociation” refers to a coupling between two entities, such as a peanut allergen and a substrate, e.g., a carrier molecule, that is significantly reduced or eliminated by exposure of the two entities to a change in environmental pH. In certain embodiments, relevant pH-sensitive dissociations may satisfy any of the relationships or combinations thereof provided herein.


In certain exemplary embodiments, a pharmaceutical composition comprises nanocarriers and/or microcarriers, such as synthetic nanocarriers and/or synthetic microcarriers. As used herein, a “synthetic nanocarrier” or “synthetic microcarrier” refers to a discrete object that possesses at least one dimension that is less than or equal to 5 microns in size.


In some embodiments, the mass balance is compared between one or more peanut allergens in one or more compositions to be compared, such as in a sampling from a composition over a period of time across different time points, or between different batches made at different times, and/or by different methods. The comparative data can be presented as a relative quantitation, and are typically expressed as a fraction or as a percentage.


Pharmaceutical compositions containing peanut allergens can generally be formulated with carriers, excipients, and other agents that provide suitable transfer, delivery, tolerance, and the like. Exemplary formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTIN™), anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. “Compendium of excipients for parenteral formulations” PDA (1998) J. Pharm Sci. Technol. 52:238-311.


“Pharmaceutically acceptable excipient” refers to a pharmacologically inactive substance added to a composition (e.g., a therapeutic composition) to further facilitate administration of the composition. Pharmaceutically acceptable excipients include, but are not limited to, calcium carbonate, calcium phosphate, various diluents, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, polyethylene glycols and the like.


“Dosage form” refers to a drug (e.g., one or more peanut allergens) in a medium, carrier, vehicle or device suitable for administration to a subject.


“Effective amount” refers to that amount effective for a certain purpose. For example, when the effective amount is for a therapeutic purpose, an effective amount is an amount that treats, alleviates, ameliorates, relieves, delays onset of, inhibits progression of, reduces severity of and/or reduces incidence of one or more symptoms or features of a disease, disorder and/or condition provided herein, e.g., a peanut allergy.


“Subject” refers an animal, including mammals such as humans and non-human primates; avians; domestic household or farm animals such as cats, dogs, sheep, goats, cattle, horses and pigs; laboratory animals such as mice, rats and guinea pigs; fish; and the like.


As used herein, “vaccine” refers to a composition of matter that improves the immune response to a particular disease or disorder. A vaccine typically contains factors that stimulate a subject's immune system to recognize a specific antigen (e.g., a peanut antigen) as foreign and eliminate it from the subject's body. A vaccine also establishes an immunologic “memory” such the antigen will be quickly recognized and responded to if a subject is re-challenged. Vaccines can be prophylactic (for example to prevent future infection by any pathogen) or therapeutic (for example a vaccine against a peanut antigen for the treatment of peanut allergy).


“Administering” or “administration” means providing a drug to a subject in a manner that is pharmacologically useful.


As used herein, an “epitope” refers to a binding site including an amino acid motif of between approximately six and fifteen amino acids which can be bound by an immunoglobulin (e.g., IgE, IgG, etc.) or recognized by a T-cell receptor when presented by an APC in conjunction with the major histocompatibility complex (WIC). A linear epitope is one where the amino acids are recognized in the context of a simple linear sequence. A conformational epitope is one where the amino acids are recognized in the context of a particular three dimensional structure. The peanut allergen signatures identified by the methods featured in the invention may comprise one or more peanut epitopes. An immunogenic epitope can provoke an immune response in the body, e.g., an allergic response.


As used herein, an “immunodominant epitope” refers to an epitope which is bound by antibody in a large percentage of the sensitized population or where the titer of the antibody is high, relative to the percentage or titer of antibody reaction to other epitopes present in the same antigen. In one embodiment, an immunodominant epitope is bound by antibody in more than 50% of the sensitive population, more preferably more than 60%, 70%, 80%, 90%, 95%, or 99% of the sensitive population. The peanut allergen signatures identified by the methods featured in the invention will typically comprising one or more peanut immunodominant epitopes.


It will be readily apparent to those skilled in the art that other suitable modifications and adaptations of the methods described herein may be made using suitable equivalents without departing from the scope of the embodiments disclosed herein. Having now described certain embodiments in detail, the same will be more clearly understood by reference to the following example, which is included for purposes of illustration only and is not intended to be limiting.


Example I
Peanut Allergen Signature

The development and initial preliminary validation of a method for the relative quantitation of four peanut allergens, Ara h1, Ara h2, Ara h3 and Ara h6, are described below. The method utilized liquid chromatography coupled to tandem mass spectrometry, and was based on the measurement of representative tryptic peptides derived from each of these proteins. The peptides were chosen in each case to include the majority of the reported allergenic protein isoforms.


The profiling method was dependent on the digest reproducibility and digest efficiency for the protein of interest in a complex protein mixture. Not all proteolytic peptides are good candidates for quantitation, and whereas good ionization efficiency and consistent tryptic cleavage, as well as low probability of the peptide harboring post-translational modification, are important aspects of the selection process, issues such as interference from other peptides and other matrix effects in a complex digest, are more difficult to predict. Once selected, a series of both heavy isotope labeled peptides and non-labeled synthetic versions of the selected peptides were procured and used as internal standards and for buffer calibration curves, respectively.


Peanut extracts prepared from lightly roasted peanut flour (Golden Peanut and Tree Nuts, Alpharetta, Ga.) were digested in quadruplicate with trypsin, and then a standard was added to the digested sample. The standard contained two Ara h1 peptides, two Ara h2 peptides, two Ara h3 peptides, and two Ara h6 peptides. The peptides in the internal standard were labeled with one or both of 13C and 15N. The sequences of the internal standards are provided in Tables 1 and 2 below.









TABLE 1







Unlabeled synthetic “Light” peptides for


internal standard









Peptide sequence
peptide
formula





DLAFPGSGEQVEK
ARA_L1-1
C60 H93 N15 O22


(SEQ ID NO: 238)







GTGNLELVAVR
ARA_L1-2
C48 H85 N15 O16


(SEQ ID NO: 239)







GAGSSQHQER
ARA_L2-1
C40 H65 N17 O17


(SEQ ID NO: 240)







QQEQQFK
ARA_L2-2
C40 H62 N12 O14


(SEQ ID NO: 241)







RPFYSNAPQEIFIQQGR
ARA_L3-1
C93 H139 N27 O26


(SEQ ID NO: 242)







AHVQVVDSNGNR
ARA_L3-2
C52 H86 N20 O19


(SEQ ID NO: 243)







IMGEQEQYDSYDIR
ARA_L6-1
C74 H111 N19 O28 S1


(SEQ ID NO: 244)







QMVQQFK
ARA_L6-2
C40 H65 N11 O11 S1


(SEQ ID NO: 245)
















TABLE 2







Labeled synthetic “Heavy” peptides for


internal standard









Peptide sequence
peptide
formula





DLAFPGSGEQVEK*
ARA_H1-1
13C6 C54 H93 15N2 


(SEQ ID NO: 246)

N13 O22





GTGNLELVAVR*
ARA_H1-2
13C6 C42 H85 15N4 


(SEQ ID NO: 247)

N11 O16





GAGSSQHQER*
ARA_H2-1
13C6 C34 H65 15N4 


(SEQ ID NO: 248)

N13 O17





QQEQQFK*
ARA_H2-2
13C6 C34 H62 15N2 


(SEQ ID NO: 249)

N10 O14





RPFYSNAPQEIFIQQGR*
ARA_H3-1
13C6 C87 H139 15N4 


(SEQ ID NO: 250)

N23 O26





AHVQVVDSNGNR*
ARA_H3-2
13C6 C46 H86 15N4 


(SEQ ID NO: 251)

N16 O19





IMGEQEQYDSYDIR*
ARA_H6-1
13C6 C68 H111 15N4 


(SEQ ID NO: 252)

N14 O28 S1





QMVQQFK*
ARA_H6-2
13C6 C34 H65 15N4 


(SEQ ID NO: 253)

N7 O11 S1





K* and R* are heavy isotope labeled amino acids where the carbon-12 has been replaced with carbon-13 and nitrogen-14 with nitrogen-15 resulting in 13C6 15N2 for lysine and 13C6 15N4 for arginine






Notably, additional peptides could have been selected and the particular characteristics of the proteins made it difficult to fulfill all desirable features for all peptides. For example, the two peptides for Ara h6 contain methionine, which is undesirable but difficult to avoid as Ara h6 contains an unusually high content of S-containing amino acids. Similarly, Ara h3 contains N-terminal “missed” cleavage sites that have been found to be stable. Often, tryptic cleavage sites that are very close to each other will result in preferential cleavage of one of the sites, not both.


The internal standards were used to quantitate the digested products. Buffer calibration curves of synthetic unlabeled signature peptides were generated with heavy isotope-labeled internal standards added to both the peanut extract digest and the calibration curve. Relative quantification was performed applying the internal standard and calibration curve to determine the relative amount of signature peptides in the peanut extract digest.


The samples containing the digested test allergens and the standards were analyzed by nanoHPLC-MS-MS gas phase fragmentation (Orbitrap ThermoScientific). Using the top-15 method, one sample (out of the four total) was analyzed three times, and the remaining three samples were analyzed one time, to generate a total of six data files. These data files were analyzed by Proteome Discoverer™ Software (Thermo Scientific). Applying the top-15 method to identify the most abundant proteins assured that peptides of high ionization and fragmentation efficiency would be identified in a consistent manner across the data sets.


The identities of the resulting fragments were determined using the UniProt sequence database at uniprot.org.


Over 90% of all known peanut allergens were detected from the peanut flour sample, including Ara h1, Ara h2, Ara h3, Ara h5, Ara h6, Ara h7, Ara h8, Ara h10, Ara h11, Ara h14, Ara h15, and Ara h Agglutinin. A survey of the identified allergens is illustrated in Tables 3-18 below.


Ara h1, Ara h2, Ara h3 and Ara h6 represent the major peanut allergens.









TABLE 3







Ara h1 isoforms detected in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 1
B3IXL2
No



Ara h 1
E5G076
Yes



Ara h 1
N1NEW2
No



Ara h 1
N1NG13
No



Ara h 1
P43237
Yes



Ara h 1
Q6PSU3
Yes



Ara h 1
Q6PSU4
Yes



Ara h 1
Q6PSU5
Yes



Ara h 1
Q6PSU6
Yes



Ara h 1.0101
P43238
Yes

















TABLE 4







Ara h2 isoforms detected in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 2
C0LJJ1
No



Ara h 2.0101
Q6PSU2
Yes



Ara h 2.0201
Q6PSU2
Yes

















TABLE 5







Ara h3 isoforms detected in peanut flour













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 3
A1DZF0
Yes



Ara h 3
A1DZF1
Yes



Ara h 3
B5TYU1
Yes



Ara h 3
E5G077
Yes



Ara h 3
Q0GM57
Yes



Ara h 3
Q5I6T2
Yes



Ara h 3
Q647H2
Yes



Ara h 3
Q647H3
Yes



Ara h 3
Q647H4
Yes



Ara h 3
Q6IWG5
Yes



Ara h 3
Q6T2T4
Yes



Ara h 3
Q8LKN1
Yes



Ara h 3
Q8LL03
Yes



Ara h 3
Q9FZ11
Yes



Ara h 3.0101
O82580
Yes



Ara h 3.0201
Q9SQH7
Yes



n/a
O82580
Yes



n/a
Q9SQH7
Yes

















TABLE 6







Ara h5 isoforms identified in peanut flour













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 5
D3K177
Yes



Ara h 5
L7QH52
Yes



Ara h 5
Q5XXQ5
Yes



Ara h 5.0101
Q9SQI9
Yes

















TABLE 7







Ara h6 isoforms identified in peanut flour













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 6
A1DZE9
Yes



Ara h 6
A5Z1R0
No



Ara h 6.0101
Q647G9
Yes

















TABLE 8







Ara h7 isoforms identified in peanut flour













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 7
Q647G8
Yes



Ara h 7.0101
Q9SQH1
Yes



Ara h 7.0201
B4XID4
Yes

















TABLE 9







Ara h8 isoforms identified in peanut flour













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 8
B1PYZ4
Yes



Ara h 8
B2ZGS2
Yes



Ara h 8
Q0PKR4
Yes



Ara h 8
Q2YHR1
Yes



Ara h 8.0101
Q6VT83
Yes



Ara h 8.0201
B0YIU5
Yes

















TABLE 10







Ara h9 isoforms identified in peanut flour













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 9.0101
B6CEX8
Yes



Ara h 9.0201
B6CG41
Yes

















TABLE 11







Ara h10 isoforms identified in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 10.0101
Q647G5
Yes



Ara h 10.0102
Q647G4
Yes

















TABLE 12







Ara h11 isoforms identified in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 11.0101
Q45W87
Yes



Ara h 11.0102
Q45W86
Yes

















TABLE 13







Ara h12 isoforms identified in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 12.0101
EY396089
No

















TABLE 14







Ara h13 isoforms identified in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 13.0101
EY396019
No

















TABLE 15







Ara h14 isoforms identified in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 14.0101
Q9AXI1
Yes



Ara h 14.0102
Q9AXI0
Yes



Ara h 14.0103
Q6J1J8
Yes

















TABLE 16







Ara h15 isoforms identified in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h 15.0101
Q647G3
Yes

















TABLE 17







Ara h Agglutinin isoforms identified in peanut flour.













Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara h Agglutinin
P02872
Yes



Ara h Agglutinin
Q38711
Yes



Ara h Agglutinin
Q43373
Yes



Ara h Agglutinin
Q43375
Yes



Ara h Agglutinin
Q8W0P8
Yes

















TABLE 18







Ara h hypothetical isoforms identified in peanut flour.











Hypothetical

Peak Identified by



Peanut Allergen
UniProt Ref
nanoHPLC-MS-MS







Ara i 2
A5Z1Q9
No



Ara i 6
A5Z1Q6
No



Ara d 2
A5Z1Q8
No



Ara d 2
A8VT41
No



Ara d 2
A8VT44
No



Ara d 2
A8VT45
No



Ara d 2
A8VT50
No



Ara d 6
A5Z1Q5
No










Peptides of the major allergens are identified to form a collection of peptides for use as an indicator of peanut allergen content in a composition (e.g., a “peanut peptide signature”). An ideal peptide signature utilizes peptides that represent completely cleaved true tryptic digest products (with the possible exception of peptides that contain sequential arginine and/or lysine residues), where one or the other cleavage site is typically preferentially cleaved. Selected peptides also typically have sequence conservation with a False Discovery Rate (FDR) of less than 1%, and sequence conservation across multiple isoforms. Further, selected peptides typically have no or minimal post-translational modifications, including oxidation and glycosylation, and have high detection quality based on manual curation of tandem mass spectrometry (MS-MS). Isoforms are represented by genetic variations, including peptides generated by truncated or deleted sequences. An example of a high quality peptide as identified by MS-MS is the Ara h1 peptide illustrated in FIGS. 1A to 1C.


For example, Ara h1k heterogeneity was determined by comparing sequence variations in the UniProt database records. Exemplary Ara h1 allergen digest products identified from UniProt reference P43238 are shown in Table 19. Sequences from other UniProt Ara h1 records were also compared (see Table 20). Preferred Ara h1 allergen digest products were selected as being useful in a peanut allergen signature based on: 1) preferentially zero missed tryptic cleavage sites; 2) presence in all sequences with a False Discovery Rate (FDR) of less than 1%; 3) no or minimal post-translational modification sites; 4) high quality fragments as determined by manual curation of tandem Mass Spectroscopy (MS-MS) data; 5) an indication by BLAST search that the sequence is unique within the peanut proteome; and 6) presence of the sequence a maximum number of isoforms. The resulting initial list of allergen digest products that were determined to be most useful candidates for profiling peanut antigens in the peanut flour extract are shown in Tables 20-23, representing peanut allergens from Ara h1, Ara h2, Ara h3 and Ara h6, respectively.









TABLE 19







Ara h1 allergen digest products from UniProt


Ref. P43238.











SEQ




ID



Ara h1 Peptide Sequence
NO:







ACESRCTKLEYDPR
  1







ACESRCTKLEYDPRCVYDPR
  2







AMVIVVVNK
  3







AMVIVVVNKGTGNLELVAVR
  4







AMVIVVVNKGTGNLELVAVRK
  5







CLQSCQQEPDDLK
  6







CLQSCQQEPDDLKQK
  7







CLQSCQQEPDDLKQKACESR
  8







CLQSCQQEPDDLKQKACESRCTK
  9







CTKLEYDPR
 10







CTKLEYDPRCVYDPR
 11







CTKLEYDPRCVYDPRGHTGTTNQR
 12







CVYDPR
 13







CVYDPRGHTGTTNQR
 14







CVYDPRGHTGTTNQRSPPGER
 15







CVYDPRGHTGTTNQRSPPGERTR
 16







DLAFPGSGEQVEK
 17







DLAFPGSGEQVEKLIK
 18







DNVIDQIEKQAK
 19







DQSSYLQGFSR
 20







DQSSYLQGFSRNTLEAAFNAEFNEIRR
 21







EDQEEENQGGKGPLLSILK
 22







EDWRRPSHQQPR
 23







EEDWRQPREDWR
 24







EEDWRQPREDWRRPSHQQPR
 25







EEEEDEDEEEEGSNREVRRYTAR
 26







EEGGRWGPAGPR
 27







EEGGRWGPAGPRER
 28







EEGGRWGPAGPRERER
 29







EETSRNNPFYFPSR
 30







EETSRNNPFYFPSRR
 31







EETSRNNPFYFPSRRFSTR
 32







EGALMLPHFNSK
 33







EGALMLPHFNSKAMVIVVVNK
 34







EGDVFIMPAAHPVAINASSELHLLGFGINAENNHRIFLAGDK
 35







EGDVFIMPAAHPVAINASSELHLLGFGINAENNHRIFLAGDK
 36



DNVIDQIEK








EGEPDLSNNFGK
 37







EGEPDLSNNFGKLFEVKPDKK
 38







EGEPDLSNNFGKLFEVKPDKKNPQLQDLDMMLTCVEIK
 39







EGEQEWGTPGSHVR
 40







EGEQEWGTPGSHVREETSRNNPFYFPSRR
 41







EHVEELTK
 42







EHVEELTKHAK
 43







EQQQRGRR
 44







EQQQRGRREEEEDEDEEEEGSNR
 45







EREEDWR
 46







EREEDWRQPR
 47







EREREEDWR
 48







EREREEDWRQPR
 49







ESHFVSARPQSQSQSPSSPEK
 50







ESHFVSARPQSQSQSPSSPEKESPEKEDQEEENQGGK
 51







ESPEKEDQEEENQGGK
 52







EVRRYTAR
 53







FDQRSR
 54







FDQRSRQFQNLQNHR
 55







FSTRYGNQNGR
 56







FSTRYGNQNGRIR
 57







FSTRYGNQNGRIR
 58







GHTGTTNQRSPPGERTR
 59







GHTGTTNQRSPPGERTRGR
 60







GQRRWSTR
 61







GRQPGDYDDDR
 62







GRQPGDYDDDRR
 63







GRQPGDYDDDRRQPR
 64







GRQPGDYDDDRRQPR
 65







GRREEEEDEDEEEEGSNR
 66







GRREEEEDEDEEEEGSNREVR
 67







GSEEEGDITNPINLR
 68







GSEEEGDITNPINLREGEPDLSNNFGK
 69







GTGNLELVAVR
 70







GTGNLELVAVRK
 71







HADADNILVIQQGQATVTVANGNNR
 72







HADADNILVIQQGQATVTVANGNNRK
 73







HADADNILVIQQGQATVTVANGNNRKSFNLDEGHALR
 74







HAKSVSKK
 75







HAKSVSKKGSEEEGDITNPINLR
 76







HDNQNLR
 77







HDNQNLRVAKISMPVNTPGQFEDFFPASSR
 78







IFLAGDKDNVIDQIEK
 79







IFLAGDKDNVIDQIEKQAK
 80







IFLAGDKDNVIDQIEKQAKDLAFPGSGEQVEK
 81







IPSGFISYILNR
 82







IPSGFISYILNRHDNQNLR
 83







IPSGFISYILNRHDNQNLRVAK
 84







IPSGFISYILNRHDNQNLRVAKISMPVNTPGQFEDFFPASSR
 85







IRPEGREGEQEWGTPGSHVREETSR
 86







IRPEGREGEQEWGTPGSHVREETSRNNPFYFPSR
 87







IRVLQRFDQR
 88







ISMPVNTPGQFEDFFPASSR
 89







ISMPVNTPGQFEDFFPASSRDQSSYLQGFSR
 90







ISMPVNTPGQFEDFFPASSRDQSSYLQGFSRNTLEAAFNAEF
 91



NEIR








ISMPVNTPGQFEDFFPASSRDQSSYLQGFSRNTLEAAFNAEF
 92



NEIRR








IVQIEAKPNTLVLPK
 93







KEQQQR
 94







KEQQQRGRR
 95







KGSEEEGDITNPINLR
 96







KGSEEEGDITNPINLREGEPDLSNNFGK
 97







KGSEEEGDITNPINLREGEPDLSNNFGKLFEVKPDK
 98







KIRPEGREGEQEWGTPGSHVREETSR
 99







KNPQLQDLDMMLTCVEIK
100







KNPQLQDLDMMLTCVEIKEGALMLPHFNSK
101







KNPQLQDLDMMLTCVEIKEGALMLPHFNSKAMVIVVVNK
102







KSFNLDEGHALR
103







KSFNLDEGHALRIPSGFISYILNR
104







KSFNLDEGHALRIPSGFISYILNRHDNQNLR
105







KTENPCAQR
106







KTENPCAQRCLQSCQQEPDDLK
107







KTENPCAQRCLQSCQQEPDDLKQK
108







LEYDPR
109







LEYDPRCVYDPR
110







LEYDPRCVYDPRGHTGTTNQR
111







LEYDPRCVYDPRGHTGTTNQRSPPGER
112







LFEVKPDK
113







LFEVKPDKK
114







LFEVKPDKKNPQLQDLDMMLTCVEIK
115







LFEVKPDKKNPQLQDLDMMLTCVEIKEGALMLPHFNSK
116







LIKNQKESHFVSARPQSQSQSPSSPEK
117







NNPFYFPSR
118







NNPFYFPSRR
119







NNPFYFPSRRFSTR
120







NNPFYFPSRRFSTRYGNQNGR
121







NPQLQDLDMMLTCVEIK
122







NPQLQDLDMMLTCVEIKEGALMLPHFNSK
123







NPQLQDLDMMLTCVEIKEGALMLPHFNSKAMVIVVVNK
124







NQKESHFVSARPQSQSQSPSSPEK
125







NQKESHFVSARPQSQSQSPSSPEKESPEKEDQEEENQGGK
126







NTLEAAFNAEFNEIR
127







NTLEAAFNAEFNEIRR
128







NTLEAAFNAEFNEIRRVLLEENAGGEQEER
129







NTLEAAFNAEFNEIRRVLLEENAGGEQEERGQR
130







QAKDLAFPGSGEQVEK
131







QFQNLQNHR
132







QFQNLQNHRIVQIEAKPNTLVLPKHADADNILVIQQGQATVT
133



VANGNNRK








QKACESR
134







QKACESRCTKLEYDPR
135







QPGDYDDDRR
136







QPGDYDDDRRQPR
137







QPGDYDDDRRQPRR
138







QPREDWRRPSHQQPR
139







QPREDWRRPSHQQPRK
140







QPRREEGGR
141







QPRREEGGRWGPAGPR
142







REEEEDEDEEEEGSNR
143







REEEEDEDEEEEGSNREVR
144







REEEEDEDEEEEGSNREVRR
145







REEGGR
146







REEGGRWGPAGPR
147







REEGGRWGPAGPRER
148







RFSTRYGNQNGR
149







RFSTRYGNQNGRIR
150







RPSHQQPR
151







RVLLEENAGGEQEER
152







RVLLEENAGGEQEERGQR
153







RWSTRSSENNEGVIVK
154







SFNLDEGHALR
155







SFNLDEGHALRIPSGFISYILNR
156







SFNLDEGHALRIPSGFISYILNRHDNQNLR
157







SPPGERTRGR
158







SPPGERTRGRQPGDYDDDR
159







SRQFQNLQNHR
160







SRQFQNLQNHRIVQIEAKPNTLVLPKHADADNILVIQQGQAT
161



VTVANGNNR








SSENNEGVIVK
162







SSENNEGVIVKVSK
164







SSENNEGVIVKVSKEHVEELTK
164







SSPYQKK
165







SSPYQKKTENPCAQR
166







SSPYQKKTENPCAQRCLQSCQQEPDDLK
167







SVSKKGSEEEGDITNPINLR
168







TENPCAQR
169







TENPCAQRCLQSCQQEPDDLK
170







TENPCAQRCLQSCQQEPDDLKQK
171







TENPCAQRCLQSCQQEPDDLKQKACESR
172







TRGRQPGDYDDDR
173







TRGRQPGDYDDDRR
174







VAKISMPVNTPGQFEDFFPASSR
175







VAKISMPVNTPGQFEDFFPASSRDQSSYLQGFSRNTLEAAFN
176



AEFNEIR








VLLEENAGGEQEER
177







VLLEENAGGEQEERGQR
178







VLLEENAGGEQEERGQRR
179







VLLEENAGGEQEERGQRRWSTR
180







VLQRFDQRSRQFQNLQNHR
181







VSKEHVEELTK
182







VSPLMLLLGILVLASVSATHAK
183







WGPAGPR
184







WGPAGPRER
185







WGPAGPRERER
186







WSTRSSENNEGVIVK
187







WSTRSSENNEGVIVKVSKEHVEELTK
188







YGNQNGR
189







YGNQNGRIRVLQR
190







YGNQNGRIRVLQRFDQR
191







YTARLKEGDVFIMPAAHPVAINASSELHLLGFGINAENNHRI
192



FLAGDK

















TABLE 20







Ara h1 Sequences Identified in Prepared Peanut 


Extract










#




Appearance




in 7
UniProt


Ara h1 Peptide
UniProt
Reference


Sequence
References
Nos





DLAFPGSGEQVEK
7
P43238,


(SEQ ID NO: 17)

P43237,




E5G076,




Q6PSU3,




Q6PSU6,




Q6PSU5,




Q6PSU4





GTGNLELVAVR
7
P43238,


(SEQ ID NO: 70)

P43237,




E5G076,




Q6PSU3,




Q6PSU6,




Q6PSU5,




Q6PSU4





VLLEENAGGEQEER
7
P43238,


(SEQ ID NO: 177)

P43237,




E5G076,




Q6PSU3,




Q6PSU6,




Q6PSU5,




Q6PSU4





DQSSYLQGFSR
5
P43238,


(SEQ ID NO: 20)

P43237,




E5G076,




Q6PSU3,




Q6PSU4





HADADNILVIQQGQATVTVANGNNR
5
P43238,


(SEQ ID NO: 72)

P43237,




E5G076,




Q6PSU3,




Q6PSU4





NTLEAAFNAEFNEIR
5
P43238,


(SEQ ID NO: 127)

P43237,




E5G076,




Q6PSU3,




Q6PSU4





EQEWEEEEEDEEEEGSNR
4
P43237,


(SEQ ID NO: 193)

E5G076,




Q6PSU3,




Q6PSU6





IPSGFISYILNR
4
P43238,


(SEQ ID NO: 82)

P43237,




Q6PSU3,




Q6PSU4





NNPFYFPSR
4
P43238,


(SEQ ID NO: 118)

P43237,




E5G076,




Q6PSU3





SFNLDEGHALR
4
P43238,


(SEQ ID NO: 155)

P43237,




Q6PSU3,




Q6PSU4





GSEEEDITNPINLR
3
P43237,


(SEQ ID NO: 194)

Q6PSU3,




Q6PSU6





IVQIEAKPNTLVLPK
3
P43238,


(SEQ ID NO: 93)

E5G076,




Q6PSU4





EGEQEWGTPGSEVR
2
P43237,


(SEQ ID NO: 195)

Q6PSU3





EGEQEWGTPGSHVR
2
P43238,


(SEQ ID NO: 40)

E5G076





IVQIEARPNTLVLPK
2
P43237,


(SEQ ID NO: 196)

Q6PSU3
















TABLE 21







Ara h2 Sequences Identified in Prepared


Peanut Extract










#




Appearance



Ara h2
in 1
UniProt


Peptide
UniProt
Reference


Sequence
References
Nos





GAGSSQHQER
1
Q6PSU2


(SEQ ID NO: 197)







QQEQQFK
1
Q6PSU2


(SEQ ID NO: 198)
















TABLE 22







Ara h3 Sequences Identified in Prepared


Peanut Extract










#




Appearance




in 16
UniProt


Ara h3
UniProt
Reference


Peptide Sequence
References
Nos





RPFYSNAPQEIFIQQGR
12
Q5I6T2,


(SEQ ID NO: 199)

B5TYU1,




Q9FZ11,




A1DZF1,




Q82580,




Q8LL03,




Q9SQH7,




Q647H4,




Q6T2T4,




Q8LKN1,




Q647H3,




A1DZF0





AHVQVVDSNGNR
10
Q5I6T2,


(SEQ ID NO: 200)

B5TYU1,




Q9FZ11,




O82580,




Q0GM57,




E5G077,




Q9SQH7,




Q647H3,




Q6IWG5,




A1DZF0





FNLAGNHEQEFLR
10
Q5I6T2,


(SEQ ID NO: 201)

B5TYU1,




Q9FZ11,




Q8LL03,




Q9SQH7,




Q647H4,




Q6T2T4,




Q8LKN1,




Q647H3,




A1DZF0





NALFVPHYNTNAHSIIYALR
 9
Q5I6T2 (3x),


(SEQ ID NO: 202)

B5TYU1 (3x),




Q9FZ11 (3x),




Q9SQH7 (3x),




Q647H4 (3x),




Q6T2T4 (3x),




Q8LKN1 (3x),




Q647H3 (3x),




A1DZF0 (3x)





SPDIYNPQAGSLK
 9
Q5I6T2,


(SEQ ID NO: 203)

B5TYU1,




Q9FZ11,




O82580,




Q647H4,




Q6T2T4,




Q8LKN1,




Q647H3,




A1DZF0





WLGLSAEYGNLYR
 9
Q5I6T2,


(SEQ ID NO: 204)

B5TYU1,




Q9FZ11,




Q9SQH7,




Q647H4,




Q6T2T4,




Q8LKN1,




Q647H3,




A1DZF0





VYDEELQEGHVLVVPQNFAVAGK
 7
Q5I6T2,


(SEQ ID NO: 205)

B5TYU1,




Q9FZ11,




O82580,




Q9SQH7,




Q647H3,




A1DZF0





SQSENFEYVAFK
 6
O82580 (3x),


(SEQ ID NO: 206)

Q9SQH7 (3x),




Q647H4 (3x),




Q6T2T4 (3x),




Q8LKN1 (3x),




A1DZF0 (3x)





AGQEQENEGGNIFSGFTPEFLAQA
 4
Q647H4 (3x),


FQVDDR

Q6T2T4 (3x),


(SEQ ID N0: 207)

Q8LKN1 (3x),




Q647H3 (3x)





GENESDEQGAIVTVR
 4
Q647H4,


(SEQ ID NO: 208)

Q6T2T4,




Q8LKN1,




Q647H3





QQYERPDEEEEYDEDEYEYDEEER
 4
Q647H4,


(SEQ ID NO: 209)

Q6T2T4,




Q8LKN1,




Q647H3





SQSDNFEYVAFK
 4
Q5I6T2,


(SEQ ID NO: 210)

B5TYU1,




Q9FZ11,




Q647H3





TANDLNLLILR
 4
Q5I6T2,


(SEQ ID NO: 211)

Q9FZ11,




O82580,




Q8LKN1





TANELNLLILR
 4
B5TYU1 (3x),


(SEQ ID NO: 212)

Q647H4 (3x),




Q6T2T4 (3x),




A1DZF0 (3x)





TDSRPSIANLAGENSFIDNLPEEV
 4
Q647H4 (3x),


VANSYGLPR

Q6T2T4 (3x),


(SEQ ID NO: 213)

Q8LKN1 (3x),




A1DZF0 (3x)





AHVQVVDSNGDR
 3
Q647H4,


(SEQ ID NO: 214)

Q6T2T4,




Q8LKN1





AQSENYEYLAFK
 3
Q0GM57,


(SEQ ID NO: 215)

E5G077,




Q6IWG5





FNEGDLIAVPTGVAFWLYNDHDTD
 3
B5TYU1,


VVAVSLTDTNNNDNQLDQFPR

Q9SQH7,


(SEQ ID NO: 216)

A1DZF0





GADEEEEYDEDEYEYDEEDR
 3
Q5I6T2,


(SEQ ID NO: 217)

B5TYU1,




Q9FZ11





SSNPDIYNPQAGSLR
 3
Q0GM57,


(SEQ ID NO: 218)

E5G077,




Q6IWG5





SVNELDLPILGWLGLSAQHGTIYR
 3
Q0GM57,


(SEQ ID NO: 219)

E5G077,




Q6IWG5





VFDEELQEGHVLVVPQNFAVAGK
 3
Q647H4,


(SEQ ID NO: 220)

Q6T2T4,




Q8LKN1





VYDEELQEGHVLVVPQNFAVAAK
 3
Q0GM57,


(SEQ ID NO: 221)

E5G077,




Q6IWG5





AGQEEENEGGNIFSGFTPEFLAQA
 2
B5TYU1,


FQVDDR

Q9FZ11


(SEQ ID NO: 222)







AGQEEENEGGNIFSGFTPEFLEQA
 2
Q5I6T2,


FQVDDR

O82580


(SEQ ID NO: 223)







FFVPPFQQSPR
 2
Q9SQH7,


(SEQ ID NO: 224)

A1DZF0





TDSRPSIANLAGENSIIDNLPEEV
 2
Q0GM57 (3x),


VANSYR

Q6IWG5 (3x)


(SEQ ID NO: 225)







AGQEEEDEGGNIFSGFTPEFLEQA
 1
Q9SQH7


FQVDDR




(SEQ ID NO: 226)







AGQEQENEGGNIFSGFTSEFLAQA
 1
A1DZF0


FQVDDR




(SEQ ID NO: 227)







GENESEEEGAIVTVK
 1
Q9FZ11


(SEQ ID NO: 228)







GENESEEQGAIVTVK
 1
A1DZF0 (3x)


(SEQ ID NO: 229)







SPDEEEEYDEDEYAEEER
 1
A1DZF0


(SEQ ID NO: 230)







SQSEHFLYVAFK
 1
Q647H2


(SEQ ID NO: 231)







TDSRPSIANLAGENSIIDNLPEEV
 1
Q647H3 (3x)


VANSYGLPR




(SEQ ID NO: 232)







TDSRPSIANLAGENSVIDNLPEEV
 1
B5TYU1


VANSYGLPR




(SEQ ID NO: 233)







TDSRPSIANQAGENSIIDNLPEEV
 1
E5G077


VANSYR




(SEQ ID NO: 234)







VFDEELQEGQSLVVPQNFAVAAK
 1
Q647H2


(SEQ ID NO: 235)
















TABLE 23







Ara h6 Sequences Identified in Prepared


Peanut Extract










#




Appearance



Ara h6
in 2
UniProt


Peptide
UniProt
Reference


Sequence
References
Nos





IMGEQEQYDSYDIR
2
Q647G9,


(SEQ ID NO: 236)

A1DZE9





QMVQQFK
1
Q647G9


(SEQ ID NO: 237)









EQUIVALENTS

The disclosure may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting of the disclosure. Scope of the disclosure is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are therefore intended to be embraced herein.

Claims
  • 1. A method for determining a signature of peanut allergens in an aqueous medium comprising: digesting peanut allergens present in an aqueous medium to generate allergen digest products;fragmenting the allergen digest products to generate peptide fragments; anddetermining the signature of allergen digest products of peanut allergens of the aqueous medium.
  • 2. The method of claim 1, wherein the aqueous medium is a dissolution medium, a release medium, or an analytical sample, and/or wherein the aqueous medium further comprises an internal standard.
  • 3. (canceled)
  • 4. The method of claim 1, wherein the amount of peanut allergens in the medium is less than about 2 μg/ml, less than about 1.5 μg/ml, less than about 1 μg/ml, or less than about 0.5 μg/ml.
  • 5. The method of claim 1, further comprising the step of comparing the signature to a signature standard, and/or wherein the steps of fragmenting the allergen digest products and determining the signature are performed by a method selected from the group consisting of one or any combination of LC-MS, LC-MS-MS, nano-LC-MS-MS, and nanoHPLC-MS-MS.
  • 6. The method of claim 1, wherein the allergen digest products are between about 4 amino acids and about 50 amino acids in length, between about 6 amino acids and about 30 amino acids in length, or between about 15 amino acids and about 20 amino acids in length.
  • 7-9. (canceled)
  • 10. The method of claim 1, wherein the signature comprises: Ara h1 allergen digest products having an amino acid sequence selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:177, SEQ ID NO:155, SEQ ID NO:93 and SEQ ID NO:40;allergen digest products from Ara h1, Ara h2 and Ara h6;allergen digest products from Ara h1, Ara h2, Ara h3 and Ara h6, optionally having having amino acid sequences selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:177, SEQ ID NO:197, SEQ ID NO:198, SEQ ID NO:199, SEQ ID NO:200, SEQ ID NO:201, SEQ ID NO:236 and SEQ ID NO:237; ordigest products that do not contain missed proteolytic cleavage sites.
  • 11-13. (canceled)
  • 14. The method of claim 1, wherein the allergen digest products are present: in a majority of isoforms of Ara h1;in at least 90% of isoforms of Ara h1;in all isoforms of Ara h1;in a majority of isoforms of Ara h1, Ara h2 and Ara h6; orin a majority of isoforms of Ara h1, Ara h2, Ara h3 and Ara h6.
  • 15-18. (canceled)
  • 19. The method of claim 1, wherein the peanut allergens are digested with one or more proteases selected from the group consisting of trypsin, endoproteinase Lys-C and endoproteinase Arg-C.
  • 20. (canceled)
  • 21. The method of claim 1, wherein the internal standard comprises one or more heavy isotopes.
  • 22. (canceled)
  • 23. A method for determining an in vitro release profile of an aqueous medium comprising peanut allergens comprising: obtaining a sample from the aqueous medium at each of a plurality of time points;digesting the peanut allergens present in the samples to generate allergen digest products;fragmenting the allergen digest products to generate peptide fragments; anddetecting the peptide fragments for at least two of the plurality of time points to identify the allergen digest products, thereby determining the in vitro release profile of the peanut allergens from the aqueous medium.
  • 24. The method of claim 23, wherein the aqueous medium is a dissolution medium, release medium, or an analytical sample.
  • 25. The method of claim 23, wherein the amount of peanut allergens in the aqueous medium is less than about 2 μg/ml, less than about 1.5 μg/ml, less than about 1 μg/ml, or less than about 0.5 μg/ml.
  • 26. (canceled)
  • 27. The method of claim 23, wherein the allergen digest products are between about 4 amino acids and about 50 amino acids in length, between about 6 amino acids and about 30 amino acids in length, or are between about 15 amino acids and about 20 amino acids in length.
  • 28-29. (canceled)
  • 30. The method of claim 23, wherein the steps of fragmenting the allergen digest products and detecting the peptide fragments are performed by a method selected from the group consisting of one or a combination of Liquid Chromatography-tandem Mass Spectroscopy (LC-MS-MS), nanoLC-MS-MS, and nano High Performance Liquid Chromatography-tandem Mass Spectroscopy (nanoHPLC-MS-MS).
  • 31. The method of claim 23, wherein the signatures comprise: Ara h1 allergen digest products having amino acid sequences selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:177, SEQ ID NO:155, SEQ ID NO:93 and SEQ ID NO:40;allergen digest products from Ara h1, Ara h2 and Ara h6;allergen digest products from Ara h1, Ara h2, Ara h3 and Ara h6; optionally selected from the group consisting of SEQ ID NO:17, SEQ ID NO:70, SEQ ID NO:177, SEQ ID NO:197, SEQ ID NO:198, SEQ ID NO:199, SEQ ID NO:200, SEQ ID NO:201, SEQ ID NO:236 and SEQ ID NO:237; orallergen digest fragments that do not contain missed proteolytic cleavage sites.
  • 32-34. (canceled)
  • 35. The method of claim 23, wherein the allergen digest products are present: in a majority of isoforms of Ara h1;in at least 90% of isoforms of Ara h1;in all isoforms of Ara h1; orin a majority of isoforms of Ara h1, Ara h2 and Ara h6.
  • 36-38. (canceled)
  • 39. The method of claim 23, wherein the peanut allergens are digested with one or more proteases selected from the group consisting of trypsin, endoproteinase Lys-C and endoproteinase Arg-C.
  • 40. The method of claim 23, wherein the aqueous medium further comprises the use of an internal standard, wherein the internal standard comprises one or more heavy isotopes.
  • 41-42. (canceled)
  • 43. The method of claim 23, wherein the composition comprises one or both of nanoparticles and microparticles.
  • 44. The method of claim 23, wherein the release profile is obtained over a three-hour period of time, over a six-hour period of time, over a twelve-hour period of time, or over a twenty-four hour period of time.
  • 45-47. (canceled)
Priority Claims (1)
Number Date Country Kind
163306642.6 Dec 2016 FR national
RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 16/580,638, filed Sep. 24, 2019, which is a division of U.S. patent application Ser. No. 15/392,233, filed Dec. 28, 2016, now U.S. Pat. No. 10,466,250, which claims the benefit of U.S. Provisional Application No. 62/272,094, filed Dec. 29, 2015, and French Patent Application No. 163306642.6, filed Dec. 8, 2016, each of which is incorporated herein by reference in its entirety.

Provisional Applications (1)
Number Date Country
62272094 Dec 2015 US
Divisions (1)
Number Date Country
Parent 15392233 Dec 2016 US
Child 16580638 US
Continuations (1)
Number Date Country
Parent 16580638 Sep 2019 US
Child 17523050 US