WO2016018978A1 - Quantifying monoclonal antibody therapeutics by lc-ms/ms - Google Patents

Quantifying monoclonal antibody therapeutics by lc-ms/ms Download PDF

Info

Publication number
WO2016018978A1
WO2016018978A1 PCT/US2015/042580 US2015042580W WO2016018978A1 WO 2016018978 A1 WO2016018978 A1 WO 2016018978A1 US 2015042580 W US2015042580 W US 2015042580W WO 2016018978 A1 WO2016018978 A1 WO 2016018978A1
Authority
WO
WIPO (PCT)
Prior art keywords
sample
monoclonal antibody
infliximab
antibody therapeutic
internal standard
Prior art date
Application number
PCT/US2015/042580
Other languages
French (fr)
Inventor
David R. Barnidge
Maria Alice Vieira WILLRICH
David L. Murray
Melissa SNYDER
Original Assignee
Mayo Foundation For Medical Education And Research
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 Mayo Foundation For Medical Education And Research filed Critical Mayo Foundation For Medical Education And Research
Priority to US15/329,512 priority Critical patent/US10690676B2/en
Priority to EP15827198.1A priority patent/EP3175242A4/en
Publication of WO2016018978A1 publication Critical patent/WO2016018978A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • G01N33/6854Immunoglobulins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • This disclosure relates to methods for quantifying monoclonal antibody therapeutics using mass spectrometry techniques.
  • Human monoclonal antibodies contain two identical heavy chain polypeptides (each about 54 kilodaltons in MW) and two identical light chain polypeptides (each about 24 kilodaltons in molecular weight) which are bound together by disulfide bonds.
  • Each light chain and each heavy chain include a constant region and a variable region. The variable region is located on the N-terminal portion of each chain and the constant region is located on the C-terminal portion of each chain.
  • the constant regions of the light chains and heavy chains have different amino acid sequences, and can be used to identify the isotype of the heavy or light chain.
  • a method for quantifying a monoclonal antibody therapeutic in a sample comprising: providing a sample comprising a monoclonal antibody therapeutic; adding an internal standard to the sample, wherein the internal standard is a non- isotopically labeled immunoglobulin from a non-human species; and subjecting the sample to a mass spectrometry technique to quantify the monoclonal antibody therapeutic in the sample.
  • the monoclonal antibody therapeutic is a humanized monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is a human monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituximab and adalimumab. For example, the monoclonal antibody therapeutic can be infliximab.
  • the non-isotopically labeled immunoglobulin from a non- human species is a non-human IgG antibody. In some embodiments, the non-isotopically labeled immunoglobulin from a non-human species is a horse IgG.
  • the method further comprises digesting the antibodies in the sample prior to the mass spectrometry step.
  • digesting the antibodies comprises digestion using trypsin.
  • the method further comprises purifying the sample prior to the mass spectrometry step.
  • the sample may be purified using antibody affinity resin purification.
  • the mass spectrometry technique comprises a tandem mass spectrometry (MS/MS) technique.
  • the mass spectrometry technique comprises an LC-MS/MS technique.
  • the mass spectrometry technique can be a LC-ESI TRIPLE QUAD MS.
  • the mass spectrometry technique comprises the use of positive ion mode.
  • the mass spectrometry technique comprises selective reaction monitoring (SRM) analysis.
  • the SRM analysis comprises monitoring the horse IgG constant region.
  • the monoclonal antibody therapeutic light chains are decoupled from the monoclonal antibody therapeutic heavy chains prior to the mass spectrometry step.
  • quantifying the monoclonal antibody therapeutic in the sample comprises measuring the variable region peptide of the monoclonal antibody therapeutic.
  • quantifying the monoclonal antibody therapeutic in the sample comprises subjecting the sample to a LC-MS/MS mass spectrometry technique.
  • the sample is a biological sample.
  • the biological sample is a whole blood sample, serum sample, saliva sample, plasma sample, or urine sample.
  • the biological sample can be a serum sample.
  • Also provided herein is a method of monitoring a treatment of a disorder in a subject, wherein the disorder is treated with a monoclonal antibody therapeutic, comprising:
  • the internal standard is a horse IgG
  • the monitoring a treatment of a disorder in a subject comprises quantifying a monoclonal antibody therapeutic in a sample.
  • the disorder is one that is treated with a humanized monoclonal antibody therapeutic. In some embodiments, the disorder is one that is treated with a human monoclonal antibody therapeutic. In some embodiments, the disorder is one that is treated with infliximab, alemtuzumab, eculizumab, rituxumab or adalimumab. For example, the disorder can be one that is treated with infliximab.
  • FIG. 1 provides liquid chromatograms with peaks for heavy and light chain labeled infliximab internal standard (top graph), and with horse IgG and infliximab heavy and light chain transitions (bottom graph).
  • FIG. 2 shows a standard curve of infliximab in commercial pooled serum using heavy chain transition HCy4.
  • FIG. 3 shows a comparison of infliximab concentrations calculated using LC- MS/MS and those obtained from an ELISA method.
  • FIG. 4 provides results for infliximab levels measured by SRM LC-MS/MS in patients receiving infliximab infusions.
  • Visit #1 was measured at trough levels; visit #2 at 48-72 hours after infliximab infusion; visit #3 at 28-32 days after infliximab infusion.
  • FIG. 5 shows liquid chromatograms of normal human serum after Ig Melon Gel purification and trypsin digest (top graph) and normal human serum spiked with 25 ⁇ g/mL alemtuzumab after Ig Melon Gel purification and trypsin digest (bottom graph).
  • FIG. 6 shows liquid chromatograms used in an SRM analysis for eculizumab.
  • Top graph shows a test sample.
  • Second graph shows a chromatogram of the horse IgGl standard.
  • Third graph shows a chromatogram of eculizumab heavy chain (HC).
  • Bottom graph shows a chromatogram of eculizumab light chain (LC).
  • FIG. 7 provides a linear regression graph used to quantify eculizumab in human serum using the area ratio of the light chain to the horse IgG constant region.
  • FIG. 8 provides a light chain standard curve for rituximab. Expected
  • Monoclonal antibody therapeutics is a rapidly growing class of drugs. This class of protein drug presents unique challenges when compared to small molecule drugs for quantitating levels in patients. Since the monoclonal antibody therapeutics have very similar molecular structure to normal background polyclonal antibodies in patients sera, most methods used for quantifying them rely on the interaction of the monoclonal antibody with its target antigen. Such methods, while useful, can be subject to cross reactivity with other antibodies present in the sera. This method directly quantitates the monoclonal antibody without the need for interaction with the antigen.
  • Figure 1 presents an example of how in silico prediction of peptide digestion patterns on a monoclonal antibody therapeutic would be correlated with an LC-MS/MS chromatogram.
  • a sample containing an antibody therapeutic would first be processed, for example, by enzymatic digestion, and then analyzed by mass spectrometry, for example, using LC-MS/MS.
  • the most abundant peptides unique to the heavy and light chain variable regions of the monoclonal antibody therapeutic would be chosen for monitoring.
  • the expected LC pattern in an LC-MS/MS would exhibit two peaks correlating to the peptides from the heavy and light chains of the monoclonal antibody therapeutic (top graph).
  • a non-human monoclonal antibody for example, horse IgG
  • horse IgG can also be added as an internal standard to samples containing the human monoclonal antibody therapeutic.
  • the intact horse immunoglobulin represents an improvement over commonly used isotopically labeled peptides since it is present in the purification and digestion step and normalizes for sample to sample differences in sample preparation.
  • Quantitation of the monoclonal antibody therapeutic may be accomplished using standard SRM analysis to generate a standard curve, for example, Figure 2, to be used to calculate concentrations in biological samples containing unknown amounts of the monoclonal antibody therapeutic.
  • a method for quantifying a monoclonal antibody therapeutic in a sample comprising: providing a sample comprising a monoclonal antibody therapeutic; adding an internal standard to the sample, wherein the internal standard is a non- isotopically labeled immunoglobulin from a non-human species; and subjecting the sample to a mass spectrometry technique to quantify the monoclonal antibody therapeutic in the sample.
  • the monoclonal antibody therapeutic is a humanized monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is a human monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituximab, adalimumab, and mixtures thereof. For example, the monoclonal antibody therapeutic can be infliximab.
  • the non-isotopically labeled immunoglobulin from a non- human species is a non-human IgG antibody. In some embodiments, the non-isotopically labeled immunoglobulin from a non-human species is a horse IgG. In some
  • the non-isotopically labeled immunoglobulin from a non-human species is a mouse IgG.
  • the immunoglobulins are purified from the sample before digestion using precipitation, protein A/G affinity chromatography, affinity resin purification, chemical fractionation (e.g., antibody purification kits, such as Melon Gel Purification).
  • the sample may be purified using affinity resin purification.
  • the method further comprises digesting one or more antibodies (e.g., the monoclonal antibody therapeutic and the internal standard) in the sample prior to the mass spectrometry step.
  • digesting the antibodies comprises digestion using trypsin.
  • the method further comprises purifying the sample prior to the mass spectrometry step.
  • the sample may be purified via centrifugation, filtration, ultrafiltration, dialysis, ion exchange chromatography, size exclusion chromatography, gel electrophoresis, or capillary electrophoresis.
  • the mass spectrometry technique comprises a tandem mass spectrometry (MS/MS) technique.
  • the mass spectrometry technique comprises an LC-MS/MS technique.
  • the mass spectrometry technique can be LC-ESI TRIPLE QUAD MS.
  • the mass spectrometry technique comprises the use of positive ion mode.
  • the mass spectrometry technique comprises selective reaction monitoring (SRM) analysis.
  • the SRM analysis comprises monitoring the horse IgG constant region.
  • the monoclonal antibody therapeutic light chains are decoupled from the monoclonal antibody therapeutic heavy chains prior to the mass spectrometry step.
  • quantifying the monoclonal antibody therapeutic in the sample comprises measuring the variable region peptide of the monoclonal antibody therapeutic. In some embodiments, quantifying the monoclonal antibody therapeutic in the sample comprises subjecting the sample to a LC-MS/MS mass spectrometry technique.
  • the sample is a biological sample.
  • the biological sample is a whole blood sample, serum sample, saliva sample, plasma sample, or urine sample.
  • the biological sample can be a serum sample.
  • Also provided herein is a method of monitoring a treatment of a disorder in a subject, wherein the disorder is treated with a monoclonal antibody therapeutic, comprising:
  • the monitoring a treatment of a disorder in a subject comprises quantifying a monoclonal antibody therapeutic in a sample.
  • the disorder is one that is treated with a humanized monoclonal antibody therapeutic. In some embodiments, the disorder is one that is treated with a human monoclonal antibody therapeutic. In some embodiments, the disorder is one that is treated with infliximab, alemtuzumab, eculizumab, rituximab, adalimumab, or a combination thereof. For example, the disorder can be one that is treated with infliximab.
  • a sample for analysis can be any biological sample, such as a tissue (e.g., adipose, liver, kidney, heart, muscle, bone, or skin tissue) or biological fluid (e.g., blood, serum, plasma, urine, lachrymal fluid, or saliva) sample.
  • the biological sample can be from a subject that has been treated with a human monoclonal antibody, which includes, but is not limited to, a mammal, e.g. a human, dog, cat, primate, rodent, pig, sheep, cow, and horse.
  • the biological sample comprises an exogenous monoclonal antibody.
  • a sample can also be a man-made reagent, such as a mixture of known composition or a control sample.
  • a sample can be treated to remove components that could interfere with the mass spectrometry technique.
  • a variety of techniques known to those having skill in the art can be used based on the sample type. Solid and/or tissue samples can be ground and extracted to free the analytes of interest from interfering components. In such cases, a sample can be centrifuged, filtered, and/or subjected to chromatographic techniques to remove interfering components (e.g., cells or tissue fragments). In yet other cases, reagents known to precipitate or bind the interfering components can be added. For example, whole blood samples can be treated using conventional clotting techniques to remove red and white blood cells and platelets.
  • Monoclonal antibodies can be isolated from the samples or enriched (i.e.
  • the samples can be enriched or purified using centrifugation, filtration, ultrafiltration, dialysis, ion exchange chromatography, size exclusion chromatography, protein A/G affinity chromatography, affinity purification, precipitation, gel electrophoresis, capillary electrophoresis, and chemical fractionation (e.g., antibody purification kits, such as Melon Gel Purification).
  • the monoclonal antibodies can be purified by chemical-based fractionation, e.g., Melon Gel
  • a sample such as a 10 - 250 ⁇ sample, e.g., a 50 ⁇ , can be directly subjected to Melon Gel, Protein A, Protein G, or Protein L purification.
  • Size exclusion principles such as a TurboFlow column can also be employed to separate the non-monoclonal antibody contaminants from a sample.
  • a urine sample can be buffered, e.g., a 50 ⁇ urine sample can be diluted first with 50 ⁇ of 50 mM ammonium bicarbonate.
  • the monoclonal antibodies, or the heavy and/or light chains thereof are substantially isolated.
  • substantially isolated is meant that the monoclonal antibodies are at least partially or substantially separated from the sample from which they were provided.
  • Partial separation can include, for example, a sample enriched in the monoclonal antibodies (i.e., the heavy and/or light chains).
  • Substantial separation can include samples containing at least about 10%, at least about 20%, at least about 30%), at least about 40%>, at least about 50%>, at least about 60%>, at least about 70%>, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the monoclonal antibody, or the heavy and/or light chains thereof.
  • Intact monoclonal antibodies can be further processed to decouple the light chains in a total monoclonal antibody sample from the heavy chains.
  • Decoupling can be achieved by treating the total monoclonal antibodies with a reducing agent, such as DTT (2,3 dihydroxybutane-l,4-dithiol), DTE (2,3 dihydroxybutane-l,4-dithiol), thioglycolate, cysteine, sulfites, bisulfites, sulfides, bisulfides, TCEP (tris(2-carboxyethyl)phosphine), 2-mercaptoethanol, and salt forms thereof.
  • a reducing agent such as DTT (2,3 dihydroxybutane-l,4-dithiol), DTE (2,3 dihydroxybutane-l,4-dithiol), thioglycolate, cysteine, sulfites, bisulfites, sulfides, bisulfides,
  • the reducing step is performed at elevated temperature, e.g., in a range from about 30 °C to about 65 °C, such as about 55 °C, in order to denature the proteins.
  • the sample is further treated, e.g., by modifying the pH of the sample or buffering the sample.
  • the sample can be acidified.
  • the sample can be neutralized (e.g., by the addition of a base such as bicarbonate).
  • a monoclonal antibody sample such as a trypsin digested immunoglobulin enriched serum
  • MS mass spectrometry
  • LC-MS/MS is an analytical technique that combines the physical separation capabilities of liquid chromatography with the mass analysis capabilities of mass spectrometry, and is suitable for detection and potential identification of chemicals in a complex mixture.
  • Any LC-MS instrument can be used, e.g., the ABSciex 5000 Mass Spectrometer (AB SCIEX, Framingham, MA, USA).
  • a reversed-phase CI 8 liquid chromatography HPLC column can be utilized.
  • Any suitable reversed-phase CI 8 liquid chromatography HPLC column can be used, e.g., the Atlantis T3 3x100mm.
  • the ion mass spectrum can be analyzed for one or more peaks corresponding to one or more heavy or light chain peptides in the sample. In some embodiments, the ratio is determined by the peak area of the selected ion peak(s).
  • electrospray ionization coupled to a quadrupole mass spectrometry can be used to analyze the mass spectrum of a monoclonal antibody sample.
  • a quadrupole mass analyzer (Q) consists of four cylindrical rods, set parallel to each other. In a quadrupole mass spectrometer, the quadrupole is the component of the instrument responsible for filtering sample ions based on their mass-to-charge ratio (m/z). Any ESI Triple Quad mass spectrometer can be used, e.g., the ABSciex API 5000 mass spectrometer.
  • the mass spectrometry based methods provided herein can also be used for monitoring the treatment of a disorder in a subject.
  • the disorder may be a disorder that is treated with a monoclonal antibody therapeutic.
  • the methods provided herein may be used to monitor the levels of monoclonal antibody therapeutic present in the subject during or after a treatment regimen. For example, in a treatment regimen that involves repeated monthly dosing of a monoclonal antibody therapeutic, the method may be used to quantitate the monoclonal antibody therapeutic at trough level prior to the subsequent dose. In some embodiments, information on the trough level may be used to determine whether the desired concentration for therapeutic treatment of the disorder is being reached, or whether the frequency or dosage of the therapy needs to be adjusted.
  • the method provided herein may monitor a disorder selected from rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease (including adult and pediatric Crohn's disease), plaque psoriasis, ulcerative colitis (including adult and pediatric ulcerative colitis), B-cell chronic lymphocytic leukemia, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), granulomatosis with polyangiitis (Wegener's granulomatosis), microscopic polyangiitis (MP A), CD-20 positive chronic lymphocytic leukemia, and non-Hodgkin lymphoma (including large B-cell and low-grade or follicular non-Hodgkin lymphoma).
  • the disorder can be rheumatoid arthritis.
  • the monoclonal antibody therapeutic is a human monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is selected from infliximab, alemtuzumab, eculizumab, rituximab,
  • the monoclonal antibody therapeutic can be infliximab.
  • Serum and Monoclonal antibody Reagents Serum was collected from waste samples obtained from the clinical laboratory.
  • Ammonium bicarbonate, dithiothreitol (DTT), and formic acid were purchased from Sigma- Aldrich (St. Louis, MO). Melon Gel was purchased from Thermo-Fisher Scientific (Waltham MA). Water, acetonitrile, and 2-propanol were purchased from Honeywell Burdick and Jackson (Muskegon, MI).
  • Serum A volume of 50 ⁇ of serum was enriched for monoclonal antibodies using Melon Gel following the manufacturer's instructions. After monoclonal antibody enrichment, 25 of sample was reduced by adding 25 ⁇ , of 100 mM DTT and 25 ⁇ , of 50 mM ammonium bicarbonate then incubated at 55°C for 15 minutes before injection. Samples were placed into 96 deep-well PCR plates (300 ⁇ , volume) at 9°C while waiting for injection.
  • ESI-Q-TOF MS Spectra were collected on an ABSciex Triple Quad 5000 mass spectrometer (ABSciex, Vaughan ON, CA) in ESI positive mode with a Turbo V dual ion source with an automated calibrant delivery system (CDS).
  • Source conditions were: IS: 5500, Temp: 500, CUR: 45, GS1 : 35, GS2: 30, CE: 50 ⁇ 5.
  • TOF MS scans were acquired from m/z 600-2500 with an acquisition time of 100 ms.
  • Fragment ion scans were acquired from m/z 350-2000 with an acquisition time of 100 ms.
  • the instrument was calibrated every 5 injections through the CDS using calibration solution supplied by the
  • Infliximab (RemicadeTM, Janssen Biotech, Inc.) was reconstituted to 10 mg/mL in 50 mM ammonium bicarbonate, reduced, alkylated and digested with trypsin (1 : 20 enzyme : substrate ratio) at 37°C for 4 hours.
  • infliximab Quantitation of infliximab was accomplished using standard SRM analysis on an ABSciex API 5000 using pooled human serum from healthy controls or 50 mM ammonium bicarbonate, each spiked with infliximab. A 9-point standard curve was generated [blank, 0.25, 0.5, 1, 2, 5, 10, 20 and 50 ⁇ g/mL].
  • FIG. 1 shows liquid chromatograms with peaks for heavy and light chain labeled infliximab internal standard (top graph), and with horse IgG and infliximab heavy and light chain transitions (bottom graph).
  • the LC-MS/MS method was applied to a series of patients who received infliximab treatments. The method was compared to a commercially available ELISA kit for infliximab (Immunodiagnostik, Germany). The resulting concentration versus time curves demonstrate the ability of the method to quantitate over time (Figure 3).
  • Figure 3 shows the infliximab concentration as measured by the LC-MS/MS method (y-axis; ⁇ g/mL) as compared with that measured using the ELISA method (x-axis; ⁇ g/mL).
  • the protocol was as performed in Example 1. Patients' immunoglobulins were purified by precipitation with 55 ⁇ ⁇ of a saturated ammonium sulfate solution prior to digestion and analysis.
  • FIG. 4 shows the change in infliximab concentration (y-axis; ⁇ / ⁇ .) over a series of three patient visits (x-axis) at trough level (visit #1), at 48-72 hours after infliximab infusion (visit #2), and at 28-32 days after infusion (visit #3).
  • a list of tryptic peptides unique to the heavy and light chain variable regions were predicted by in silico digestion of alemtuzumab variable region sequences found in the IMGT database (http://www.imgt.org/3Dstructure-DB).
  • Alemtuzumab (CampathTM, Genzyme, Inc.) was reconstituted to 10 mg/mL in 50 mM ammonium bicarbonate, reduced, alkylated and digested with trypsin (1 :20 enzyme: substrate ratio) at 37°C for 4 hours.
  • Digests were analyzed by IDA LC-ESI-Q-TOF MS. The most abundant peptides matching the in silico list were chosen for subsequent studies.
  • Alemtuzumab was then spiked into normal human serum at a 25 ⁇ g/mL level. A dilution series was made from the 25 ⁇ g stock. Samples were purified using Melon Gel and digested in a similar manner to Example 1. Identified alemtuzumab tryptic peptides were present in the all spiked samples and no significant amount of the peptides was observed in the normal pooled sera (Figure 5).
  • Eculizumab was reconstituted to 10 mg/mL in 50 mM ammonium bicarbonate, reduced, alkylated and digested with trypsin (1 :20
  • Immunoglobulins were isolated by precipitation using a saturated ammonium sulfate solution, followed by trypsin digestion. Peptides were separated on reverse-phase
  • Human serum was spiked with 50 ⁇ g of infliximab and the concentration was measured using the method of Example 1. Twenty replicates of the same sample were measured using and the data was either normalized to isotopically labeled peptides internal standards or to a peptide from the constant region of the horse immunoglobulin. The coefficient of variation (CV) for each method was determined for each analysis. The CV's of quantitation using the horse constant region was significantly lower (12% versus 34%) for the heavy chain peptide and 8% versus 22% for the light chain peptide) as demonstrated in Table 1.
  • Rituximab was then spiked into normal human serum at 250 mg/ml. A dilution series was made from the stock. Samples were purified using by precipitation in a saturated ammonium sulfate solution and digested according to Example 1. Identified rituximab tryptic peptides were present in the all spiked samples and no appreciable quantities of the peptides were seen in the normal pooled sera (c.f. Figure 5). The resulting rituximab light chain standard curve is shown in Figure 8. Expected concentration (y-axis; ⁇ g/mL) correlated with measured concentration (x-axis; ⁇ g/mL).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Biophysics (AREA)
  • Endocrinology (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Other Investigation Or Analysis Of Materials By Electrical Means (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

This document relates to methods for quantifying antibody therapeutics using mass spectrometry techniques by providing a sample comprising a monoclonal antibody therapeutic, adding an internal standard to the sample, wherein the internal standard is a nonisotopically labeled immunoglobulin from a non-human species, and subjecting the sample to a mass spectrometry technique to quantify the monoclonal antibody therapeutic in the sample.

Description

Quantifying Monoclonal Antibody Therapeutics by LC-MS/MS
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Serial No. 62/030,493 filed July 29, 2014. The disclosure of the prior application is considered part of (and is incorporated by reference in) the disclosure of this application.
TECHNICAL FIELD
This disclosure relates to methods for quantifying monoclonal antibody therapeutics using mass spectrometry techniques.
BACKGROUND
Human monoclonal antibodies contain two identical heavy chain polypeptides (each about 54 kilodaltons in MW) and two identical light chain polypeptides (each about 24 kilodaltons in molecular weight) which are bound together by disulfide bonds. Each light chain and each heavy chain include a constant region and a variable region. The variable region is located on the N-terminal portion of each chain and the constant region is located on the C-terminal portion of each chain. The constant regions of the light chains and heavy chains have different amino acid sequences, and can be used to identify the isotype of the heavy or light chain. In humans, there are two different isotypes of light chain polypeptides referred to as either kappa or lambda; and five different isotypes of heavy chain polypeptides referred to as gamma (IgG), alpha (IgA), mu (IgM), epsilon (IgE), and delta (IgD).
Clinical laboratories currently monitor and quantify the levels of monoclonal antibody therapeutics in patients receiving such treatment using immunoassays, such as ELISA.
SUMMARY
Provided herein is a method for quantifying a monoclonal antibody therapeutic in a sample, comprising: providing a sample comprising a monoclonal antibody therapeutic; adding an internal standard to the sample, wherein the internal standard is a non- isotopically labeled immunoglobulin from a non-human species; and subjecting the sample to a mass spectrometry technique to quantify the monoclonal antibody therapeutic in the sample.
In some embodiments, the monoclonal antibody therapeutic is a humanized monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is a human monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituximab and adalimumab. For example, the monoclonal antibody therapeutic can be infliximab.
In some embodiments, the non-isotopically labeled immunoglobulin from a non- human species is a non-human IgG antibody. In some embodiments, the non-isotopically labeled immunoglobulin from a non-human species is a horse IgG.
In some embodiments, the method further comprises digesting the antibodies in the sample prior to the mass spectrometry step. In some embodiments, digesting the antibodies comprises digestion using trypsin.
In some embodiments, the method further comprises purifying the sample prior to the mass spectrometry step. For example, the sample may be purified using antibody affinity resin purification.
In some embodiments, the mass spectrometry technique comprises a tandem mass spectrometry (MS/MS) technique. In some embodiments, the mass spectrometry technique comprises an LC-MS/MS technique. For example, the mass spectrometry technique can be a LC-ESI TRIPLE QUAD MS. In some embodiments, the mass spectrometry technique comprises the use of positive ion mode. In some embodiments, the mass spectrometry technique comprises selective reaction monitoring (SRM) analysis. In some embodiments, the SRM analysis comprises monitoring the horse IgG constant region.
In some embodiments, the monoclonal antibody therapeutic light chains are decoupled from the monoclonal antibody therapeutic heavy chains prior to the mass spectrometry step. In some embodiments, quantifying the monoclonal antibody therapeutic in the sample comprises measuring the variable region peptide of the monoclonal antibody therapeutic. In some embodiments, quantifying the monoclonal antibody therapeutic in the sample comprises subjecting the sample to a LC-MS/MS mass spectrometry technique.
In some embodiments, the sample is a biological sample. In some embodiments, the biological sample is a whole blood sample, serum sample, saliva sample, plasma sample, or urine sample. For example, the biological sample can be a serum sample.
Also provided herein is a method of monitoring a treatment of a disorder in a subject, wherein the disorder is treated with a monoclonal antibody therapeutic, comprising:
a) providing a first sample of the subject during treatment;
b) providing a second sample of the subject on a subsequent treatment or after treatment;
c) subjecting the first and second sample to a mass spectrometry technique
comprising an internal standard to obtain a mass spectrum of the sample, wherein the internal standard is a horse IgG;
d) quantifying the monoclonal antibody therapeutic in the first and second sample; and
e) comparing the amounts from the first and second sample.
In some embodiments, the monitoring a treatment of a disorder in a subject comprises quantifying a monoclonal antibody therapeutic in a sample.
In some embodiments, the disorder is one that is treated with a humanized monoclonal antibody therapeutic. In some embodiments, the disorder is one that is treated with a human monoclonal antibody therapeutic. In some embodiments, the disorder is one that is treated with infliximab, alemtuzumab, eculizumab, rituxumab or adalimumab. For example, the disorder can be one that is treated with infliximab.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this description belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
DESCRIPTION OF DRAWINGS
FIG. 1 provides liquid chromatograms with peaks for heavy and light chain labeled infliximab internal standard (top graph), and with horse IgG and infliximab heavy and light chain transitions (bottom graph).
FIG. 2 shows a standard curve of infliximab in commercial pooled serum using heavy chain transition HCy4.
FIG. 3 shows a comparison of infliximab concentrations calculated using LC- MS/MS and those obtained from an ELISA method.
FIG. 4 provides results for infliximab levels measured by SRM LC-MS/MS in patients receiving infliximab infusions. Visit #1 was measured at trough levels; visit #2 at 48-72 hours after infliximab infusion; visit #3 at 28-32 days after infliximab infusion.
FIG. 5 shows liquid chromatograms of normal human serum after Ig Melon Gel purification and trypsin digest (top graph) and normal human serum spiked with 25 μg/mL alemtuzumab after Ig Melon Gel purification and trypsin digest (bottom graph).
FIG. 6 shows liquid chromatograms used in an SRM analysis for eculizumab. Top graph shows a test sample. Second graph shows a chromatogram of the horse IgGl standard. Third graph shows a chromatogram of eculizumab heavy chain (HC). Bottom graph shows a chromatogram of eculizumab light chain (LC).
FIG. 7 provides a linear regression graph used to quantify eculizumab in human serum using the area ratio of the light chain to the horse IgG constant region.
FIG. 8 provides a light chain standard curve for rituximab. Expected
concentration ^g/mL) is shown on the y-axis; measured concentration ^g/mL) is shown on the x-axis. DETAILED DESCRIPTION
Monoclonal antibody therapeutics is a rapidly growing class of drugs. This class of protein drug presents unique challenges when compared to small molecule drugs for quantitating levels in patients. Since the monoclonal antibody therapeutics have very similar molecular structure to normal background polyclonal antibodies in patients sera, most methods used for quantifying them rely on the interaction of the monoclonal antibody with its target antigen. Such methods, while useful, can be subject to cross reactivity with other antibodies present in the sera. This method directly quantitates the monoclonal antibody without the need for interaction with the antigen. Provided herein are methods of quantifying monoclonal antibody therapeutics that include monitoring tryptic peptides from the unique variable regions of the monoclonal antibody and comparing them to the constant regions of a non-human antibody added as an internal standard. This method offers advantages compared to other methods as the addition of the non-human antibody as internal standard affords enhanced accuracy. For example, not only does the standard have a similar, yet distinct, mass profile, but it is also similarly reactive through all preparatory steps including, for example, enzymatic digestion and purification.
Figure 1 presents an example of how in silico prediction of peptide digestion patterns on a monoclonal antibody therapeutic would be correlated with an LC-MS/MS chromatogram. A sample containing an antibody therapeutic would first be processed, for example, by enzymatic digestion, and then analyzed by mass spectrometry, for example, using LC-MS/MS. The most abundant peptides unique to the heavy and light chain variable regions of the monoclonal antibody therapeutic would be chosen for monitoring. The expected LC pattern in an LC-MS/MS would exhibit two peaks correlating to the peptides from the heavy and light chains of the monoclonal antibody therapeutic (top graph). Known amounts of a non-human monoclonal antibody, for example, horse IgG, can also be added as an internal standard to samples containing the human monoclonal antibody therapeutic. The intact horse immunoglobulin represents an improvement over commonly used isotopically labeled peptides since it is present in the purification and digestion step and normalizes for sample to sample differences in sample preparation. Quantitation of the monoclonal antibody therapeutic may be accomplished using standard SRM analysis to generate a standard curve, for example, Figure 2, to be used to calculate concentrations in biological samples containing unknown amounts of the monoclonal antibody therapeutic.
Provided herein is a method for quantifying a monoclonal antibody therapeutic in a sample, comprising: providing a sample comprising a monoclonal antibody therapeutic; adding an internal standard to the sample, wherein the internal standard is a non- isotopically labeled immunoglobulin from a non-human species; and subjecting the sample to a mass spectrometry technique to quantify the monoclonal antibody therapeutic in the sample.
In some embodiments, the monoclonal antibody therapeutic is a humanized monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is a human monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituximab, adalimumab, and mixtures thereof. For example, the monoclonal antibody therapeutic can be infliximab.
In some embodiments, the non-isotopically labeled immunoglobulin from a non- human species is a non-human IgG antibody. In some embodiments, the non-isotopically labeled immunoglobulin from a non-human species is a horse IgG. In some
embodiments, the non-isotopically labeled immunoglobulin from a non-human species is a mouse IgG.
In some embodiments, the immunoglobulins are purified from the sample before digestion using precipitation, protein A/G affinity chromatography, affinity resin purification, chemical fractionation (e.g., antibody purification kits, such as Melon Gel Purification). For example, the sample may be purified using affinity resin purification.
In some embodiments, the method further comprises digesting one or more antibodies (e.g., the monoclonal antibody therapeutic and the internal standard) in the sample prior to the mass spectrometry step. In some embodiments, digesting the antibodies comprises digestion using trypsin. In some embodiments, the method further comprises purifying the sample prior to the mass spectrometry step. In some embodiments, the sample may be purified via centrifugation, filtration, ultrafiltration, dialysis, ion exchange chromatography, size exclusion chromatography, gel electrophoresis, or capillary electrophoresis.
In some embodiments, the mass spectrometry technique comprises a tandem mass spectrometry (MS/MS) technique. In some embodiments, the mass spectrometry technique comprises an LC-MS/MS technique. For example, the mass spectrometry technique can be LC-ESI TRIPLE QUAD MS. In some embodiments, the mass spectrometry technique comprises the use of positive ion mode. In some embodiments, the mass spectrometry technique comprises selective reaction monitoring (SRM) analysis. In some embodiments, the SRM analysis comprises monitoring the horse IgG constant region.
In some embodiments, the monoclonal antibody therapeutic light chains are decoupled from the monoclonal antibody therapeutic heavy chains prior to the mass spectrometry step.
In some embodiments, quantifying the monoclonal antibody therapeutic in the sample comprises measuring the variable region peptide of the monoclonal antibody therapeutic. In some embodiments, quantifying the monoclonal antibody therapeutic in the sample comprises subjecting the sample to a LC-MS/MS mass spectrometry technique.
In some embodiments, the sample is a biological sample. In some embodiments, the biological sample is a whole blood sample, serum sample, saliva sample, plasma sample, or urine sample. For example, the biological sample can be a serum sample.
Also provided herein is a method of monitoring a treatment of a disorder in a subject, wherein the disorder is treated with a monoclonal antibody therapeutic, comprising:
a) providing a first sample of the subject during treatment;
b) providing a second sample of the subject on a subsequent treatment or after treatment; c) subjecting the first and second sample to a mass spectrometry technique comprising an internal standard to obtain a mass spectrum of the sample, wherein the internal standard is a horse IgG;
d) quantifying the monoclonal antibody therapeutic in the first and second sample; and
e) comparing the amounts from the first and second sample.
In some embodiments, the monitoring a treatment of a disorder in a subject comprises quantifying a monoclonal antibody therapeutic in a sample.
In some embodiments, the disorder is one that is treated with a humanized monoclonal antibody therapeutic. In some embodiments, the disorder is one that is treated with a human monoclonal antibody therapeutic. In some embodiments, the disorder is one that is treated with infliximab, alemtuzumab, eculizumab, rituximab, adalimumab, or a combination thereof. For example, the disorder can be one that is treated with infliximab.
Samples and Sample Preparation
A sample for analysis can be any biological sample, such as a tissue (e.g., adipose, liver, kidney, heart, muscle, bone, or skin tissue) or biological fluid (e.g., blood, serum, plasma, urine, lachrymal fluid, or saliva) sample. The biological sample can be from a subject that has been treated with a human monoclonal antibody, which includes, but is not limited to, a mammal, e.g. a human, dog, cat, primate, rodent, pig, sheep, cow, and horse. In some embodiments the biological sample comprises an exogenous monoclonal antibody. A sample can also be a man-made reagent, such as a mixture of known composition or a control sample.
A sample can be treated to remove components that could interfere with the mass spectrometry technique. A variety of techniques known to those having skill in the art can be used based on the sample type. Solid and/or tissue samples can be ground and extracted to free the analytes of interest from interfering components. In such cases, a sample can be centrifuged, filtered, and/or subjected to chromatographic techniques to remove interfering components (e.g., cells or tissue fragments). In yet other cases, reagents known to precipitate or bind the interfering components can be added. For example, whole blood samples can be treated using conventional clotting techniques to remove red and white blood cells and platelets.
Monoclonal antibodies can be isolated from the samples or enriched (i.e.
concentrated) in a sample using standard methods known in the art. Such methods include removing one or more non-monoclonal antibody contaminants from a sample. In some embodiments, the samples can be enriched or purified using centrifugation, filtration, ultrafiltration, dialysis, ion exchange chromatography, size exclusion chromatography, protein A/G affinity chromatography, affinity purification, precipitation, gel electrophoresis, capillary electrophoresis, and chemical fractionation (e.g., antibody purification kits, such as Melon Gel Purification). For example, the monoclonal antibodies can be purified by chemical-based fractionation, e.g., Melon Gel
Chromatography (Thermo Scientific), where Melon Gel resins bind to non-monoclonal antibody proteins in a sample and allow monoclonal antibodies to be collected in the flow-through fraction; or by affinity purification, e.g., by Protein A, Protein G, or Protein L purification, where monoclonal antibodies are bound by those proteins at physiologic pH and then released from the proteins by lowering the pH. When serum, plasma, or whole blood samples are used, a sample, such as a 10 - 250 μΐ sample, e.g., a 50 μΐ, can be directly subjected to Melon Gel, Protein A, Protein G, or Protein L purification. Size exclusion principles such as a TurboFlow column can also be employed to separate the non-monoclonal antibody contaminants from a sample. When urine samples are used, a urine sample can be buffered, e.g., a 50 μΐ urine sample can be diluted first with 50 μΐ of 50 mM ammonium bicarbonate.
In some embodiments, the monoclonal antibodies, or the heavy and/or light chains thereof, are substantially isolated. By "substantially isolated" is meant that the monoclonal antibodies are at least partially or substantially separated from the sample from which they were provided. Partial separation can include, for example, a sample enriched in the monoclonal antibodies (i.e., the heavy and/or light chains). Substantial separation can include samples containing at least about 10%, at least about 20%, at least about 30%), at least about 40%>, at least about 50%>, at least about 60%>, at least about 70%>, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the monoclonal antibody, or the heavy and/or light chains thereof. Methods for isolating monoclonal antibodies, such as those described above, are routine in the art.
Intact monoclonal antibodies can be further processed to decouple the light chains in a total monoclonal antibody sample from the heavy chains. Decoupling can be achieved by treating the total monoclonal antibodies with a reducing agent, such as DTT (2,3 dihydroxybutane-l,4-dithiol), DTE (2,3 dihydroxybutane-l,4-dithiol), thioglycolate, cysteine, sulfites, bisulfites, sulfides, bisulfides, TCEP (tris(2-carboxyethyl)phosphine), 2-mercaptoethanol, and salt forms thereof. In some embodiments, the reducing step is performed at elevated temperature, e.g., in a range from about 30 °C to about 65 °C, such as about 55 °C, in order to denature the proteins. In some embodiments, the sample is further treated, e.g., by modifying the pH of the sample or buffering the sample. In some embodiments, the sample can be acidified. In some embodiments, the sample can be neutralized (e.g., by the addition of a base such as bicarbonate).
Mass Spectrometry Methods
After sample preparation, a monoclonal antibody sample, such as a trypsin digested immunoglobulin enriched serum, can be subjected to a mass spectrometry (MS) technique, either directly or after separation on a high performance liquid
chromatography column (HPLC). LC-MS/MS is an analytical technique that combines the physical separation capabilities of liquid chromatography with the mass analysis capabilities of mass spectrometry, and is suitable for detection and potential identification of chemicals in a complex mixture. Any LC-MS instrument can be used, e.g., the ABSciex 5000 Mass Spectrometer (AB SCIEX, Framingham, MA, USA). In some embodiments, a reversed-phase CI 8 liquid chromatography HPLC column can be utilized. Any suitable reversed-phase CI 8 liquid chromatography HPLC column can be used, e.g., the Atlantis T3 3x100mm. The ion mass spectrum can be analyzed for one or more peaks corresponding to one or more heavy or light chain peptides in the sample. In some embodiments, the ratio is determined by the peak area of the selected ion peak(s). In some embodiments, electrospray ionization coupled to a quadrupole mass spectrometry (ESI Triple Quad MS) can be used to analyze the mass spectrum of a monoclonal antibody sample. A quadrupole mass analyzer (Q) consists of four cylindrical rods, set parallel to each other. In a quadrupole mass spectrometer, the quadrupole is the component of the instrument responsible for filtering sample ions based on their mass-to-charge ratio (m/z). Any ESI Triple Quad mass spectrometer can be used, e.g., the ABSciex API 5000 mass spectrometer.
Methods for screening biological samples and for diagnosing and monitoring disorders
The mass spectrometry based methods provided herein can also be used for monitoring the treatment of a disorder in a subject. The disorder may be a disorder that is treated with a monoclonal antibody therapeutic. In such cases, the methods provided herein may be used to monitor the levels of monoclonal antibody therapeutic present in the subject during or after a treatment regimen. For example, in a treatment regimen that involves repeated monthly dosing of a monoclonal antibody therapeutic, the method may be used to quantitate the monoclonal antibody therapeutic at trough level prior to the subsequent dose. In some embodiments, information on the trough level may be used to determine whether the desired concentration for therapeutic treatment of the disorder is being reached, or whether the frequency or dosage of the therapy needs to be adjusted.
In some embodiments, the method provided herein may monitor a disorder selected from rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease (including adult and pediatric Crohn's disease), plaque psoriasis, ulcerative colitis (including adult and pediatric ulcerative colitis), B-cell chronic lymphocytic leukemia, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), granulomatosis with polyangiitis (Wegener's granulomatosis), microscopic polyangiitis (MP A), CD-20 positive chronic lymphocytic leukemia, and non-Hodgkin lymphoma (including large B-cell and low-grade or follicular non-Hodgkin lymphoma). For example, the disorder can be rheumatoid arthritis.
In some embodiments, the monoclonal antibody therapeutic is a human monoclonal antibody therapeutic. In some embodiments, the monoclonal antibody therapeutic is selected from infliximab, alemtuzumab, eculizumab, rituximab,
adalimumab, or any combination thereof. For example, the monoclonal antibody therapeutic can be infliximab.
EXAMPLES
General Methods.
Serum and Monoclonal antibody Reagents: Serum was collected from waste samples obtained from the clinical laboratory.
Reagents: Ammonium bicarbonate, dithiothreitol (DTT), and formic acid were purchased from Sigma- Aldrich (St. Louis, MO). Melon Gel was purchased from Thermo-Fisher Scientific (Waltham MA). Water, acetonitrile, and 2-propanol were purchased from Honeywell Burdick and Jackson (Muskegon, MI).
Serum: A volume of 50 μί of serum was enriched for monoclonal antibodies using Melon Gel following the manufacturer's instructions. After monoclonal antibody enrichment, 25 of sample was reduced by adding 25 μΐ, of 100 mM DTT and 25 μΐ, of 50 mM ammonium bicarbonate then incubated at 55°C for 15 minutes before injection. Samples were placed into 96 deep-well PCR plates (300 μΐ, volume) at 9°C while waiting for injection. ESI-Q-TOF MS: Spectra were collected on an ABSciex Triple Quad 5000 mass spectrometer (ABSciex, Vaughan ON, CA) in ESI positive mode with a Turbo V dual ion source with an automated calibrant delivery system (CDS). Source conditions were: IS: 5500, Temp: 500, CUR: 45, GS1 : 35, GS2: 30, CE: 50±5. TOF MS scans were acquired from m/z 600-2500 with an acquisition time of 100 ms. Fragment ion scans were acquired from m/z 350-2000 with an acquisition time of 100 ms. The instrument was calibrated every 5 injections through the CDS using calibration solution supplied by the
manufacturer.
MS Data Analysis: Analyst TF vl .6 was used for instrument control. Data were viewed using Analyst TF vl .6 and Peak View vl .2.0.3. Peptides were separated on reverse-phase C18 liquid chromatography (Atlantis T3 3x100 mm) and subjected to MS/MS using an ABSciex 5000 triple quad MS instrument. Example 1 - Infliximab
Preparation of sample
A list of tryptic peptides unique to the heavy and light chain variable regions were predicted by in silico digestion of infliximab variable region sequences found in the
IMGT database (http://www.imgt.org/3Dstructure-DB). Infliximab (Remicade™, Janssen Biotech, Inc.) was reconstituted to 10 mg/mL in 50 mM ammonium bicarbonate, reduced, alkylated and digested with trypsin (1 : 20 enzyme : substrate ratio) at 37°C for 4 hours.
Digests were analyzed by IDA LC-ESI TRIPLE QUADMS. The most abundant peptides matching the in silico list were chosen for subsequent studies.
Quantitation of infliximab was accomplished using standard SRM analysis on an ABSciex API 5000 using pooled human serum from healthy controls or 50 mM ammonium bicarbonate, each spiked with infliximab. A 9-point standard curve was generated [blank, 0.25, 0.5, 1, 2, 5, 10, 20 and 50 μg/mL].
A known concentration of purified horse IgG (200 ug/mL) with a unique non- human constant region peptide was added to each sample as a pre-analytical digestion control along with stable isotope-labeled peptide internal standards to monitor HPLC retention times.
LC-MS/MS Analysis
Samples were processed to remove non-immunoglobulin proteins using the Melon Gel purification kit (Pierce, Rockford, IL), followed by trypsin digestion. Peptides were separated on reverse-phase CI 8 liquid chromatography (Atlantis T3 3x100 mm) and subjected to MS/MS. A typical chromatogram for the SRM analysis monitoring for the peptides for horse IgG constant region, and the heavy chain and light chain peptide from infliximab are shown in Figure 1. Figure 1 shows liquid chromatograms with peaks for heavy and light chain labeled infliximab internal standard (top graph), and with horse IgG and infliximab heavy and light chain transitions (bottom graph).
Correlation of Experimental Data to Expected Theoretical Results
The ability to accurately quantitate the level of infliximab in patient sera was demonstrated by spiking known amounts of infliximab into patient serum and plotting the heavy chain peptide response as a function of drug level. The results of the normal pooled serum spiked at various levels of infliximab demonstrate that the peak areas are linearly correlated with infliximab levels (Figure 2). Figure 2 shows the measured and calculated infliximab concentration (y-axis; μg/mL) in comparison to the theoretical infliximab concentration (x-axis; μg/mL). The standard curve was generated with infliximab at 0 to 100 μg/mL concentrations in commercial pooled human serum using heavy chain transition HCy4.
Correlation ofLC-MS/MS Concentrations to Other Methods
The LC-MS/MS method was applied to a series of patients who received infliximab treatments. The method was compared to a commercially available ELISA kit for infliximab (Immunodiagnostik, Germany). The resulting concentration versus time curves demonstrate the ability of the method to quantitate over time (Figure 3). Figure 3 shows the infliximab concentration as measured by the LC-MS/MS method (y-axis; μg/mL) as compared with that measured using the ELISA method (x-axis; μg/mL). The results demonstrate the LC-MS/MC method correlates to the ELISA method with a linear regression of R2 = 0.82.
Example 2 - Infliximab in Patients
The protocol was as performed in Example 1. Patients' immunoglobulins were purified by precipitation with 55 μΐ^ of a saturated ammonium sulfate solution prior to digestion and analysis.
The LC-MS/MS method was used to monitor consenting patients who were receiving infliximab therapy. The results confirmed that this method may be useful to monitor concentrations of infliximab in patients. Figure 4 shows the change in infliximab concentration (y-axis; μ§/ηιΙ.) over a series of three patient visits (x-axis) at trough level (visit #1), at 48-72 hours after infliximab infusion (visit #2), and at 28-32 days after infusion (visit #3).
Example 3 - Alemtuzumab
A list of tryptic peptides unique to the heavy and light chain variable regions were predicted by in silico digestion of alemtuzumab variable region sequences found in the IMGT database (http://www.imgt.org/3Dstructure-DB). Alemtuzumab (Campath™, Genzyme, Inc.) was reconstituted to 10 mg/mL in 50 mM ammonium bicarbonate, reduced, alkylated and digested with trypsin (1 :20 enzyme: substrate ratio) at 37°C for 4 hours. Digests were analyzed by IDA LC-ESI-Q-TOF MS. The most abundant peptides matching the in silico list were chosen for subsequent studies.
Alemtuzumab was then spiked into normal human serum at a 25 μg/mL level. A dilution series was made from the 25 μg stock. Samples were purified using Melon Gel and digested in a similar manner to Example 1. Identified alemtuzumab tryptic peptides were present in the all spiked samples and no significant amount of the peptides was observed in the normal pooled sera (Figure 5).
Example 4 - Eculizumab
A list of tryptic peptides unique to the heavy and light chain variable regions were predicted by in silico digestion of eculizumab variable region sequences provided by the manufacturer (Alexion). Eculizumab was reconstituted to 10 mg/mL in 50 mM ammonium bicarbonate, reduced, alkylated and digested with trypsin (1 :20
enzyme: substrate) at 37oC for 4 hours. Digests were analyzed by IDA LC-ESI-Q- TOFMS; the most abundant peptides matching the in silico list was chosen for subsequent studies.
Quantitation of eculizumab was accomplished using standard SRM analysis on as described in Example 1 using the horse IgG as an internal digestion standard.
Immunoglobulins were isolated by precipitation using a saturated ammonium sulfate solution, followed by trypsin digestion. Peptides were separated on reverse-phase
C18 liquid chromatography (Atlantis T3 3x100 mm) and subjected to MS/MS. Exemplary chromatograms for the SRM analysis monitoring for the peptides for horse IgG constant region, and the heavy chain and light chain peptide from eculizumab are shown in Figure 6. The ability to accurately quantitate the level of infliximab in patient sera was demonstrated by spiking known amounts of infliximab into patient serum and plotting the heavy chain peptide response as a function of drug level (Figure 7).
Example 5 - Comparison with and without horse IgG
Human serum was spiked with 50 μg of infliximab and the concentration was measured using the method of Example 1. Twenty replicates of the same sample were measured using and the data was either normalized to isotopically labeled peptides internal standards or to a peptide from the constant region of the horse immunoglobulin. The coefficient of variation (CV) for each method was determined for each analysis. The CV's of quantitation using the horse constant region was significantly lower (12% versus 34%) for the heavy chain peptide and 8% versus 22% for the light chain peptide) as demonstrated in Table 1.
Example 6 - Rituximab
A list of tryptic peptides unique to the heavy and light chain variable regions were predicted by in silico digestion of Rituximab variable region sequences found in the IMGT database (http://www.imgt.org/3Dstructure-DB). Rituximab (Rituxan, Genentech) was reconstituted to 10 mg/mL in 50 mM ammonium bicarbonate, reduced, alkylated and digested with trypsin (1 :20 enzyme: substrate) at 37°C for 4 hours. Digests were analyzed by IDA LC-ESI-Q-TOFMS; the most abundant peptides matching the in silico list were chosen for subsequent studies. A set of tryptic peptides for the rat heavy chain and light chain were identified.
Rituximab was then spiked into normal human serum at 250 mg/ml. A dilution series was made from the stock. Samples were purified using by precipitation in a saturated ammonium sulfate solution and digested according to Example 1. Identified rituximab tryptic peptides were present in the all spiked samples and no appreciable quantities of the peptides were seen in the normal pooled sera (c.f. Figure 5). The resulting rituximab light chain standard curve is shown in Figure 8. Expected concentration (y-axis; μg/mL) correlated with measured concentration (x-axis; μg/mL).
Table 1. Comparison of the intra-assay CV of a human serum spiked with infliximab using isotopically labeled peptides versus the horse IgG
Figure imgf000019_0001
OTHER EMBODIMENTS
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method for quantifying a monoclonal antibody therapeutic in a sample, the method comprising:
a) providing a sample comprising a monoclonal antibody therapeutic;
b) adding an internal standard to the sample, wherein the internal standard is a non- isotopically labeled immunoglobulin from a non-human species; and
c) subjecting the sample to a mass spectrometry technique to quantify the monoclonal antibody therapeutic in the sample.
2. The method of claim 1, wherein the method further comprises digesting the
antibodies in the sample prior to step (c).
3. The method of any one of claims 1-2, wherein the sample is purified prior to step (c).
4. The method of any one of claims 1-3, wherein the sample is purified using antibody affinity resin purification.
5. The method of any one of claims 1-4, wherein the wherein the mass spectrometry technique comprises a tandem mass spectrometry (MS/MS) technique.
6. The method of any one of claims 1-5, wherein the mass spectrometry technique
comprises an LC-MS/MS technique.
7. A method for quantifying a monoclonal antibody therapeutic in a sample, the method comprising:
a) providing a sample comprising a monoclonal antibody therapeutic;
b) adding an internal standard to the sample, wherein the internal standard is a non- human IgG antibody; and c) subjecting the sample to a LC-MS/MS mass spectrometry technique comprising monitoring the non-human IgG antibody constant region to quantify the monoclonal antibody therapeutic in the sample.
8. The method of claim 7, wherein the method further comprises digesting the
antibodies in the sample prior to step (c).
9. The method of claim 8, wherein the non-human IgG antibody is a horse IgG.
10. The method of any one of claims 1-9, wherein the mass spectrometry technique comprises a liquid chromatography electrospray ionization coupled to a quadrupole mass spectrometry (ESI Triple Quad MS) technique.
11. The method of any one of claims 1-10, wherein the mass spectrometry technique comprises the use of positive ion mode.
12. The method of any one of claims 1-11, wherein the mass spectrometry technique comprises selective reaction monitoring (SRM) analysis.
13. The method of any one of claims 1-12, wherein the SRM analysis comprises
monitoring the horse IgG constant region.
14. The method of any one of claims 1-13, wherein the monoclonal antibody therapeutic light chains are decoupled from the monoclonal antibody therapeutic heavy chains prior to step (c).
15. The method of any one of claims 1-14, wherein quantifying the monoclonal antibody therapeutic in the sample comprises measuring the variable region peptide of the monoclonal antibody therapeutic.
16. The method of any one of claims 1-15, wherein the sample is a biological sample.
17. The method of claim 16, wherein the biological sample is a whole blood sample, serum sample, saliva sample, plasma sample, or urine sample.
18. The method of claim 16 or 17, wherein the biological sample is a serum sample.
19. A method of monitoring a treatment of a disorder in a subject, wherein the disorder is treated with a monoclonal antibody therapeutic, the method comprising:
a) providing a first sample of the subject during treatment;
b) providing a second sample of the subject on a subsequent treatment or after treatment;
c) subjecting the first and second sample to a mass spectrometry technique
comprising an internal standard to obtain a mass spectrum of the sample, wherein the internal standard is a horse IgG;
d) quantifying the human monoclonal antibody therapeutic in the first and second sample; and
e) comparing the amounts from the first and second sample.
20. The method of any one of claims 1-19, wherein the monoclonal antibody therapeutic is selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituximab and adalimumab.
21. The method of any one of claims 1-20, wherein the monoclonal antibody therapeutic is infliximab.
22. A method for quantifying a monoclonal antibody therapeutic in a human serum
sample, the method comprising: a) providing a human serum sample comprising a monoclonal antibody therapeutic selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituxumab and adalimumab;
b) adding an internal standard to the sample, wherein the internal standard is a horse IgG;
c) digesting the antibodies, wherein the digesting comprises trypsin digestion; and d) subjecting the sample to a LC-MS/MS mass spectrometry technique to quantify the monoclonal antibody therapeutic in the sample.
23. A method for quantifying a monoclonal antibody therapeutic in a human serum
sample, the method comprising:
a) providing a human serum sample comprising a monoclonal antibody therapeutic selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituxumab and adalimumab;
b) adding an internal standard to the sample, wherein the internal standard is a horse IgG;
c) purifying the sample;
d) digesting the antibodies present, wherein the digesting comprises trypsin digestion; and
e) subjecting the sample to a LC-MS/MS mass spectrometry technique to quantify the human monoclonal antibody therapeutic in the sample.
24. A method for quantifying a monoclonal antibody therapeutic in a human serum
sample, the method comprising:
a) providing a human serum sample comprising a monoclonal antibody therapeutic selected from the group consisting of: infliximab, alemtuzumab, and adalimumab; b) adding an internal standard to the sample, wherein the internal standard is a horse IgG;
c) purifying the sample; d) digesting the antibodies present, wherein the digesting comprises trypsin digestion; and
e) subjecting the sample to a LC-MS/MS mass spectrometry technique to quantify the monoclonal antibody therapeutic, wherein the quantifying the antibody therapeutic comprises measuring the variable region of the antibody therapeutic in the sample.
25. A method for quantifying infliximab in a human serum sample, the method
comprising:
a) providing a human serum sample comprising infliximab;
b) adding an internal standard to the sample, wherein the internal standard is a horse IgG;
c) purifying the sample using antibody affinity resin purification;
d) digesting the antibodies present, wherein the digesting comprises trypsin digestion; and
e) subjecting the sample to a LC-MS/MS mass spectrometry technique to quantify infliximab in the sample.
26. A method for quantifying infliximab in a human serum sample, the method
comprising:
a) providing a sample comprising a monoclonal antibody therapeutic, wherein the sample is a human serum sample and the monoclonal antibody therapeutic is infliximab;
b) adding an internal standard to the sample, wherein the internal standard is a horse IgG;
c) purifying the sample using antibody affinity resin purification;
d) digesting the antibodies present, wherein the digesting comprises trypsin digestion; and
e) subjecting the sample to a LC-ESI TRIPLE QUAD MS mass spectrometry technique to quantify infliximab in the sample.
27. A method for quantifying infliximab in a sample, the method comprising:
a) providing a sample comprising infliximab;
b) adding an internal standard to the sample, wherein the internal standard is a horse IgG;
c) purifying the sample using antibody affinity resin purification;
d) digesting the antibodies present, wherein the digesting comprises trypsin digestion; and
e) subjecting the sample to a LC-ESI TRIPLE QUAD MS mass spectrometry technique to quantify infliximab in the sample by measuring the horse IgG constant region and the variable region of infliximab.
28. A method of monitoring a treatment of a disorder in a subject being treated with a monoclonal antibody therapeutic selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituxumab and adalimumab, the method comprising: a) providing a first sample of the subject during treatment;
b) providing a second sample of the subject on a subsequent treatment or after treatment;
c) subjecting the first and second sample to a LC-MS/MS mass spectrometry
technique comprising an internal standard to obtain a mass spectrum of the sample, wherein the internal standard is a horse IgG;
d) quantifying the monoclonal antibody therapeutic in the first and second sample; and
e) comparing the amounts of monoclonal antibody therapeutic from the first and second sample.
29. A method of monitoring a treatment of a disorder in a subject being treated with a monoclonal antibody therapeutic selected from the group consisting of: infliximab, alemtuzumab, eculizumab, rituxumab and adalimumab, the method comprising: a) providing a first sample of the subject during treatment, wherein the first sample is a human serum sample;
b) providing a second sample of the subject on a subsequent treatment or after treatment, wherein the second sample is a human serum sample;
c) subjecting the first and second sample to a LC-MS/MS mass spectrometry
technique comprising an internal standard to obtain a mass spectrum of the sample, wherein the internal standard is a horse IgG;
d) quantifying the monoclonal antibody therapeutic in the first and second sample; and
e) comparing the amounts of monoclonal antibody therapeutic from the first and second sample.
30. A method of monitoring a treatment of a disorder in a subject being treated with infliximab, the method comprising:
a) providing a first human serum sample of the subject during treatment;
b) providing a second human serum sample of the subject on a subsequent
treatment or after treatment;
c) subjecting the first and second sample to a LC-MS/MS mass spectrometry
technique comprising an internal standard to obtain a mass spectrum of the sample, wherein the internal standard is a horse IgG;
d) quantifying infliximab in the first and second sample; and
e) comparing the amounts of infliximab from the first and second sample.
31. A method of monitoring a treatment of a disorder in a subject being treated with infliximab, the method comprising:
a) providing a first human serum sample of the subject during treatment;
b) providing a second human serum sample of the subject on a subsequent treatment or after treatment; c) subjecting the first and second sample to a LC-MS/MS mass spectrometry technique comprising an internal standard to obtain a mass spectrum of the sample, wherein the internal standard is a horse IgG;
d) quantifying infliximab in the first and second sample, wherein the quantifying comprises measuring the horse IgG constant region and the variable region of infliximab; and
e) comparing the amounts of infliximab from the first and second sample.
PCT/US2015/042580 2014-07-29 2015-07-29 Quantifying monoclonal antibody therapeutics by lc-ms/ms WO2016018978A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/329,512 US10690676B2 (en) 2014-07-29 2015-07-29 Quantifying monoclonal antibody therapeutics by LC-MS/MS
EP15827198.1A EP3175242A4 (en) 2014-07-29 2015-07-29 Quantifying monoclonal antibody therapeutics by lc-ms/ms

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462030493P 2014-07-29 2014-07-29
US62/030,493 2014-07-29

Publications (1)

Publication Number Publication Date
WO2016018978A1 true WO2016018978A1 (en) 2016-02-04

Family

ID=55218259

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/042580 WO2016018978A1 (en) 2014-07-29 2015-07-29 Quantifying monoclonal antibody therapeutics by lc-ms/ms

Country Status (3)

Country Link
US (1) US10690676B2 (en)
EP (1) EP3175242A4 (en)
WO (1) WO2016018978A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018183449A1 (en) * 2017-03-31 2018-10-04 Alexion Pharmaceuticals, Inc. Method for simultaneous quantification of alxn1210 and eculizumab in human serum or urine
JP2019509483A (en) * 2016-02-25 2019-04-04 ザ バインディング サイト グループ リミティド Mass spectrometry kit
US10267806B2 (en) 2014-04-04 2019-04-23 Mayo Foundation For Medical Education And Research Isotyping immunoglobulins using accurate molecular mass
WO2019178151A1 (en) * 2018-03-13 2019-09-19 Amgen Inc. Methods for the preparation of trypsin-resistant polypeptides for mass spectrometric analysis
EP3633383A1 (en) * 2018-10-04 2020-04-08 Regeneron Pharmaceuticals, Inc. Fast protein sequencing
US10690676B2 (en) 2014-07-29 2020-06-23 Mayo Roundation for Medical Education and Research Quantifying monoclonal antibody therapeutics by LC-MS/MS
WO2020150491A1 (en) * 2019-01-16 2020-07-23 Regeneron Pharmaceuticals, Inc. Methods for characterizing disulfide bonds
WO2020150328A1 (en) * 2019-01-16 2020-07-23 Regeneron Pharmaceuticals, Inc. Method and system of identifying and quantifying antibody fragmentation
US10955420B2 (en) 2016-09-07 2021-03-23 Mayo Foundation For Medical Education And Research Identification and monitoring of cleaved immunoglobulins by molecular mass
US11209439B2 (en) 2015-09-24 2021-12-28 Mayo Foundation For Medical Education And Research Identification of immunoglobulin free light chains by mass spectrometry
US11946937B2 (en) 2017-09-13 2024-04-02 Mayo Foundation For Medical Education And Research Identification and monitoring of apoptosis inhibitor of macrophage

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021237035A1 (en) * 2020-05-21 2021-11-25 Mayo Foundation For Medical Education And Research Calibration methods for mass spectrometry measurements
EP4162250A1 (en) * 2020-06-09 2023-04-12 Regeneron Pharmaceuticals, Inc. A high-throughput and mass-spectrometry-based method for quantitating antibodies
CN113804804A (en) * 2021-09-08 2021-12-17 杭州佰辰医学检验所有限公司 Monitoring method for rapidly determining monoclonal antibody drug in plasma by ultra-high performance liquid chromatography tandem mass spectrometry

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050064422A1 (en) * 2001-11-29 2005-03-24 Barnidge David R Polypeptide quantitation
US20140045276A1 (en) * 2011-02-17 2014-02-13 Nestec S.A. Assays for detecting autoantibodies to anti-tnfalpha drugs
WO2014109927A1 (en) * 2013-01-11 2014-07-17 The Regents Of The University Of Michigan Synthesis and isolation of dendrimer based imaging systems

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030027216A1 (en) 2001-07-02 2003-02-06 Kiernan Urban A. Analysis of proteins from biological fluids using mass spectrometric immunoassay
JP2005504263A (en) 2001-02-01 2005-02-10 シファーゲン バイオシステムズ, インコーポレイテッド Improved method for protein identification, characterization and sequencing by tandem mass spectrometry
AU2002309423B2 (en) 2001-06-06 2007-11-22 Fujirebio Diagnostics Ab Method to measure gene expression ratio of key genes
DE60228132D1 (en) 2001-12-08 2008-09-18 Micromass Ltd Massenspektormetrie method
US7267942B2 (en) 2003-03-24 2007-09-11 The University Of Hong Kong Diagnostic assay for the human virus causing severe acute respiratory syndrome (SARS)
US7709610B2 (en) 2003-05-08 2010-05-04 Facet Biotech Corporation Therapeutic use of anti-CS1 antibodies
NZ552476A (en) 2004-06-10 2009-09-25 Viventia Biotech Inc Tumor specific antibody
CA2611728A1 (en) 2005-06-08 2006-12-14 Millennium Pharmaceuticals, Inc. Methods for the identification, assessment, and treatment of patients with cancer therapy
US20110177492A1 (en) 2005-06-16 2011-07-21 3M Innovative Properties Company Method of classifying chemically crosslinked cellular samples using mass spectra
EP2013356B1 (en) 2006-05-02 2010-09-01 Genentech, Inc. Microwave assisted deglycosylation of proteins for molecular weight determination by mass spectrometry
WO2008021183A2 (en) 2006-08-10 2008-02-21 Millennium Pharmaceuticals, Inc. For the identification, assessment, and treatment of patients with cancer therapy
US20090203602A1 (en) 2006-09-01 2009-08-13 Cohava Gelber Compositions and methods for diagnosis and treatment of type 2 diabetes
CA2664333A1 (en) 2006-09-15 2008-03-20 Memorial Sloan Kettering Cancer Center Methods of diagnosing, treating, or preventing plasma cell disorders
FI20075278A0 (en) 2007-04-20 2007-04-20 Biotie Therapies Corp Novel completely human anti-VAP-1 monoclonal antibodies
US8501907B2 (en) 2007-08-10 2013-08-06 Janssen Biotech, Inc. Immunoglobulin cleavage fragments as disease indicators and compositions for detecting and binding such
BRPI0820221A2 (en) 2007-11-22 2015-05-26 Symphogen As Method for characterization of a recombinant polyclonal protein
WO2009100390A2 (en) 2008-02-08 2009-08-13 Mayo Foundation For Medical Education And Research Classifying amyloidosis
WO2009148528A2 (en) 2008-05-30 2009-12-10 Millennium Pharmaceuticals, Inc. Assessment of chromosomal alterations to predict clinical outcome of bortezomib treatment
WO2010002911A2 (en) 2008-06-30 2010-01-07 H. Lee Moffitt Cancer Center And Research Institute, Inc. Methods and materials for monitoring myeloma using quantitative mass spetrometry
JP6016892B2 (en) 2011-04-29 2016-10-26 セルジーン コーポレイション Methods for treating cancer and inflammatory diseases using cereblon as a predictor
CA2833212C (en) 2011-05-12 2020-06-09 Genentech, Inc. Multiple reaction monitoring lc-ms/ms method to detect therapeutic antibodies in animal samples using framework signature peptides
JP2014523589A (en) 2011-07-13 2014-09-11 ザ マルチプル ミエローマ リサーチ ファウンデーション, インコーポレイテッド Methods for data collection and distribution
GB2495113A (en) 2011-09-29 2013-04-03 Bioinvent Int Ab Anti-ICAM-1 antibody for treating multiple-myeloma-related disorders
CN108152513B (en) 2011-09-29 2020-07-24 西雅图基因公司 Overall molecular weight determination of protein-coupled reagent compounds
CA2859808A1 (en) 2011-12-19 2013-06-27 The Washington University Methods for diagnosing alzheimer's disease
AU2013258834B2 (en) 2012-05-10 2017-09-07 Zymeworks Bc Inc. Heteromultimer constructs of immunoglobulin heavy chains with mutations in the Fc domain
WO2013181576A2 (en) * 2012-06-01 2013-12-05 Momenta Pharmaceuticals, Inc. Methods of evaluating and making biologics
WO2013191908A1 (en) 2012-06-20 2013-12-27 The University Of North Carolina At Chapel Hill Integrated sample processing for electrospray ionization devices
US20140186332A1 (en) 2012-12-28 2014-07-03 NX Pharmagen Biomarkers of preterm birth
WO2014150170A1 (en) 2013-03-15 2014-09-25 Mayo Foundation For Medical Education And Research Identification and monitoring of monoclonal immunoglobulins by molecular mass
EP3043806A4 (en) 2013-09-11 2017-05-17 University Of Southern California A composition of stem cells having highly expressed fas ligand
US10267806B2 (en) 2014-04-04 2019-04-23 Mayo Foundation For Medical Education And Research Isotyping immunoglobulins using accurate molecular mass
WO2016018978A1 (en) 2014-07-29 2016-02-04 Mayo Foundation For Medical Education And Research Quantifying monoclonal antibody therapeutics by lc-ms/ms
JP6666916B2 (en) 2015-08-06 2020-03-18 積水メディカル株式会社 Testing methods for kidney disease
WO2017053932A1 (en) 2015-09-24 2017-03-30 Mayo Foundation For Medical Education And Research Identification of immunoglobulin free light chains by mass spectrometry
EP3523647B1 (en) 2016-09-07 2024-06-26 Mayo Foundation for Medical Education and Research Identification and monitoring of cleaved immunoglobulins by molecular mass

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050064422A1 (en) * 2001-11-29 2005-03-24 Barnidge David R Polypeptide quantitation
US20140045276A1 (en) * 2011-02-17 2014-02-13 Nestec S.A. Assays for detecting autoantibodies to anti-tnfalpha drugs
WO2014109927A1 (en) * 2013-01-11 2014-07-17 The Regents Of The University Of Michigan Synthesis and isolation of dendrimer based imaging systems

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
HEUDI ET AL., ANALYTICAL CHEMISTRY, vol. 80, no. 11, 1 June 2008 (2008-06-01), pages 4200 - 4207
LADWIG ET AL., CLINICAL CHEMISTRY, vol. 60, no. 8, 5 May 2014 (2014-05-05), pages 1080 - 1088
LI ET AL.: "Simultaneous Analysis of Multiple Monoclonal Antibody Biotherapeutics by LC-MS/MS Method in Rat Plasma Following Cassette-Dosing", AAPS JOURNAL, vol. 15, no. 2, 2013, pages 337 - 346, XP055390980 *
See also references of EP3175242A4 *
WILLRICH ET AL., GASTROENTEROLOGY AGA ABSTRACTS, 1 May 2014 (2014-05-01)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10753945B2 (en) 2014-04-04 2020-08-25 Mayo Foundation For Medical Education And Research Isotyping immunoglobulins using accurate molecular mass
US10267806B2 (en) 2014-04-04 2019-04-23 Mayo Foundation For Medical Education And Research Isotyping immunoglobulins using accurate molecular mass
US11604196B2 (en) 2014-04-04 2023-03-14 Mayo Foundation For Medical Education And Research Isotyping immunoglobulins using accurate molecular mass
US10690676B2 (en) 2014-07-29 2020-06-23 Mayo Roundation for Medical Education and Research Quantifying monoclonal antibody therapeutics by LC-MS/MS
US11209439B2 (en) 2015-09-24 2021-12-28 Mayo Foundation For Medical Education And Research Identification of immunoglobulin free light chains by mass spectrometry
JP2019509483A (en) * 2016-02-25 2019-04-04 ザ バインディング サイト グループ リミティド Mass spectrometry kit
US10955420B2 (en) 2016-09-07 2021-03-23 Mayo Foundation For Medical Education And Research Identification and monitoring of cleaved immunoglobulins by molecular mass
US11112411B2 (en) 2017-03-31 2021-09-07 Alexion Pharmaceuticals, Inc. Method for simultaneous quantification of ALXN1210 and eculizumab in human serum or urine
JP2020516865A (en) * 2017-03-31 2020-06-11 アレクシオン ファーマシューティカルズ, インコーポレイテッド Method for simultaneous quantification of ALXN1210 and eculizumab in human serum or urine
WO2018183449A1 (en) * 2017-03-31 2018-10-04 Alexion Pharmaceuticals, Inc. Method for simultaneous quantification of alxn1210 and eculizumab in human serum or urine
JP7247099B2 (en) 2017-03-31 2023-03-28 アレクシオン ファーマシューティカルズ, インコーポレイテッド Method for Simultaneous Quantitation of ALXN1210 and Eculizumab in Human Serum or Urine
US11946937B2 (en) 2017-09-13 2024-04-02 Mayo Foundation For Medical Education And Research Identification and monitoring of apoptosis inhibitor of macrophage
WO2019178151A1 (en) * 2018-03-13 2019-09-19 Amgen Inc. Methods for the preparation of trypsin-resistant polypeptides for mass spectrometric analysis
EP4321870A3 (en) * 2018-03-13 2024-04-03 Amgen Inc. Methods for the preparation of trypsin-resistant polypeptides for mass spectrometric analysis
EP3633383A1 (en) * 2018-10-04 2020-04-08 Regeneron Pharmaceuticals, Inc. Fast protein sequencing
EP3964836A1 (en) * 2018-10-04 2022-03-09 Regeneron Pharmaceuticals, Inc. Fast protein sequencing
US11726096B2 (en) 2018-10-04 2023-08-15 Regeneron Pharmaceuticals, Inc. Fast protein sequencing
US11579150B2 (en) 2019-01-16 2023-02-14 Regeneron Pharmaceuticals, Inc. Methods for characterizing disulfide bonds
WO2020150328A1 (en) * 2019-01-16 2020-07-23 Regeneron Pharmaceuticals, Inc. Method and system of identifying and quantifying antibody fragmentation
US11486864B2 (en) 2019-01-16 2022-11-01 Regeneron Pharmaceuticals, Inc. Method and system of identifying and quantifying antibody fragmentation
EP4220173A3 (en) * 2019-01-16 2023-10-18 Regeneron Pharmaceuticals, Inc. Methods for characterizing disulfide bonds
CN113272650A (en) * 2019-01-16 2021-08-17 瑞泽恩制药公司 Methods for characterizing disulfide bonds
WO2020150491A1 (en) * 2019-01-16 2020-07-23 Regeneron Pharmaceuticals, Inc. Methods for characterizing disulfide bonds
US12000839B2 (en) 2019-01-16 2024-06-04 Regeneron Pharmaceuticals, Inc. Methods for characterizing disulfide bonds
US12000811B2 (en) 2019-01-16 2024-06-04 Regeneron Pharmaceuticals, Inc. Method and system of identifying and quantifying antibody fragmentation

Also Published As

Publication number Publication date
EP3175242A1 (en) 2017-06-07
US20180106815A1 (en) 2018-04-19
US10690676B2 (en) 2020-06-23
EP3175242A4 (en) 2017-12-27

Similar Documents

Publication Publication Date Title
US10690676B2 (en) Quantifying monoclonal antibody therapeutics by LC-MS/MS
AU2019268150B2 (en) Isotyping immunoglobulins using accurate molecular mass
EP2966985B1 (en) Identification and monitoring of monoclonal immunoglobulins by molecular mass
EP3165922B1 (en) A method for quantifying therapeutic antibodies
CN103217489B (en) A kind ofly measure the glycosylation of sample in protein purification technological process and the method for end modified situation
AU2017325022B2 (en) Identification and monitoring of cleaved immunoglobulins by molecular mass
CN109900815B (en) Absolute quantitative analysis of IgG glycopeptides in serum
US11630111B2 (en) Method for quantifying anti-TNF antibodies
US11946937B2 (en) Identification and monitoring of apoptosis inhibitor of macrophage
US20200284800A1 (en) Identification and monitoring of immunoglobulin j chains
EP3682249A1 (en) Identification and monitoring of acid hydrolysis products of immunoglobulin heavy chains
JP2022502683A (en) Quantification of antibodies in biological samples
EP3371602B1 (en) A method for quantifying anti-tnf antibodies
EP3371592B1 (en) A method for quantifying therapeutic antibodies

Legal Events

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

Ref document number: 15827198

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015827198

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015827198

Country of ref document: EP