US20230417761A1 - Detection and quantification of natalizumab - Google Patents

Detection and quantification of natalizumab Download PDF

Info

Publication number
US20230417761A1
US20230417761A1 US18/312,915 US202318312915A US2023417761A1 US 20230417761 A1 US20230417761 A1 US 20230417761A1 US 202318312915 A US202318312915 A US 202318312915A US 2023417761 A1 US2023417761 A1 US 2023417761A1
Authority
US
United States
Prior art keywords
natalizumab
peptide
antibody
test device
test
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/312,915
Inventor
Bradley T. Messmer
Dina UZRI
Jessie-Farah Fecteau
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Abreos Biosciences Inc
Original Assignee
Abreos Biosciences Inc
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 Abreos Biosciences Inc filed Critical Abreos Biosciences Inc
Priority to US18/312,915 priority Critical patent/US20230417761A1/en
Assigned to ABREOS BIOSCIENCES, INC. reassignment ABREOS BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MESSMER, BRADLEY T., FECTEAU, JESSIE-FARAH, UZRI, Dina
Publication of US20230417761A1 publication Critical patent/US20230417761A1/en
Pending legal-status Critical Current

Links

Images

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
    • G01N33/6854Immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70546Integrin superfamily, e.g. VLAs, leuCAM, GPIIb/GPIIIa, LPAM

Definitions

  • Monoclonal antibodies (mAb) and other biologics are targeted therapies that are increasingly being used to treat indications such as cancer and autoimmune disease, such as multiple sclerosis (MS).
  • MS is a leading cause of neurologic disability, and the disease is characterized by multiple inflammatory lesions and demyelination within the white matter of the central nervous system (CNS).
  • Natalizumab (marketed as Tysabri by Biogen Idec) is the top-selling biologic drug indicated for treatment of MS and is being used to treat 25,000 to 50,000 of the 400,000 MS patients in the US.
  • described herein are methods, assays, complexes, and devices for measuring natalizumab in a sample.
  • described herein is a method of capturing an antibody in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23, allowing binding of the peptide with the antibody to form an antibody-peptide complex, and detecting the antibody-peptide complex.
  • the peptide is selected from the group consisting of SEQ ID NOs:1, 2, 4, 13, 16, and 18-23.
  • the antibody is not complexed to an epitope of a target protein.
  • the antibody is natalizumab.
  • the biological sample is selected from the group consisting of body fluids, tissues, body swabs, and body smears.
  • the biological sample is a fluid.
  • the fluid contains antibody at a concentration of between about 0.5 mcg/mL to 120 mcg/mL.
  • the fluid is selected from the group consisting of serum, plasma, whole blood, red blood cell concentrates, platelet concentrates, leukocytes concentrates, urine, cerebral spinal fluid, and sputum.
  • the biological sample is obtained from a human.
  • described herein is a method of capturing natalizumab in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23, wherein the peptide is attached to a solid support.
  • the peptide binds to the antigen binding site of the antibody.
  • described herein is a method of capturing an antibody in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23, allowing binding of the peptide with the antibody to form an antibody-peptide complex, and detecting the antibody-peptide complex, wherein detection of the antibody-peptide complex is performed by detection of a detectable label on the antibody or the peptide.
  • detection of the antibody-peptide complex is performed by Western blot analysis, dot blot analysis, flow cytometry, enzyme-linked immunosorbent assay (ELISA), lateral flow immunoassay, radioimmunoassay (RIA), competition immunoassay, dual antibody sandwich assay, chemiluminescent assay, bioluminescent assay, fluorescent assay, or agglutination assay.
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • competition immunoassay dual antibody sandwich assay
  • chemiluminescent assay bioluminescent assay
  • fluorescent assay or agglutination assay.
  • a natalizumab-mimetope complex comprising: a mimetope comprising a peptide between 7 and 26 amino acids long; and natalizumab.
  • the peptide is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23.
  • test device comprising: a sample pad for receiving a biologic; a conjugate pad; and a test membrane comprising at least one test line comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23.
  • the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane.
  • the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab.
  • the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane and an antibody specific for natalizumab.
  • the antibody specific for natalizumab binds natalizumab at a variable region.
  • the antibody specific for natalizumab binds natalizumab at a constant region.
  • the test membrane further comprises at least one test line comprising an antibody specific for the natalizumab-binding mimetope peptide.
  • the antibody is natalizumab or a biosimilar thereof or a new antibody specific for the selected peptide obtained by conventional methods, such as animal immunization using the selected peptide as an immunogen.
  • Also described herein is a method of preventing progressive multifocal leukoencephalopathy (PML) comprising: identifying a subject receiving antibody therapy at risk of developing PML; obtaining a biological fluid from the subject; contacting the biological fluid with a test device comprising: a sample pad for receiving the biological fluid; a conjugate pad; and a test membrane comprising at least one test line comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23; and, increasing the frequency of performing the method steps based on the results obtained following the step of contacting the biological fluid with a test device.
  • PML progressive multifocal leukoencephalopathy
  • the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane. In some embodiments, the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane and an antibody specific for natalizumab. In some embodiments, the antibody specific for natalizumab binds natalizumab at a variable region. In some embodiments, the antibody specific for natalizumab binds natalizumab at a constant region.
  • the test membrane further comprises at least one test line comprising an antibody specific for the natalizumab-binding mimetope peptide.
  • the antibody is natalizumab or a biosimilar thereof or a new antibody specific for the selected peptide obtained by conventional methods, such as animal immunization using the selected peptide as an immunogen.
  • the step of increasing the frequency of performing the method steps further comprises adjusting the antibody therapy based on the results obtained following the step of contacting the biological fluid with a test device.
  • a natalizumab-binding mimetope comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23. Also described herein is a method of detecting natalizumab in a biological sample comprising: contacting said biological sample with a natalizumab mimetope; allowing binding of said natalizumab mimetope with the natalizumab to form a natalizumab-natalizumab mimetope complex; and detecting the natalizumab-natalizumab mimetope complex.
  • the natalizumab mimetope is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 96% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 97% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 98% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23.
  • the natalizumab mimetope is at least 99% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is 100% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23.
  • described herein are methods, assays, complexes, and devices for measuring active natalizumab in a sample.
  • described herein is a method of capturing an antibody in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30, allowing binding of the peptide with the antibody to form an antibody-peptide complex, and detecting the antibody-peptide complex.
  • the peptide is selected from the group consisting of SEQ ID NOs:24, 29, and 30.
  • the antibody is not complexed to an epitope of a target protein.
  • the antibody is natalizumab.
  • the antibody is active natalizumab
  • described herein is a method of capturing active natalizumab in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30, wherein the peptide is attached to a solid support.
  • the peptide binds to the antigen binding site of the active natalizumab.
  • described herein is a method of capturing an antibody in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30, allowing binding of the peptide with the antibody to form an antibody-peptide complex, and detecting the antibody-peptide complex, wherein detection of the antibody-peptide complex is performed by detection of a detectable label on the antibody or the peptide.
  • detection of the antibody-peptide complex is performed by Western blot analysis, dot blot analysis, flow cytometry, enzyme-linked immunosorbent assay (ELISA), lateral flow immunoassay, radioimmunoassay (RIA), competition immunoassay, dual antibody sandwich assay, chemiluminescent assay, bioluminescent assay, fluorescent assay, or agglutination assay.
  • ELISA enzyme-linked immunosorbent assay
  • RIA radioimmunoassay
  • competition immunoassay dual antibody sandwich assay
  • chemiluminescent assay bioluminescent assay
  • fluorescent assay or agglutination assay.
  • a natalizumab-mimetope complex comprising: a mimetope comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30; and active natalizumab.
  • test device comprising: a sample pad for receiving a biologic; a conjugate pad; and a test membrane comprising at least one test line comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30.
  • the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane.
  • the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab.
  • the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane and an antibody specific for natalizumab.
  • the antibody specific for natalizumab binds natalizumab at a variable region.
  • the antibody specific for natalizumab binds natalizumab at a constant region.
  • the test membrane further comprises at least one test line comprising an antibody specific for the natalizumab-binding mimetope peptide.
  • the antibody is natalizumab or a biosimilar thereof or a new antibody specific for the selected peptide obtained by conventional methods, such as animal immunization using the selected peptide as an immunogen.
  • Also described herein is a method of preventing progressive multifocal leukoencephalopathy (PML) comprising: identifying a subject receiving antibody therapy at risk of developing PML; obtaining a biological fluid from the subject; contacting the biological fluid with a test device comprising: a sample pad for receiving the biological fluid; a conjugate pad; and a test membrane comprising at least one test line comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30; and, increasing the frequency of performing the method steps based on the results obtained following the step of contacting the biological fluid with a test device.
  • PML progressive multifocal leukoencephalopathy
  • the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane. In some embodiments, the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane and an antibody specific for natalizumab. In some embodiments, the antibody specific for natalizumab binds natalizumab at a variable region. In some embodiments, the antibody specific for natalizumab binds natalizumab at a constant region.
  • the test membrane further comprises at least one test line comprising an antibody specific for the natalizumab-binding mimetope peptide.
  • the antibody is natalizumab or a biosimilar thereof or a new antibody specific for the selected peptide obtained by conventional methods, such as animal immunization using the selected peptide as an immunogen.
  • the step of increasing the frequency of performing the method steps further comprises adjusting the antibody therapy based on the results obtained following the step of contacting the biological fluid with a test device.
  • a natalizumab-binding mimetope comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. Also described herein is a method of detecting active natalizumab in a biological sample comprising: contacting said biological sample with a natalizumab mimetope; allowing binding of said natalizumab mimetope with the natalizumab to form a natalizumab-natalizumab mimetope complex; and detecting the natalizumab-natalizumab mimetope complex.
  • the natalizumab mimetope is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 96% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 97% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 98% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30.
  • the natalizumab mimetope is at least 99% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is 100% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30.
  • FIG. 1 is a graph showing the specificity of natalizumab (NTZ) mimetope peptides for the detection of natalizumab by enzyme-linked immunosorbent assay (ELISA).
  • FIG. 2 is a graph showing the sensitivity of a natalizumab (NTZ) mimetope peptide-based enzyme-linked immunosorbent assay (ELISA) for natalizumab spiked in 2.5% BSA/TBST buffer.
  • NTZ-01-Bio and NTZ-02-Bio peptides were coated at 25 ug/mL or 50 ug/mL in TBS.
  • FIGS. 3 A- 3 C are graphs showing a titration of the NTZ-01-Bio peptide coating concentration and calibration curves of natalizumab prepared in buffer (TBST-2.5% BSA) and human serum (0.1%).
  • FIGS. 4 A and 4 B are graphs showing the sensitivity and specificity of the NTZ-01-Bio mimetope peptide-based ELISA for natalizumab spiked in human serum (0.1%).
  • FIG. 4 A presents results from 2 independent experiments (mean+/ ⁇ SD). The dashed line indicates OD without natalizumab present in the samples.
  • FIG. 4 B depicts results with all wells coated with 7.5 ug/mL NTZ-01-Bio (except the no coating control). Natalizumab was spiked at 2560 ng/mL and the other therapeutic monoclonal antibodies were spiked at 2000 ng/mL in human serum (0.1%).
  • FIG. 5 is a graph illustrating the sensitivity of the NTZ-01-Bio, NTZ-04-Bio, and NTZ-05-Bio mimetope peptide-based ELISAs for natalizumab spiked in human serum (0.4%). Results from a single experiment are presented. All wells were coated with 9 uM peptide except no coating control. The calibration curve was prepared using natalizumab-spiked human serum and exposed to 3 mM GSH for 18 hours at 37° C. to generate monovalent antibody.
  • FIGS. 6 A and 6 B are graphs illustrating the detection of the monovalent form of natalizumab in human serum (0.4%) using the mimetope peptide-based ELISA (NTZ-01-Bio).
  • FIG. 7 depicts an exemplary lateral flow immunoassay (LFA) device for rapid measurement of natalizumab levels in a finger-stick blood sample.
  • LFA lateral flow immunoassay
  • FIG. 8 depicts detection of free natalizumab levels using a combination of sandwich and competitive assays in the same lateral flow immunoassay (LFA).
  • FIG. 9 depicts a flow cytometry histogram of binding between natalizumab and its cellular target, following incubation of natalizumab with varying concentrations of either a natalizumab-specific VERITOPETM or an irrelevant peptide. Jeko-1 cells, expressing CD49d, were used as a model.
  • FIGS. 10 A and B depicts a graphical comparison of a natalizumab-specific VERITOPETM ( FIG. 10 A ) and anti-idiotype ( FIG. 10 B ) ELISA binding.
  • FIGS. 11 A and B illustrate the characterization of a natalizumab-specific VERITOPETM (NTZ-01-Bio; SEQ ID NO:24) and an anti-idiotype.
  • natalizumab-specific VERITOPETM is the capture reagent and anti-idiotype is the competitive reagent.
  • anti-idiotype is the capture reagent and natalizumab-specific VERITOPETM is the competitive reagent.
  • FIGS. 12 A and B demonstrate that heat-inactivation of natalizumab alters bivalent natalizumab ( FIG. 12 A ) and arm-exchanged (bispecific) natalizumab ( FIG. 12 B ) binding to its cellular target.
  • FIGS. 13 A and B depict a comparison between anti-idiotype ELISA ( FIG. 13 A ) and natalizumab-specific VERITOPETM ( FIG. 13 B ; NTZ-01-Bio; SEQ ID NO:24).
  • FIG. 14 depicts natalizumab-specific VERITOPETM (NTZ-01-Bio; SEQ ID NO:24) ELISA assay calibration curve.
  • FIG. 15 depicts stability of human serum samples spiked with a natalizumab-specific VERITOPETM (NTZ-01-Bio; SEQ ID NO:24) up to 8 days.
  • FIG. 16 depicts a comparison of the calibration curves for NTZ01 (SEQ ID NO:24), NTZ06 (SEQ ID NO:29), and NTZ07 (SEQ ID NO:30) natalizumab-specific VERITOPESTM.
  • FIG. 17 depicts the specificity of natalizumab-specific VERITOPETM NTZ07 (SEQ ID NO:30) compared to 4 other monoclonal antibodies.
  • natalizumab (marketed as Tysabri by Biogen Idec).
  • Natalizumab is one of the most effective treatments available to reduce relapse frequency in multiple sclerosis (MS) patients. Like most mAbs, natalizumab displays highly variable pharmacokinetics (PK) across patients, compounded by the standard dosing that is not body mass or surface area adjusted.
  • PK pharmacokinetics
  • Natalizumab is the top selling biologic drug indicated for treatment of MS and is being used to treat 25,000-50,000 of the 400,000 MS patients in the US and also used in the treatment of Crohn's disease.
  • Natalizumab is a humanized recombinant mAb that targets the ⁇ 4 chain of ⁇ 4 ⁇ 1 integrin (also known as very late activation antigen 4; VLA-4) and ⁇ 4 ⁇ 7 integrin and is thought to function by blocking migration of immune cells across the blood-brain barrier into the central nervous system (CNS), thus suppressing inflammation in patients with relapsing-remitting multiple sclerosis (Vennegoor A, et al. Clinical relevance of serum natalizumab concentration and anti - natalizumab antibodies in multiple sclerosis . Mult Scler [Internet].
  • Natalizumab is a full-length antibody of the IgG4 subclass and consists of two heavy and two light chains connected by four inter-chain disulfide bonds. Like other IgG4 antibodies, natalizumab demonstrates reduced binding to Fc gamma receptors and a lack of ability to fix complement in vitro. As a result, natalizumab can block interaction of ⁇ 4-integrins with their cognate receptors with minimal cell killing.
  • Natalizumab given by infusion, is very expensive and costs approximately $4,000-5,000 for a single dose, leading to an annual cost of close to $65,000 if the drug is taken every 4 weeks as recommended on the label.
  • the immunosuppressive activity of natalizumab has been associated with reawakening of JC polyomavirus, which may lead to progressive multifocal leukoencephalopathy (PML), a serious and often-fatal opportunistic brain infection.
  • PML progressive multifocal leukoencephalopathy
  • the standard dosing regimen for natalizumab is 300 mg by IV infusion every 4 weeks.
  • the current dosing strategy for natalizumab is not customized for each patient, and there is growing evidence of considerable variability in the rate at which different patients clear the drug from their bodies.
  • a 2011 study by Foley reported that patients with MS who were receiving monthly doses of natalizumab exhibited patient-to-patient variability in their serum natalizumab levels. Furthermore, natalizumab was found to accumulate in the serum in some patients who did not clear the drug within the 4-week period (Foley J. Progressive escalation of natalizumab serum concentration as a potential kinetic marker for PML risk assessment . Oral communication, abstract S51.004, April 2011. American Academy of Neurology.
  • Peptide-based immunoassays can be developed for monitoring mAb levels.
  • Phage displayed peptide libraries are used to select peptide sequences that mimic the target antigen of a given mAb.
  • Peptide libraries displayed on bacteriophage are routinely used to identify peptide epitopes, or mimetopes (also referred to as VERITOPESTM), recognized by antibodies.
  • Phage display works best with concentrated and highly purified proteins, and as such therapeutic mAb are ideal targets. When short peptides, 7 to 26 amino acids long, are screened, the selected peptides almost invariably bind to the antigen-binding site of the antibody and are competed by the natural ligand (Sanchez A B, et al.
  • Mimetope peptides may also be selected from a library that contains cysteines flanking the peptide mimetope sequence to increase the stability of the peptide through disulfide bond formation.
  • the methods, assays, complexes, and devices described herein are useful for monitoring the level of mAb in a subject.
  • the level of mAb is monitoring using a biological sample obtained from a subject.
  • the subject is a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult, or senior adult)) and/or other non-human animals, for example, mammals (e.g., primates (e.g., cynomolgus monkeys, rhesus monkeys); commercially relevant or research mammals (such as cattle, pigs, horses, sheep, goats, dogs, cats, rabbits, rats, and/or mice); and birds (e.g., commercially relevant birds such as chickens, ducks, geese, and/or turkeys).
  • mammals e.g., primates (e.g.
  • One advantage of the methods, assays, complexes, and devices described herein is that they provide means to detect, monitor, and record drug levels in a patient with precision directly at the point-of-care.
  • the drugs can be novel or biosimilar drugs, and the technology is based on the binding of a mimetope to an antibody.
  • Mimetopes are peptides that specifically detect a given biological drug (novel or biosimilar) in a biologic fluid. Mimetopes are described in U.S. Pat. No. 9,250,233, which is hereby incorporated in full by reference.
  • the tests are optionally performed with a disposable, integrated point-of-care device, or performed with a laboratory-based enzyme-linked immunosorbent assay (ELISA), or performed with a fixed reader, as a tabletop system connected to a computer.
  • Devices and systems for use with mimetopes include those performed with a personal point-of-care device.
  • the personal point-of-care device comprises a housing, a display, a test strip holder, a test strip comprising a peptide which binds to an antigen-binding site of an antibody present in a sample from a user, an imaging device for imaging the test strip, a processor, an onboard memory, and a communications element.
  • the personal point-of-care device includes an on-device display.
  • the display is embedded in a face of the device and the test strip holder is encased by the device.
  • the display is not attached to the device.
  • the display color, font, image size, contrast, or contents are user-selected.
  • the display may render various icons or messages to a user, such as test results, device status, or error messages.
  • the personal point-of-care device optionally includes an audio indicator or a light indicator.
  • contents of the on-display device outputs into audio by user-selection.
  • the device is reversibly connected to a mobile device, a computer, a GPS, an IPAD, a USB drive, a printer, a scanner, a television, a server, a car, a smart watch, smart glasses, an IPOD, a game player, a projector, a camera, or similar electronic devices.
  • the methods, assays, complexes, and devices described herein provide benefit to the patient, the medical professional, the pharmaceutical company, and the insurance company.
  • the patient benefits by receiving the optimal dose of natalizumab (avoiding high cost and adverse side effects associated with excessive dosing such as PML, or poor response to treatment associated with insufficient dosing).
  • the medical professional benefits with knowledge that a patient has a specific drug level.
  • the medical professional has the information to adjust the dose accurately in order to increase or decrease the patient's response to natalizumab.
  • the pharmaceutical company benefits by saving costs from not having natalizumab in limited supply squandered due to waste or inefficiency.
  • the insurance company benefits by not having to purchase additional natalizumab beyond the amount needed for optimal dosing of a patient.
  • therapeutic dose monitoring of natalizumab with the methods, assays, complexes and devices described herein will enable the identification of patients who have excessive free drug levels producing potentially elevated VLA-4 receptor saturations with reduced or absent immune cell trafficking and are candidates for dose extension.
  • the methods, assays, complexes and devices described herein are optionally also used to identify patients with sub-therapeutic drug levels and may indicate development of anti-drug antibodies.
  • Different therapeutic interventions are performed on patients having lower drug levels (identified via the methods, assays, complexes, and devices described herein), such as increasing the dose, or frequency of dosing, to maximize therapeutic benefit, or rapidly switching to another treatment approach more likely to succeed.
  • patients for which drug levels may be well above the range required for optimal clinical benefit could have subsequent treatments delayed, reducing the risk of PML without affecting outcomes, or minimally be placed under closer medical monitoring based on the risk associated with elevated drug levels.
  • the methods, assays, complexes, and devices described herein comprise sample pads, conjugate pads, colloidal gold conjugates (or other colored or fluorescent monodispersed types of particle conjugates such as those made of latex), test lines, and/or membranes.
  • the sample pads are pretreated with blocking agents and surfactants to improve flow and subsequent release of agents from the conjugate pad.
  • the sample pad may also include a filter that removes cells while allowing serum or plasma to flow through to the membrane.
  • the colloidal gold may be a monodisperse and uniform solution of 40 nm colloidal gold prepared by a reduction of aqueous HAuCl 4 with a cherry red appearance.
  • mimetope peptide may be conjugated to the gold colloids by passive adsorption, and the coating density of the mimetope on the gold may be optimized to enable measurement of the proportion of bivalent natalizumab in a sample.
  • Conjugate pad parameters including flow rate, release characteristics, and stability, may be optimized to enable measurement of the proportion of bivalent natalizumab in a sample.
  • Pretreatment of a conjugate pad with blocking and/or stabilizing buffers can improve these parameters.
  • colloidal gold conjugates are dried onto the pre-treated conjugate pads and are able to return to solution when sample is present.
  • the methods, assays, complexes, and devices comprise a test line comprising a mimetope peptide specific for natalizumab that serves as the first test line to capture bivalent antibody.
  • mimetope peptides are synthesized and attached to bovine serum albumin (BSA) which greatly improves adsorption of the peptide onto the membrane.
  • BSA bovine serum albumin
  • natalizumab or another antibody that specifically captures the natalizumab peptide serves as the second test line for the competitive assay portion of the test and allows measurement of both monovalent and bivalent natalizumab.
  • the mimetope test line concentration and the natalizumab test line concentration will be varied to optimize the quantitative feature of the test.
  • the mimetope test line concentration or natalizumab test line concentration is between 0.1 mg/mL and 2.5 mg/mL.
  • the mimetope test line concentration or natalizumab test line concentration is selected from the group consisting of 0.1 mg/mL, 0.2 mg/mL, 0.3 mg/mL, 0.4 mg/mL, 0.5 mg/mL, 0.6 mg/mL, 0.7 mg/mL, 0.8 mg/mL, 0.9 mg/mL, 1.0 mg/mL, 1.2 mg/mL, 1.4 mg/mL, 1.6 mg/mL, 1.8 mg/mL, 2.0 mg/mL, 2.2 mg/mL, 2.4 mg/mL, 2.6 mg/mL, 2.8 mg/mL, 3.0 mg/mL, 3.2 mg/mL, 3.4 mg/mL, 3.6 mg/mL, 3.8 mg/mL, 4.0 mg/mL, 4.2 mg/mL, 4.4 mg/mL, 4.6 mg/mL, 4.8 mg/mL, 5.0 mg/mL, 5.2 mg/mL, 5.4 mg/mL,
  • nitrocellulose membranes are used.
  • Alternative membrane materials may also be used.
  • the optimal membrane may is determined empirically by testing 5-10 different forms, varied by pore size and wicking rate. Test lines may be striped onto the membrane using an automated programmable dispenser. Membranes may be blocked to reduce non-specific binding, and blocking can influence the wicking rate.
  • the assays described herein are shelf-stable for at least two years.
  • Components that contribute to shelf life include the stability of the reagents, such as antibodies and peptides, as well as the physical components of the assay.
  • Assembled assays may be stored in foil pouches with a desiccant. Long term storage tests and exposure to non-optimal conditions of the assays described herein may be performed via accelerated stability testing by varying temperature, humidity and light.
  • sample assays are incubated at 4° C., room temperature, 37° C., and 55° C., and tested at various times. One week at 55° C. can simulate one year at room temperature.
  • Humidity testing may be performed with both open and closed packages at 30% and 80% relative humidity at room temperature and 37° C. Light exposure can be performed for several weeks.
  • the methods, assays, complexes, and devices described herein comprise specific mimetope peptide sequences for the capture and quantification of free and active natalizumab in human serum.
  • the mimetope peptide specific for natalizumab is selected from the group consisting of SEQ ID NOs: 1-23.
  • the natalizumab mimetope is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23.
  • the natalizumab mimetope is at least 96% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23.
  • the natalizumab mimetope is at least 97% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 98% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 99% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is 100% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23.
  • the mimetope peptide specific for natalizumab is selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 96% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 97% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30.
  • the natalizumab mimetope is at least 98% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 99% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is 100% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. Biotinylated peptides are optionally attached to streptavidin coated plates and used as a surrogate ligand to capture natalizumab in immunoassays.
  • an enzyme linked immunosorbent assay with a calibration range from 20 to 240 ng/mL (after the minimum required sample dilution) is described, corresponding to a concentration range from 5-60 ug/mL natalizumab in undiluted human serum.
  • ELISA enzyme linked immunosorbent assay
  • Such an ELISA can have an intra- and inter-assay coefficient of variations ranging from 1.0 to 7.9% and from 4.2% to 18.9%, respectively.
  • described herein is a lateral flow immunoassay with a lower bound of detection of 10 ug/mL and an upper bound of detection of 100 ug/mL.
  • active natalizumab is distinguished from denatured or inactive natalizumab using the methods, assays, complexes, and devices disclosed.
  • Active natalizumab as used herein, is able to bind to alpha-4 integrin and trigger downstream events, including preventing passage of immune cells, such as white blood cells, across blood vessel walls into affected organs, such as the brain, spinal cord, and bowel.
  • Natalizumab may be denatured or inactivated by factors such as heat, high or low pH, exposure to organic solvents, length of time, enzymes, oxidizing agents, other stress conditions, or post-translational modifications, such as but not limited to: asparagine deamidation, aspartate isomerization, methionine oxidation, and lysine glycation.
  • Denatured or inactive natalizumab may still bind to anti-idiotype antibodies, as discussed below, but will not lead to the beneficial therapeutic effects seen in successful natalizumab treatment.
  • the methods, assays, complexes, and devices described herein are validated using reconstructed samples and/or primary patient samples.
  • the assays described herein are evaluated using reconstructed serum samples spiked with natalizumab.
  • samples can be prepared containing increasing concentrations (0 to 200 ug/mL) of natalizumab (or irrelevant mAb) spiked into serum (obtained from at least 10 different individuals) and then incubated with 0.5 mM reduced glutathione (GSH) at 37° C. for 17-24 hours to form monovalent natalizumab.
  • GSH reduced glutathione
  • the intensity of each test line can be determined using an end-point reader at different time intervals (e.g., 5, 10, and 15 minutes) to identify the optimal and shortest assay time.
  • the concentration of bivalent natalizumab and total natalizumab in each sample may also be quantified by enzyme linked immunosorbent assay (ELISA).
  • ELISA enzyme linked immunosorbent assay
  • the total natalizumab concentration can be measured using the competitive assay mimetope peptide-based ELISA assay described herein.
  • quantitation of the bivalent form of natalizumab is determined using a double antigen sandwich ELISA with peptide coated on the bottom of the plate as the capture reagent and peptide conjugated to horseradish peroxidase (HRP) as the detection reagent.
  • HRP horseradish peroxidase
  • two separate test strips are run in parallel in the same cassette to independently measure bivalent and total natalizumab.
  • a single test strip accurately measures both bivalent and total natalizumab in a sample.
  • the natalizumab test line is replaced with anti-human IgG4 Fc to capture both forms of natalizumab via a sandwich format (which may also capture endogenous IgG4 if present).
  • the assays described herein may be cross-validated against a minimum number of primary patient samples (e.g., 20 samples).
  • the samples are isolated from MS patients receiving natalizumab therapy and have known natalizumab concentrations determined by a separate, non-mimetope peptide-based and validated ELISA.
  • the assay time is less than 15 minutes.
  • the assay uses 10-40 uL of serum and 40-200 uL of chase buffer.
  • the assay uses 10-40 uL of whole blood and 40-200 uL of chase buffer.
  • the assay uses 15 uL of serum and 85 uL of chase buffer.
  • the assay uses 15 uL of whole blood and 85 uL of chase buffer.
  • the methods, assays, complexes, and devices described herein comprise a personal point-of-care device.
  • the personal point-of-care device comprises a housing, a display, a test strip holder, a test strip comprising a peptide which binds to the antigen-binding site of natalizumab present in a sample from a user, an imaging device for imaging the test strip, a processor, an onboard memory, and a communications element.
  • the peptide comprises a sequence selected from the group consisting of SEQ ID NOs:1-28.
  • the peptide comprises a sequence selected from the group consisting of SEQ ID NOs:1-30.
  • the peptide comprises a sequence selected from the group consisting of SEQ ID NO:1, 2, 4, 13, 16, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, and 28. In further embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NO:1, 2, 4, 13, 16, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, and 30. In further embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NOs:24, 27, and 28. In further embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NOs:24, 27, 28, 29, and 30.
  • the peptide is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from the group consisting of SEQ ID NO:1-30. In some embodiments, the peptide is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from the group consisting of SEQ ID NO:24, 29 and 30. In some embodiments, the peptide is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO:29. In some embodiments, the peptide is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 30.
  • the personal point-of-care device includes an on-device display.
  • the display is embedded in the face of the device and the test strip holder is encased by the device.
  • the display is not attached to the device.
  • the display color, font, image size, contrast, or contents are user-selected.
  • the display may render various icons or messages to a user, such as test results, device status, or error messages.
  • the personal point-of-care device optionally includes an audio indicator.
  • contents of the on-display device outputs into audio by user-selection.
  • the device is reversibly connected to a mobile device, a computer, a GPS, an IPAD, a USB drive, a printer, a scanner, a television, a server, a car, a smart watch, smart glasses, an IPOD, a game player, a projector, a camera, or similar electronic devices.
  • the device is connected via Bluetooth.
  • the device is reversibly connected to a wearable device, such as a Fitbit®.
  • detection and quantification of natalizumab in human serum is measured using the following peptide-based ELISA protocol.
  • the day before the experiment generate monovalent natalizumab by preparing suspensions of human serum containing 3 mM reduced glutathione (GSH) and different concentrations of natalizumab (256, 128, 64, 32, 16, 8 or 4 ug/mL (and 0)) to create a calibration curve. Incubate samples overnight at 37° C.
  • GSH reduced glutathione
  • the day of the experiment prepare a suspension of the peptide of interest (7.5 ug/mL in 1 ⁇ TBS) and add to a neutravidin coated plate (or TBS only as background control). Incubate peptide on the plate for 1 h at room temperature (RT), then wash 5 times with TBST (1 ⁇ TBS+0.05% Tween20).
  • Mimetope peptides were selected from phage display libraries, some of which contain cysteines flanking the peptide mimetope sequence to increase stability of the peptide through disulfide bond formation. After three rounds of selection with multiple phage display libraries, individual phage plaques were isolated and sequenced. Twenty-three unique phage displayed peptide sequences were identified and are presented in Table 1.
  • Synthetic peptides were supplied as TFA salt at >84% purity confirmed by mass spec and HPLC. Peptides were reconstituted and concentration was determined using a NanoDrop spectrophotometer.
  • Synthesized peptides were validated for specificity by using them as ELISA capture reagents on neutravidin-coated plates. The results are shown in FIG. 1 . Briefly, biotinylated peptides were coated on the wells at 100 ug/mL concentration in either tris buffered saline buffer (TBS) or water. Wells were blocked and then either natalizumab (NTZ) or rituximab (RIT) antibodies spiked in 2.5% BSA/TBST buffer at 1000 ng/mL or 500 ng/mL were added to the wells.
  • TBS tris buffered saline buffer
  • RIT rituximab
  • Antibody that was captured by the peptides was subsequently detected using a goat anti-human IgG-Fc conjugated to horseradish peroxidase (HRP) combined with a colorimetric substrate.
  • HRP horseradish peroxidase
  • Sensitivity of the mimetope peptide for detection of natalizumab in buffer was determined, and the results presented in FIG. 2 .
  • the minimal coating concentration of peptide (NTZ-01) to achieve desired sensitivity in buffer and human serum (0.1%) was determined, as shown in FIG. 3 .
  • Sensitivity of different mimetope peptides (NTZ-01, NTZ-04, and NTZ-05) for detection of natalizumab (monovalent form) in human serum was determined, and the results are presented in FIGS. 4 and 5 .
  • the dynamic range of the assay is between 20 and 240 ng/mL of natalizumab. This allows for the accurate quantification of samples containing between 5 and 60 ug/mL natalizumab after applying the minimum sample dilution required for the assay.
  • a subject undergoing natalizumab therapy has her finger pricked using a lancet.
  • a fixed and predetermined volume of blood (specific for each test) is collected using a transfer pipet and applied to a test device as shown in FIG. 7 .
  • IgG4 antibodies such as natalizumab (NTZ) can exchange Fab-arms with other endogenous IgG4 molecules, leading to the formation of monovalent molecules (hybrid IgG4 molecules with only one arm targeting VLA-4) in addition to the original bivalent form.
  • NTZ natalizumab
  • This test measures both bivalent and total circulating levels of NTZ in the biologic sample.
  • the conjugate pad contains colloidal gold conjugated to mimetope peptide specific for NTZ.
  • the first test line coated with mimetope peptide, captures bivalent NTZ via one Fab arm while the other Fab arm is detected by the mimetope-gold conjugate (i.e., a double antigen sandwich assay).
  • the second test line is coated with NTZ antibody.
  • the mimetope-gold conjugate binds to the NTZ test line and the line develops color. If sufficient NTZ is present in the sample, both bivalent and monovalent forms compete with the NTZ test line for binding to the mimetope-gold conjugate and less color develops on the test line in a dose-dependent manner (i.e., a competitive assay).
  • the independent control line indicates that the device ran properly. The intensity of the color changes are measured and quantified by an end-point reader.
  • This lateral flow immunoassay (LFA) design is able to measure both bivalent and total circulating levels of natalizumab in biologic samples, as shown in FIG. 8 .
  • LFA lateral flow immunoassay
  • both bivalent and monovalent forms compete with the test line for binding to the mimetope-gold conjugate and less color is observed on the test line.
  • the coating densities of the mimetope on the gold, the mimetope on the first test line, and the antibody on the second test line are optimized to enable measurement of bivalent and total natalizumab in the sample to fall within the dynamic range.
  • Test line intensity is quantified by an off-the-shelf end-point digital reader.
  • Natalizumab was incubated with increasing concentrations of natalizumab-specific VERITOPETM (NTZ-01-Bio; SEQ ID NO:24) or irrelevant peptide (1,000 or 10,000 fold molar excess) and binding to its cellular target was measured by flow cytometry. Jeko-1 cells, expressing CD49d, were used as a model. As shows in FIG. 9 , natalizumab-specific VERITOPETM can inhibit binding of natalizumab to its cell surface target.
  • the histogram peaks, from left to right, are: Jeko-1 cells only; x-IgG4-A647; 10,000-fold natalizumab-specific VERITOPETM; 1,000-fold natalizumab-specific VERITOPETM; 1,000-fold irrelevant VERITOPETM; and 10,000-fold irrelevant VERITOPETM.
  • FIGS. 10 A and B illustrates a comparison of VERITOPETM ( FIG. 10 A ) and anti-idiotype ( FIG. 10 B ) ELISA binding.
  • the anti-idiotype antibody (Type 1 or neutralizing) binds to the antigen binding site of natalizumab, specifically recognizes free natalizumab but not free human alpha-4/beta-1 integrin, and is suitable for pharmacokinetic (PK) studies.
  • FIGS. 11 A and B demonstrate that both VERITOPETM ( FIG.
  • FIG. 11 A the natalizumab-specific VERITOPETM is attached to the surface of the plate as the capture reagent for natalizumab (TYSABRITM), and natalizumab-specific VERITOPETM (left side) or the anti-idiotype (right side) at different concentrations is the competitive reagent.
  • FIGS. 12 A and B demonstrate that heat-inactivation of natalizumab alters natalizumab binding to its cellular target. Natalizumab was heat-inactivated by incubation at 60° C. for various periods of time. Alteration of the binding capacity of natalizumab to its cellular target was measured by flow cytometry. Jeko-1 cells, expressing CD49d, were used as a model.
  • FIG. 12 A is the bivalent natalizumab and FIG. 12 B is the arm-exchanged (bispecific) natalizumab.
  • Example 7 VERITOPETM ELISA Specifically Captures Active Drug Compared to Anti-Idiotype ELISA
  • FIGS. 13 A and B depict a comparison between anti-idiotype ELISA ( FIG. 13 A ) and VERITOPETM ELISA ( FIG. 13 B ), demonstrating that VERITOPETM ELISA is more sensitive than anti-idiotype ELISA to changes that affect the binding pocket of natalizumab. Natalizumab was heat-inactivated by incubation at 60° C. for various periods of time. The ability of anti-idiotype and VERITOPETM (NTZ-01-Bio; SEQ ID NO:24) capture reagents to distinguish between active and inactive drug was measured by ELISA. Anti-idiotype ELISA measures both inactive and active drug whereas VERITOPETM ELISA selectively distinguishes & quantifies active drug.
  • the natalizumab-specific VERITOPETM (NTZ-01-Bio; SEQ ID NO:24) was characterized by a CLIA-certified/CAP-accredited lab and the assay specifications are presented in FIG. 14 .
  • the Lower Limit of Quantification (LLOQ) is 2.0 ug/mL natalizumab in undiluted serum, which corresponds to 8 ng/mL after applying the minimum required dilution of 1/250
  • the Upper Limit of Quantification (ULOQ) is 16.0 ug/mL natalizumab in undiluted serum, which corresponds to 64 ng/mL after applying the minimum required dilution of 1/250; the data presented are from 5 independent runs, each sample ran in triplicates (mean+/ ⁇ SD) (Limit of Blank: 0.6 ug/mL; Limit of Detection: 0.8 ug/mL).
  • Spiked serum sample stability of the natalizumab-specific VERITOPETM (NTZ-01-Bio; SEQ ID NO:24) was measured and the results presented in FIG. 15 .
  • Serum samples spiked with 8 ug/mL natalizumab were quantified using the VERITOPETM ELISA after exposure to different storage conditions to determine sample stability. Exposure to room temperature (RT), 4° C. or ⁇ 80° C. for 1, 2 and 8 days did not affect stability.
  • the sensitivity of different mimetope peptides (NTZ-06-Bio, SEQ ID NO:29; NTZ-07-Bio, SEQ ID NO:30; and NTZ-01-Bio, SEQ ID NO:24) for detection of natalizumab in human serum was determined, and the results are presented in FIG. 16 .
  • Experimental conditions used were similar to those described in Example 3, above.
  • the Limit of Blank for NTZ-07-Bio was 85.6+/ ⁇ 41 ng/ml.
  • the selectivity for the natalizumab-specific VERITOPETM (NTZ-07-Bio; SEQ ID NO:30) for natalizumab over other therapeutic monoclonal antibodies was measured and the results presented in FIG. 17 .
  • VERITOPETM selectively captures natalizumab in presence of other circulating human IgG and VERITOPETM does not capture other mAb drugs.

Abstract

Methods and assays for detecting natalizumab in a sample, natalizumab-peptide complexes in a sample, and point-of-care devices for detecting natalizumab in a sample are described herein.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of U.S. application Ser. No. 16/324,878 filed on Feb. 11, 2019, which is a national application of International Application No. PCT/US2017/046,499 filed on Aug. 11, 2017, which claims the benefit of U.S. Provisional Patent Application No. 62/374,217 filed Aug. 12, 2016. Priority is claimed pursuant to 35 U.S.C. § 119. The above noted patent applications are incorporated by reference as if set forth fully herein.
  • STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
  • The inventions described herein were made with the support of the United States government under grants 1R41CA192697-01 and 1R43CA183241-01 awarded by the National Institutes of Health Small Business Innovation Research (NIH-SBIR). The government has certain rights in the disclosed subject matter.
  • 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 Aug. 10, 2017, is named 47085-712_601_SL.txt and is 10,449 bytes in size.
  • BACKGROUND OF THE INVENTION
  • Accurate drug dosing is critical for optimal patient treatment. Actual drug levels vary enormously among people given the same standard dose. Insufficient dosing can result in a poor response to treatment, whereas excessive dosing results in higher costs, wasted resources, and troublesome side effects.
  • Monoclonal antibodies (mAb) and other biologics are targeted therapies that are increasingly being used to treat indications such as cancer and autoimmune disease, such as multiple sclerosis (MS). MS is a leading cause of neurologic disability, and the disease is characterized by multiple inflammatory lesions and demyelination within the white matter of the central nervous system (CNS). Natalizumab (marketed as Tysabri by Biogen Idec) is the top-selling biologic drug indicated for treatment of MS and is being used to treat 25,000 to 50,000 of the 400,000 MS patients in the US.
  • SUMMARY OF THE INVENTION
  • In some embodiments, described herein are methods, assays, complexes, and devices for measuring natalizumab in a sample. In some embodiments, described herein is a method of capturing an antibody in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23, allowing binding of the peptide with the antibody to form an antibody-peptide complex, and detecting the antibody-peptide complex. In some embodiments, the peptide is selected from the group consisting of SEQ ID NOs:1, 2, 4, 13, 16, and 18-23. In some embodiments, the antibody is not complexed to an epitope of a target protein. In some embodiments, the antibody is natalizumab.
  • Also described herein are methods, assays, complexes, and device for monitoring natalizumab in a biological sample obtained from a subject. In some embodiments, the biological sample is selected from the group consisting of body fluids, tissues, body swabs, and body smears. In some embodiments, the biological sample is a fluid. In some embodiments, the fluid contains antibody at a concentration of between about 0.5 mcg/mL to 120 mcg/mL. In some embodiments, the fluid is selected from the group consisting of serum, plasma, whole blood, red blood cell concentrates, platelet concentrates, leukocytes concentrates, urine, cerebral spinal fluid, and sputum. In some embodiments, the biological sample is obtained from a human.
  • In some embodiments, described herein is a method of capturing natalizumab in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23, wherein the peptide is attached to a solid support. In some embodiments, the peptide binds to the antigen binding site of the antibody. In further embodiments, described herein is a method of capturing an antibody in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23, allowing binding of the peptide with the antibody to form an antibody-peptide complex, and detecting the antibody-peptide complex, wherein detection of the antibody-peptide complex is performed by detection of a detectable label on the antibody or the peptide. In further embodiments, detection of the antibody-peptide complex is performed by Western blot analysis, dot blot analysis, flow cytometry, enzyme-linked immunosorbent assay (ELISA), lateral flow immunoassay, radioimmunoassay (RIA), competition immunoassay, dual antibody sandwich assay, chemiluminescent assay, bioluminescent assay, fluorescent assay, or agglutination assay.
  • Also described herein is a natalizumab-mimetope complex comprising: a mimetope comprising a peptide between 7 and 26 amino acids long; and natalizumab. In some embodiments, the peptide is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23.
  • Also described herein is a test device comprising: a sample pad for receiving a biologic; a conjugate pad; and a test membrane comprising at least one test line comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane. In some embodiments, the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane and an antibody specific for natalizumab. In further embodiments, the antibody specific for natalizumab binds natalizumab at a variable region. In other embodiments, the antibody specific for natalizumab binds natalizumab at a constant region. In some embodiments, the test membrane further comprises at least one test line comprising an antibody specific for the natalizumab-binding mimetope peptide. In some embodiments, the antibody is natalizumab or a biosimilar thereof or a new antibody specific for the selected peptide obtained by conventional methods, such as animal immunization using the selected peptide as an immunogen.
  • Also described herein is a method of preventing progressive multifocal leukoencephalopathy (PML) comprising: identifying a subject receiving antibody therapy at risk of developing PML; obtaining a biological fluid from the subject; contacting the biological fluid with a test device comprising: a sample pad for receiving the biological fluid; a conjugate pad; and a test membrane comprising at least one test line comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23; and, increasing the frequency of performing the method steps based on the results obtained following the step of contacting the biological fluid with a test device. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane. In some embodiments, the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane and an antibody specific for natalizumab. In some embodiments, the antibody specific for natalizumab binds natalizumab at a variable region. In some embodiments, the antibody specific for natalizumab binds natalizumab at a constant region. In some embodiments, the test membrane further comprises at least one test line comprising an antibody specific for the natalizumab-binding mimetope peptide. In some embodiments, the antibody is natalizumab or a biosimilar thereof or a new antibody specific for the selected peptide obtained by conventional methods, such as animal immunization using the selected peptide as an immunogen. In some embodiments, the step of increasing the frequency of performing the method steps further comprises adjusting the antibody therapy based on the results obtained following the step of contacting the biological fluid with a test device.
  • Also described herein is a natalizumab-binding mimetope comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23. Also described herein is a method of detecting natalizumab in a biological sample comprising: contacting said biological sample with a natalizumab mimetope; allowing binding of said natalizumab mimetope with the natalizumab to form a natalizumab-natalizumab mimetope complex; and detecting the natalizumab-natalizumab mimetope complex. In some embodiments, the natalizumab mimetope is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 96% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 97% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 98% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 99% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is 100% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23.
  • In some embodiments, described herein are methods, assays, complexes, and devices for measuring active natalizumab in a sample. In some embodiments, described herein is a method of capturing an antibody in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30, allowing binding of the peptide with the antibody to form an antibody-peptide complex, and detecting the antibody-peptide complex. In some embodiments, the peptide is selected from the group consisting of SEQ ID NOs:24, 29, and 30. In some embodiments, the antibody is not complexed to an epitope of a target protein. In some embodiments, the antibody is natalizumab. In some embodiments the antibody is active natalizumab
  • In some embodiments, described herein is a method of capturing active natalizumab in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30, wherein the peptide is attached to a solid support. In some embodiments, the peptide binds to the antigen binding site of the active natalizumab. In further embodiments, described herein is a method of capturing an antibody in a sample comprising contacting a sample with a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30, allowing binding of the peptide with the antibody to form an antibody-peptide complex, and detecting the antibody-peptide complex, wherein detection of the antibody-peptide complex is performed by detection of a detectable label on the antibody or the peptide. In further embodiments, detection of the antibody-peptide complex is performed by Western blot analysis, dot blot analysis, flow cytometry, enzyme-linked immunosorbent assay (ELISA), lateral flow immunoassay, radioimmunoassay (RIA), competition immunoassay, dual antibody sandwich assay, chemiluminescent assay, bioluminescent assay, fluorescent assay, or agglutination assay.
  • Also described herein is a natalizumab-mimetope complex comprising: a mimetope comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30; and active natalizumab.
  • Also described herein is a test device comprising: a sample pad for receiving a biologic; a conjugate pad; and a test membrane comprising at least one test line comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane. In some embodiments, the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane and an antibody specific for natalizumab. In further embodiments, the antibody specific for natalizumab binds natalizumab at a variable region. In other embodiments, the antibody specific for natalizumab binds natalizumab at a constant region. In some embodiments, the test membrane further comprises at least one test line comprising an antibody specific for the natalizumab-binding mimetope peptide. In some embodiments, the antibody is natalizumab or a biosimilar thereof or a new antibody specific for the selected peptide obtained by conventional methods, such as animal immunization using the selected peptide as an immunogen.
  • Also described herein is a method of preventing progressive multifocal leukoencephalopathy (PML) comprising: identifying a subject receiving antibody therapy at risk of developing PML; obtaining a biological fluid from the subject; contacting the biological fluid with a test device comprising: a sample pad for receiving the biological fluid; a conjugate pad; and a test membrane comprising at least one test line comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30; and, increasing the frequency of performing the method steps based on the results obtained following the step of contacting the biological fluid with a test device. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane. In some embodiments, the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab. In some embodiments, the conjugate pad comprises a detection reagent conjugated to a peptide at least 95% identical to the peptide in the test membrane and an antibody specific for natalizumab. In some embodiments, the antibody specific for natalizumab binds natalizumab at a variable region. In some embodiments, the antibody specific for natalizumab binds natalizumab at a constant region. In some embodiments, the test membrane further comprises at least one test line comprising an antibody specific for the natalizumab-binding mimetope peptide. In some embodiments, the antibody is natalizumab or a biosimilar thereof or a new antibody specific for the selected peptide obtained by conventional methods, such as animal immunization using the selected peptide as an immunogen. In some embodiments, the step of increasing the frequency of performing the method steps further comprises adjusting the antibody therapy based on the results obtained following the step of contacting the biological fluid with a test device.
  • Also described herein is a natalizumab-binding mimetope comprising a peptide at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. Also described herein is a method of detecting active natalizumab in a biological sample comprising: contacting said biological sample with a natalizumab mimetope; allowing binding of said natalizumab mimetope with the natalizumab to form a natalizumab-natalizumab mimetope complex; and detecting the natalizumab-natalizumab mimetope complex. In some embodiments, the natalizumab mimetope is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 96% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 97% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 98% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 99% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is 100% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph showing the specificity of natalizumab (NTZ) mimetope peptides for the detection of natalizumab by enzyme-linked immunosorbent assay (ELISA).
  • FIG. 2 is a graph showing the sensitivity of a natalizumab (NTZ) mimetope peptide-based enzyme-linked immunosorbent assay (ELISA) for natalizumab spiked in 2.5% BSA/TBST buffer. NTZ-01-Bio and NTZ-02-Bio peptides were coated at 25 ug/mL or 50 ug/mL in TBS.
  • FIGS. 3A-3C are graphs showing a titration of the NTZ-01-Bio peptide coating concentration and calibration curves of natalizumab prepared in buffer (TBST-2.5% BSA) and human serum (0.1%).
  • FIGS. 4A and 4B are graphs showing the sensitivity and specificity of the NTZ-01-Bio mimetope peptide-based ELISA for natalizumab spiked in human serum (0.1%). FIG. 4A presents results from 2 independent experiments (mean+/−SD). The dashed line indicates OD without natalizumab present in the samples. FIG. 4B depicts results with all wells coated with 7.5 ug/mL NTZ-01-Bio (except the no coating control). Natalizumab was spiked at 2560 ng/mL and the other therapeutic monoclonal antibodies were spiked at 2000 ng/mL in human serum (0.1%).
  • FIG. 5 is a graph illustrating the sensitivity of the NTZ-01-Bio, NTZ-04-Bio, and NTZ-05-Bio mimetope peptide-based ELISAs for natalizumab spiked in human serum (0.4%). Results from a single experiment are presented. All wells were coated with 9 uM peptide except no coating control. The calibration curve was prepared using natalizumab-spiked human serum and exposed to 3 mM GSH for 18 hours at 37° C. to generate monovalent antibody.
  • FIGS. 6A and 6B are graphs illustrating the detection of the monovalent form of natalizumab in human serum (0.4%) using the mimetope peptide-based ELISA (NTZ-01-Bio). FIG. 6A depicts a calibration curve prepared using natalizumab-spiked human serum that is exposed to 3 mM GSH for 18 hours at 37° C. to generate monovalent antibody. (mean+/−SD). The absence of analyte provides an OD of 0.006+/−0.003 (not depicted; (n=4; mean+/−SD). FIG. 6B depicts a precision profile determined by spiking predetermined amount of natalizumab in human-serum followed by treatment with GSH, as above. The samples were run in 6 replicates and the coefficient of variation (CV) was determined by dividing the standard deviation by the mean×100. A CV below 20% is obtained for natalizumab concentrations between 16 and 1024 ng/mL (n=4).
  • FIG. 7 depicts an exemplary lateral flow immunoassay (LFA) device for rapid measurement of natalizumab levels in a finger-stick blood sample.
  • FIG. 8 depicts detection of free natalizumab levels using a combination of sandwich and competitive assays in the same lateral flow immunoassay (LFA).
  • FIG. 9 depicts a flow cytometry histogram of binding between natalizumab and its cellular target, following incubation of natalizumab with varying concentrations of either a natalizumab-specific VERITOPE™ or an irrelevant peptide. Jeko-1 cells, expressing CD49d, were used as a model.
  • FIGS. 10A and B depicts a graphical comparison of a natalizumab-specific VERITOPE™ (FIG. 10A) and anti-idiotype (FIG. 10B) ELISA binding.
  • FIGS. 11A and B illustrate the characterization of a natalizumab-specific VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24) and an anti-idiotype. In FIG. 11A natalizumab-specific VERITOPE™ is the capture reagent and anti-idiotype is the competitive reagent. In FIG. 11B, anti-idiotype is the capture reagent and natalizumab-specific VERITOPE™ is the competitive reagent.
  • FIGS. 12A and B demonstrate that heat-inactivation of natalizumab alters bivalent natalizumab (FIG. 12A) and arm-exchanged (bispecific) natalizumab (FIG. 12B) binding to its cellular target.
  • FIGS. 13A and B depict a comparison between anti-idiotype ELISA (FIG. 13A) and natalizumab-specific VERITOPE™ (FIG. 13B; NTZ-01-Bio; SEQ ID NO:24).
  • FIG. 14 depicts natalizumab-specific VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24) ELISA assay calibration curve.
  • FIG. 15 depicts stability of human serum samples spiked with a natalizumab-specific VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24) up to 8 days.
  • FIG. 16 depicts a comparison of the calibration curves for NTZ01 (SEQ ID NO:24), NTZ06 (SEQ ID NO:29), and NTZ07 (SEQ ID NO:30) natalizumab-specific VERITOPES™.
  • FIG. 17 depicts the specificity of natalizumab-specific VERITOPE™ NTZ07 (SEQ ID NO:30) compared to 4 other monoclonal antibodies.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Accurate drug dosing is critical for optimal patient treatment. There is wide variability in the actual drug levels among patients given the same standard dose. Further, the traditional model of medical sample analysis involves centralized laboratories, where tests are performed but results are delayed for hours or days. As such, medical professionals and their patients have a strong interest in obtaining precise, personalized, point-of-care diagnosis.
  • One example of a therapy that requires accurate drug dosing is natalizumab (marketed as Tysabri by Biogen Idec). Natalizumab is one of the most effective treatments available to reduce relapse frequency in multiple sclerosis (MS) patients. Like most mAbs, natalizumab displays highly variable pharmacokinetics (PK) across patients, compounded by the standard dosing that is not body mass or surface area adjusted. Natalizumab is the top selling biologic drug indicated for treatment of MS and is being used to treat 25,000-50,000 of the 400,000 MS patients in the US and also used in the treatment of Crohn's disease. Natalizumab is a humanized recombinant mAb that targets the α4 chain of α4β1 integrin (also known as very late activation antigen 4; VLA-4) and α4β7 integrin and is thought to function by blocking migration of immune cells across the blood-brain barrier into the central nervous system (CNS), thus suppressing inflammation in patients with relapsing-remitting multiple sclerosis (Vennegoor A, et al. Clinical relevance of serum natalizumab concentration and anti-natalizumab antibodies in multiple sclerosis. Mult Scler [Internet]. 2013; 19(5):593-600; Polman C H, et al., A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. The New England Journal of Medicine. 2006 2; 354(9):899-910). Natalizumab is a full-length antibody of the IgG4 subclass and consists of two heavy and two light chains connected by four inter-chain disulfide bonds. Like other IgG4 antibodies, natalizumab demonstrates reduced binding to Fc gamma receptors and a lack of ability to fix complement in vitro. As a result, natalizumab can block interaction of α4-integrins with their cognate receptors with minimal cell killing.
  • Unfortunately, the cost associated with treating chronic diseases such as MS can be considerable. Natalizumab, given by infusion, is very expensive and costs approximately $4,000-5,000 for a single dose, leading to an annual cost of close to $65,000 if the drug is taken every 4 weeks as recommended on the label. Furthermore, the immunosuppressive activity of natalizumab has been associated with reawakening of JC polyomavirus, which may lead to progressive multifocal leukoencephalopathy (PML), a serious and often-fatal opportunistic brain infection. Approximately 55% of MS patients are positive for anti-JC virus antibodies, which puts them at increased risk for developing PML while on natalizumab. The estimated incidence of PML is 1:1,000 after a median of 18 months of treatment, and the mortality rate of PML patients is close to 25%, and most of the survivors of PML have permanent residual brain damage. Restoring immune function by accelerating the removal of natalizumab from the body is the only intervention for PML with demonstrated efficacy (Khatri B O, et al. Effect of plasma exchange in accelerating natalizumab clearance and restoring leukocyte function. Neurology. 2009; 72:402-9). Reducing the dose of natalizumab or increasing infusion intervals could mitigate the risk of developing PML in susceptible patients (Planas R, et al. Long-term safety and efficacy of natalizumab in relapsing-remitting multiple sclerosis: impact on quality of life. Patient Relat Outcome Meas [Internet]. 2014; 5:25-33).
  • The standard dosing regimen for natalizumab is 300 mg by IV infusion every 4 weeks. The current dosing strategy for natalizumab is not customized for each patient, and there is growing evidence of considerable variability in the rate at which different patients clear the drug from their bodies. A 2011 study by Foley reported that patients with MS who were receiving monthly doses of natalizumab exhibited patient-to-patient variability in their serum natalizumab levels. Furthermore, natalizumab was found to accumulate in the serum in some patients who did not clear the drug within the 4-week period (Foley J. Progressive escalation of natalizumab serum concentration as a potential kinetic marker for PML risk assessment. Oral communication, abstract S51.004, April 2011. American Academy of Neurology. 2011; Bomprezzi R, et al. Extended interval dosing of natalizumab: a two-center, 7-year experience. Ther Adv Neurol Disord [Internet]. 2014; 7(5):227-31). Similarly, integrin saturation by natalizumab at the end of 4 weeks has been reported to range from 80% (Miller D H et al., A controlled trial of natalizumab for relapsing multiple sclerosis, New Eng J Med, 2003) to less than 40% (Hyams J S, et al. Natalizumab therapy for moderate to severe crohn disease in adolescents. J Pediatr Gastroenterol Nutr [Internet]. 2007; 44(2):185-91). Research indicates that patients with low body weight may be receiving excessive drug, which could place them at higher risk for PML (Foley J, et al., Low body weight as a potential surrogate risk factor for progressive multifocal leukoencephalopathy. In: Pulst S, editor. The 66th Annual Meeting of American Academy of Neurology. 2014. p. P2-244). Patients with higher levels of free, circulating natalizumab may have increased risk of PML, which could be reduced through dose extension schedules. An extended dosing schedule of 300 mg every 6 to 8 weeks has been suggested as one way to maintain the efficacy of natalizumab while reducing exposure to the drug, and thereby reducing the risk for PML (Bomprezzi R, et al. Extended interval dosing of natalizumab: a two-center, 7-year experience. Ther Adv Neurol Disord [Internet]. 2014; 7(5):227-31; see also, Zhovtis R., et al. Extended interval dosing of natalizumab in multiple sclerosis. J Neurol Neurosurg Psychiatry 2016; 87(8):885-9). Both the clinical literature and discussions with neurologists have highlighted the importance of frequent monitoring of natalizumab serum concentrations in individual patients in order to maximize drug efficacy and minimize risk for PML. However, there is currently no widely available assay enabling the measurement of natalizumab serum levels in treated patients, such as a routine, fast, easy-to-use, and inexpensive point-of-care lateral flow immunoassay for rapid measurement of natalizumab levels in a finger-stick blood sample.
  • Peptide-based immunoassays can be developed for monitoring mAb levels. Phage displayed peptide libraries are used to select peptide sequences that mimic the target antigen of a given mAb. Peptide libraries displayed on bacteriophage are routinely used to identify peptide epitopes, or mimetopes (also referred to as VERITOPES™), recognized by antibodies. Phage display works best with concentrated and highly purified proteins, and as such therapeutic mAb are ideal targets. When short peptides, 7 to 26 amino acids long, are screened, the selected peptides almost invariably bind to the antigen-binding site of the antibody and are competed by the natural ligand (Sanchez A B, et al. A general process for the development of peptide-based immunoassays for monoclonal antibodies. Cancer Chemother Pharmacol [Internet]. 2010/01/21 ed. 2010; 66(5):919-25). These mimetope peptides are then optionally used as capture or detection reagents in ELISA or other solid phase immunoassays such as lateral flow immunoassay (LFA) as long as the density of the peptide is sufficient to enable multivalent binding avidity to compensate for the moderate affinity. Mimetope peptides may also be selected from a library that contains cysteines flanking the peptide mimetope sequence to increase the stability of the peptide through disulfide bond formation. Described herein are methods, assays, complexes, and devices that incorporate mimetope peptide reagents selected for specific binding to natalizumab and assays (including LFAs and ELISAs) that implement these peptides for the capture of natalizumab from solution including biological samples for the purpose of measuring circulating natalizumab concentrations in patients for dose monitoring applications.
  • In some embodiments, the methods, assays, complexes, and devices described herein are useful for monitoring the level of mAb in a subject. In some embodiments, the level of mAb is monitoring using a biological sample obtained from a subject. In some embodiments, the subject is a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult, or senior adult)) and/or other non-human animals, for example, mammals (e.g., primates (e.g., cynomolgus monkeys, rhesus monkeys); commercially relevant or research mammals (such as cattle, pigs, horses, sheep, goats, dogs, cats, rabbits, rats, and/or mice); and birds (e.g., commercially relevant birds such as chickens, ducks, geese, and/or turkeys).
  • One advantage of the methods, assays, complexes, and devices described herein is that they provide means to detect, monitor, and record drug levels in a patient with precision directly at the point-of-care. The drugs can be novel or biosimilar drugs, and the technology is based on the binding of a mimetope to an antibody. Mimetopes are peptides that specifically detect a given biological drug (novel or biosimilar) in a biologic fluid. Mimetopes are described in U.S. Pat. No. 9,250,233, which is hereby incorporated in full by reference. The tests are optionally performed with a disposable, integrated point-of-care device, or performed with a laboratory-based enzyme-linked immunosorbent assay (ELISA), or performed with a fixed reader, as a tabletop system connected to a computer. Devices and systems for use with mimetopes include those performed with a personal point-of-care device. In some embodiments, the personal point-of-care device comprises a housing, a display, a test strip holder, a test strip comprising a peptide which binds to an antigen-binding site of an antibody present in a sample from a user, an imaging device for imaging the test strip, a processor, an onboard memory, and a communications element. In some embodiments, the personal point-of-care device includes an on-device display. In some embodiments, the display is embedded in a face of the device and the test strip holder is encased by the device. In some embodiments, the display is not attached to the device. In some embodiments, the display color, font, image size, contrast, or contents are user-selected. In some embodiments, the display may render various icons or messages to a user, such as test results, device status, or error messages. In some embodiments, the personal point-of-care device optionally includes an audio indicator or a light indicator. In further embodiments, contents of the on-display device outputs into audio by user-selection. In some embodiments, the device is reversibly connected to a mobile device, a computer, a GPS, an IPAD, a USB drive, a printer, a scanner, a television, a server, a car, a smart watch, smart glasses, an IPOD, a game player, a projector, a camera, or similar electronic devices.
  • The methods, assays, complexes, and devices described herein provide benefit to the patient, the medical professional, the pharmaceutical company, and the insurance company. The patient benefits by receiving the optimal dose of natalizumab (avoiding high cost and adverse side effects associated with excessive dosing such as PML, or poor response to treatment associated with insufficient dosing). The medical professional benefits with knowledge that a patient has a specific drug level. The medical professional has the information to adjust the dose accurately in order to increase or decrease the patient's response to natalizumab. The pharmaceutical company benefits by saving costs from not having natalizumab in limited supply squandered due to waste or inefficiency. Finally, the insurance company benefits by not having to purchase additional natalizumab beyond the amount needed for optimal dosing of a patient. Further, therapeutic dose monitoring of natalizumab with the methods, assays, complexes and devices described herein will enable the identification of patients who have excessive free drug levels producing potentially elevated VLA-4 receptor saturations with reduced or absent immune cell trafficking and are candidates for dose extension. The methods, assays, complexes and devices described herein are optionally also used to identify patients with sub-therapeutic drug levels and may indicate development of anti-drug antibodies. Different therapeutic interventions are performed on patients having lower drug levels (identified via the methods, assays, complexes, and devices described herein), such as increasing the dose, or frequency of dosing, to maximize therapeutic benefit, or rapidly switching to another treatment approach more likely to succeed. On the other hand, patients for which drug levels may be well above the range required for optimal clinical benefit, as identified by the methods, assays, complexes, and devices described herein, could have subsequent treatments delayed, reducing the risk of PML without affecting outcomes, or minimally be placed under closer medical monitoring based on the risk associated with elevated drug levels.
  • Certain Terminology
  • Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Any reference to “or” herein is intended to encompass “and/or” unless otherwise stated. As used herein, the term ‘about’ a number refers to that number plus or minus 10% of that number. The term ‘about’ a range refers to that range minus 10% of its lowest value and plus 10% of its greatest value.
  • In some embodiments, the methods, assays, complexes, and devices described herein comprise sample pads, conjugate pads, colloidal gold conjugates (or other colored or fluorescent monodispersed types of particle conjugates such as those made of latex), test lines, and/or membranes. In some embodiments, the sample pads are pretreated with blocking agents and surfactants to improve flow and subsequent release of agents from the conjugate pad. In the case of blood samples, the sample pad may also include a filter that removes cells while allowing serum or plasma to flow through to the membrane. The colloidal gold may be a monodisperse and uniform solution of 40 nm colloidal gold prepared by a reduction of aqueous HAuCl4 with a cherry red appearance. For detection of test lines, mimetope peptide may be conjugated to the gold colloids by passive adsorption, and the coating density of the mimetope on the gold may be optimized to enable measurement of the proportion of bivalent natalizumab in a sample. Conjugate pad parameters, including flow rate, release characteristics, and stability, may be optimized to enable measurement of the proportion of bivalent natalizumab in a sample. Pretreatment of a conjugate pad with blocking and/or stabilizing buffers can improve these parameters. In some embodiments, colloidal gold conjugates are dried onto the pre-treated conjugate pads and are able to return to solution when sample is present.
  • In some embodiments, the methods, assays, complexes, and devices comprise a test line comprising a mimetope peptide specific for natalizumab that serves as the first test line to capture bivalent antibody. In some embodiments, mimetope peptides are synthesized and attached to bovine serum albumin (BSA) which greatly improves adsorption of the peptide onto the membrane. In some embodiments, natalizumab (or another antibody that specifically captures the natalizumab peptide) serves as the second test line for the competitive assay portion of the test and allows measurement of both monovalent and bivalent natalizumab. The mimetope test line concentration and the natalizumab test line concentration will be varied to optimize the quantitative feature of the test. In some embodiments, the mimetope test line concentration or natalizumab test line concentration is between 0.1 mg/mL and 2.5 mg/mL. In further embodiments, the mimetope test line concentration or natalizumab test line concentration is selected from the group consisting of 0.1 mg/mL, 0.2 mg/mL, 0.3 mg/mL, 0.4 mg/mL, 0.5 mg/mL, 0.6 mg/mL, 0.7 mg/mL, 0.8 mg/mL, 0.9 mg/mL, 1.0 mg/mL, 1.2 mg/mL, 1.4 mg/mL, 1.6 mg/mL, 1.8 mg/mL, 2.0 mg/mL, 2.2 mg/mL, 2.4 mg/mL, 2.6 mg/mL, 2.8 mg/mL, 3.0 mg/mL, 3.2 mg/mL, 3.4 mg/mL, 3.6 mg/mL, 3.8 mg/mL, 4.0 mg/mL, 4.2 mg/mL, 4.4 mg/mL, 4.6 mg/mL, 4.8 mg/mL, 5.0 mg/mL, 5.2 mg/mL, 5.4 mg/mL, 5.6 mg/mL, 5.8 mg/mL, and 6.0-13-mg/mL. In some embodiments, nitrocellulose membranes are used. Alternative membrane materials may also be used. The optimal membrane may is determined empirically by testing 5-10 different forms, varied by pore size and wicking rate. Test lines may be striped onto the membrane using an automated programmable dispenser. Membranes may be blocked to reduce non-specific binding, and blocking can influence the wicking rate.
  • In some embodiments, the assays described herein are shelf-stable for at least two years. Components that contribute to shelf life include the stability of the reagents, such as antibodies and peptides, as well as the physical components of the assay. Assembled assays may be stored in foil pouches with a desiccant. Long term storage tests and exposure to non-optimal conditions of the assays described herein may be performed via accelerated stability testing by varying temperature, humidity and light. In some embodiments, sample assays are incubated at 4° C., room temperature, 37° C., and 55° C., and tested at various times. One week at 55° C. can simulate one year at room temperature. Humidity testing may be performed with both open and closed packages at 30% and 80% relative humidity at room temperature and 37° C. Light exposure can be performed for several weeks.
  • In some embodiments, the methods, assays, complexes, and devices described herein comprise specific mimetope peptide sequences for the capture and quantification of free and active natalizumab in human serum. In some embodiments, the mimetope peptide specific for natalizumab is selected from the group consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 96% identical to a peptide selected from the group consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 97% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 98% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is at least 99% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the natalizumab mimetope is 100% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 1-23. In some embodiments, the mimetope peptide specific for natalizumab is selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 95% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 96% identical to a peptide selected from the group consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 97% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 98% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is at least 99% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. In some embodiments, the natalizumab mimetope is 100% identical to a peptide selected from the group consisting of consisting of SEQ ID NOs: 24-30. Biotinylated peptides are optionally attached to streptavidin coated plates and used as a surrogate ligand to capture natalizumab in immunoassays. In one embodiment, an enzyme linked immunosorbent assay (ELISA) with a calibration range from 20 to 240 ng/mL (after the minimum required sample dilution) is described, corresponding to a concentration range from 5-60 ug/mL natalizumab in undiluted human serum. Such an ELISA can have an intra- and inter-assay coefficient of variations ranging from 1.0 to 7.9% and from 4.2% to 18.9%, respectively. In some embodiments, described herein is a lateral flow immunoassay with a lower bound of detection of 10 ug/mL and an upper bound of detection of 100 ug/mL.
  • In some embodiments, active natalizumab is distinguished from denatured or inactive natalizumab using the methods, assays, complexes, and devices disclosed. Active natalizumab, as used herein, is able to bind to alpha-4 integrin and trigger downstream events, including preventing passage of immune cells, such as white blood cells, across blood vessel walls into affected organs, such as the brain, spinal cord, and bowel. Natalizumab may be denatured or inactivated by factors such as heat, high or low pH, exposure to organic solvents, length of time, enzymes, oxidizing agents, other stress conditions, or post-translational modifications, such as but not limited to: asparagine deamidation, aspartate isomerization, methionine oxidation, and lysine glycation. Denatured or inactive natalizumab may still bind to anti-idiotype antibodies, as discussed below, but will not lead to the beneficial therapeutic effects seen in successful natalizumab treatment.
  • In some embodiments, the methods, assays, complexes, and devices described herein are validated using reconstructed samples and/or primary patient samples. In some embodiments, the assays described herein are evaluated using reconstructed serum samples spiked with natalizumab. To mimic the in vivo Fab-exchange behavior of natalizumab, samples can be prepared containing increasing concentrations (0 to 200 ug/mL) of natalizumab (or irrelevant mAb) spiked into serum (obtained from at least 10 different individuals) and then incubated with 0.5 mM reduced glutathione (GSH) at 37° C. for 17-24 hours to form monovalent natalizumab. These suspensions can be applied to the assays (n=5 for each concentration) to determine dynamic range, sensitivity, and specificity. The intensity of each test line can be determined using an end-point reader at different time intervals (e.g., 5, 10, and 15 minutes) to identify the optimal and shortest assay time. The concentration of bivalent natalizumab and total natalizumab in each sample may also be quantified by enzyme linked immunosorbent assay (ELISA). In some embodiments, the total natalizumab concentration can be measured using the competitive assay mimetope peptide-based ELISA assay described herein. In some embodiments, quantitation of the bivalent form of natalizumab is determined using a double antigen sandwich ELISA with peptide coated on the bottom of the plate as the capture reagent and peptide conjugated to horseradish peroxidase (HRP) as the detection reagent. In some embodiments, two separate test strips are run in parallel in the same cassette to independently measure bivalent and total natalizumab. In other embodiments, a single test strip accurately measures both bivalent and total natalizumab in a sample. In other embodiments, the natalizumab test line is replaced with anti-human IgG4 Fc to capture both forms of natalizumab via a sandwich format (which may also capture endogenous IgG4 if present).
  • In some embodiments, the assays described herein may be cross-validated against a minimum number of primary patient samples (e.g., 20 samples). In some embodiments, the samples are isolated from MS patients receiving natalizumab therapy and have known natalizumab concentrations determined by a separate, non-mimetope peptide-based and validated ELISA. In some embodiments, the assay time is less than 15 minutes. In some embodiments, the assay uses 10-40 uL of serum and 40-200 uL of chase buffer. In some embodiments, the assay uses 10-40 uL of whole blood and 40-200 uL of chase buffer. In some embodiments, the assay uses 15 uL of serum and 85 uL of chase buffer. In other embodiments, the assay uses 15 uL of whole blood and 85 uL of chase buffer.
  • In some embodiments, the methods, assays, complexes, and devices described herein comprise a personal point-of-care device. In some embodiments, the personal point-of-care device comprises a housing, a display, a test strip holder, a test strip comprising a peptide which binds to the antigen-binding site of natalizumab present in a sample from a user, an imaging device for imaging the test strip, a processor, an onboard memory, and a communications element. In some embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NOs:1-28. In some embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NOs:1-30. In further embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NO:1, 2, 4, 13, 16, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, and 28. In further embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NO:1, 2, 4, 13, 16, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, and 30. In further embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NOs:24, 27, and 28. In further embodiments, the peptide comprises a sequence selected from the group consisting of SEQ ID NOs:24, 27, 28, 29, and 30. In some embodiments, the peptide is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from the group consisting of SEQ ID NO:1-30. In some embodiments, the peptide is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from the group consisting of SEQ ID NO:24, 29 and 30. In some embodiments, the peptide is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO:29. In some embodiments, the peptide is at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to SEQ ID NO: 30.
  • In some embodiments, the personal point-of-care device includes an on-device display. In some embodiments, the display is embedded in the face of the device and the test strip holder is encased by the device. In some embodiments, the display is not attached to the device. In some embodiments, the display color, font, image size, contrast, or contents are user-selected. In some embodiments, the display may render various icons or messages to a user, such as test results, device status, or error messages. In some embodiments, the personal point-of-care device optionally includes an audio indicator. In further embodiments, contents of the on-display device outputs into audio by user-selection. In some embodiments, the device is reversibly connected to a mobile device, a computer, a GPS, an IPAD, a USB drive, a printer, a scanner, a television, a server, a car, a smart watch, smart glasses, an IPOD, a game player, a projector, a camera, or similar electronic devices. In some embodiments, the device is connected via Bluetooth. In some embodiments, the device is reversibly connected to a wearable device, such as a Fitbit®.
  • While preferred embodiments have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. It should be understood that various alternatives to the embodiments described herein may be employed in practicing the inventions described herein.
  • EXAMPLES
  • The following illustrative examples are representative of embodiments of the methods, assays, complexes, and devices described herein and are not meant to be limiting in any way.
  • Example 1: Detection and Quantification of Natalizumab in Human Serum by Veritope™ Peptide-Based ELISA
  • In one embodiment, detection and quantification of natalizumab in human serum is measured using the following peptide-based ELISA protocol. The day before the experiment, generate monovalent natalizumab by preparing suspensions of human serum containing 3 mM reduced glutathione (GSH) and different concentrations of natalizumab (256, 128, 64, 32, 16, 8 or 4 ug/mL (and 0)) to create a calibration curve. Incubate samples overnight at 37° C.
  • The day of the experiment: prepare a suspension of the peptide of interest (7.5 ug/mL in 1×TBS) and add to a neutravidin coated plate (or TBS only as background control). Incubate peptide on the plate for 1 h at room temperature (RT), then wash 5 times with TBST (1×TBS+0.05% Tween20).
  • Block nonspecific binding to the plate with the addition of 5% goat serum, then incubate 1 h at RT.
  • Use the monovalent natalizumab suspensions prepared in GSH-serum in the first step above as calibrators. Prepare 1/250 dilutions in dilution buffer (2.5% BSA-TBST) to obtain a calibration curve ranging from 1024 to 16 ng/mL natalizumab.
  • Wash wells 5 times with TBST and add samples in triplicate to the appropriate peptide-coated wells and to uncoated wells as background control for each sample. Incubate samples for 1 h at RT, and then wash 5 times with TBST. Add HRP-conjugated mouse monoclonal anti-human IgG4 Fc diluted 1:2000 in 1×TBST to the wells, then incubate for 30 min at RT.
  • Wash wells 10 times with TBST, then add TMB substrate. After a 5 minute incubation at RT, stop the reaction with 1 M H2SO4 and immediately measure optical density at 450 nm using a plate reader.
  • Example 2: Identification of Mimetope Sequences
  • Mimetope peptides were selected from phage display libraries, some of which contain cysteines flanking the peptide mimetope sequence to increase stability of the peptide through disulfide bond formation. After three rounds of selection with multiple phage display libraries, individual phage plaques were isolated and sequenced. Twenty-three unique phage displayed peptide sequences were identified and are presented in Table 1.
  • TABLE 1
    List of Natalizumab Peptide
    Sequences Identified by Phage Display
    NEB PhD C7C Library
    ACPMNESKFCGGG (SEQ ID NO: 1)
    ACPSNPSKFCGGG (SEQ ID NO: 2)
    ACNWMINKECGGG (SEQ ID NO: 3)
    ACPKNPNKFCGGG (SEQ ID NO: 4)
    ACVPSKPGLCGGG (SEQ ID NO: 5)
    NEB PhD 12 Library
    NFLGAVAKGAIHGGG (SEQ ID NO: 6)
    HASWLGSSSNVRGGG (SEQ ID NO: 7)
    TAMASTSTMLQHGGG (SEQ ID NO: 8)
    HFINVSGLATVFGGG (SEQ ID NO: 9)
    RDYHPRDHTATWGGG (SEQ ID NO: 10)
    QMAMEQTNADYQGGG (SEQ ID NO: 11)
    LPTNESSPKGSNGGG (SEQ ID NO: 12)
    QTLNHSWLHTFIGGG (SEQ ID NO: 13)
    VSRPAETTPRLTGGG (SEQ ID NO: 14)
    Custom 7C7C7 Library
    SPFHSPRCGTANSYSCLHMKITSGGG (SEQ ID NO: 15)
    IYAAYPPCPQNLSKFCRHSSSPGGGG (SEQ ID NO: 16)
    VENPWNQCMKGTFKRCSYPRIANGGG (SEQ ID NO: 17)
    AYPHGRSCPQNISKFCFDHEKTNGGG (SEQ ID NO: 18)
    QGGEWHRCMSEEGKHCVDIQFIRGGG (SEQ ID NO: 19)
    TSLTVMTCPHNPSKWCSPLPAAVGGG (SEQ ID NO: 20)
    AMASSATCTKPNSYSCLHAKLVPGGG (SEQ ID NO: 21)
    MPSPPKNCSKFHSALCKGVTWNVGGG (SEQ ID NO: 22)
    SHPQEFWCPQNFSKFCSRSYSNTGGG (SEQ ID NO: 23)
  • All of these unique phage clones were individually amplified and purified, and their ability to specifically bind natalizumab-coated wells was assessed. Of the twenty-three phage clones, eleven demonstrated specific binding to natalizumab (SEQ ID NOs: 1, 2, 4, 13, 16, and 18-23; peptide sequences shown in bold in Table 1). Selected peptides derived from validated phage clones were chemically synthesized with an N-terminal acetyl modification (in some cases), C-terminal biotin modification via a terminal lysine (all peptides), and a disulfide bridge between cysteines 2 and 10 or cysteines 8 and 16 by a contract peptide manufacturer, as shown in Table 2. SEQ ID NOs:29 and 30 were identified by affinity maturation of SEQ ID NO:1 and synthesized with the C-terminal Lys(Biotin).
  • TABLE 2
    Synthetic Mimetope Peptides
    Peptide Peptide
    motif name Peptide sequence
     1 NTZ-01-Bio Ac-ACPMNESKFCGGG{Lys(Biotin)}
    with Cys2-Cys10 bridge
    (SEQ ID NO: 24)
     2 NTZ-02-Bio Ac-ACPSNPSKFCGGG{Lys(Biotin)}
    with Cys2-Cys10 bridge
    (SEQ ID NO: 25)
     4 NTZ-03-Bio Ac-ACPKNPNKFCGGG{Lys(Biotin)}
    with Cys2-Cys10 bridge
    (SEQ ID NO: 26)
    18 NTZ-04-Bio AYPHGRSCPQNISKFCFDHEKTNGGG
    {Lys(Biotin)} with Cys8-Cys16
    bridge (SEQ ID NO: 27)
    23 NTZ-05-Bio SHPQEFWCPQNFSKFCSRSYSNTGGG
    {Lys(Biotin)} with Cys8-Cys16
    bridge (SEQ ID NO: 28)
    NTZ-06-Bio ACPRNESKFCGGG{Lys(Biotin)}
    with Cys2-Cys10 bridge
    (SEQ ID NO: 29)
    NTZ-07-Bio ACPKNPSKFCGGG{Lys(Biotin)}
    with Cys2-Cys10 bridge
    (SEQ ID NO: 30)
  • Synthetic peptides were supplied as TFA salt at >84% purity confirmed by mass spec and HPLC. Peptides were reconstituted and concentration was determined using a NanoDrop spectrophotometer.
  • Example 3: Validation of Mimetope Specificity and Assay Performance
  • Synthesized peptides were validated for specificity by using them as ELISA capture reagents on neutravidin-coated plates. The results are shown in FIG. 1 . Briefly, biotinylated peptides were coated on the wells at 100 ug/mL concentration in either tris buffered saline buffer (TBS) or water. Wells were blocked and then either natalizumab (NTZ) or rituximab (RIT) antibodies spiked in 2.5% BSA/TBST buffer at 1000 ng/mL or 500 ng/mL were added to the wells. Antibody that was captured by the peptides was subsequently detected using a goat anti-human IgG-Fc conjugated to horseradish peroxidase (HRP) combined with a colorimetric substrate. Sensitivity of the mimetope peptide for detection of natalizumab in buffer was determined, and the results presented in FIG. 2 . The minimal coating concentration of peptide (NTZ-01) to achieve desired sensitivity in buffer and human serum (0.1%) was determined, as shown in FIG. 3 . Sensitivity of different mimetope peptides (NTZ-01, NTZ-04, and NTZ-05) for detection of natalizumab (monovalent form) in human serum was determined, and the results are presented in FIGS. 4 and 5 . Assay performance was further assessed for the detection of the monovalent form of natalizumab in human serum (0.4%) using NTZ-01 mimetope peptide-based ELISA, as depicted in FIG. 6 . Spike and recovery experiments were performed where predetermined amounts (nominal concentrations) of natalizumab were spiked into human serum followed by treatment with GSH. The percent of recovery was calculated as follow: calibrated concentration/nominal concentration×100. The results are presented in Table 3:
  • TABLE 3
    Spike and Recovery
    Nominal concentration % recovery
    (ng/mL) Mean SD
    480 88 11
    240 104 3
    80 115 6
    20 112 11
  • The dynamic range of the assay is between 20 and 240 ng/mL of natalizumab. This allows for the accurate quantification of samples containing between 5 and 60 ug/mL natalizumab after applying the minimum sample dilution required for the assay.
  • Example 4: Measurement of Natalizumab Levels in Human Serum
  • A subject undergoing natalizumab therapy has her finger pricked using a lancet. A fixed and predetermined volume of blood (specific for each test) is collected using a transfer pipet and applied to a test device as shown in FIG. 7 . IgG4 antibodies such as natalizumab (NTZ) can exchange Fab-arms with other endogenous IgG4 molecules, leading to the formation of monovalent molecules (hybrid IgG4 molecules with only one arm targeting VLA-4) in addition to the original bivalent form. This test measures both bivalent and total circulating levels of NTZ in the biologic sample. The conjugate pad contains colloidal gold conjugated to mimetope peptide specific for NTZ. The first test line, coated with mimetope peptide, captures bivalent NTZ via one Fab arm while the other Fab arm is detected by the mimetope-gold conjugate (i.e., a double antigen sandwich assay). The second test line is coated with NTZ antibody. When no or little NTZ is present in the sample, the mimetope-gold conjugate binds to the NTZ test line and the line develops color. If sufficient NTZ is present in the sample, both bivalent and monovalent forms compete with the NTZ test line for binding to the mimetope-gold conjugate and less color develops on the test line in a dose-dependent manner (i.e., a competitive assay). The independent control line indicates that the device ran properly. The intensity of the color changes are measured and quantified by an end-point reader.
  • This lateral flow immunoassay (LFA) design is able to measure both bivalent and total circulating levels of natalizumab in biologic samples, as shown in FIG. 8 . When sufficient bivalent natalizumab (NTZ) is present in the sample, one arm of the mAb will bind to the mimetope test line and the other arm will bind to the mimetope-gold conjugate (i.e., a double antigen sandwich), as shown in FIG. 8A. This enables exclusive detection of the bivalent form because only natalizumab with both Fab-arms can bind to mimetope-gold conjugate and simultaneously bind mimetope on the test line. In the competitive test, shown in FIG. 8B, both bivalent and monovalent forms compete with the test line for binding to the mimetope-gold conjugate and less color is observed on the test line. The coating densities of the mimetope on the gold, the mimetope on the first test line, and the antibody on the second test line are optimized to enable measurement of bivalent and total natalizumab in the sample to fall within the dynamic range. Test line intensity is quantified by an off-the-shelf end-point digital reader.
  • Example 5: Natalizumab-Specific VERITOPE™ Inhibition of Natalizumab Binding
  • Natalizumab was incubated with increasing concentrations of natalizumab-specific VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24) or irrelevant peptide (1,000 or 10,000 fold molar excess) and binding to its cellular target was measured by flow cytometry. Jeko-1 cells, expressing CD49d, were used as a model. As shows in FIG. 9 , natalizumab-specific VERITOPE™ can inhibit binding of natalizumab to its cell surface target. The histogram peaks, from left to right, are: Jeko-1 cells only; x-IgG4-A647; 10,000-fold natalizumab-specific VERITOPE™; 1,000-fold natalizumab-specific VERITOPE™; 1,000-fold irrelevant VERITOPE™; and 10,000-fold irrelevant VERITOPE™.
  • Example 6: Comparison of VERITOPE™ and Anti-Idiotype ELISA for Natalizumab Monitoring
  • A commercially available Type 1 anti-idiotype antibody Fab fragment was obtained (AbD21375; BIO-RAD) and compared to a natalizumab-specific VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24). FIGS. 10A and B illustrates a comparison of VERITOPE™ (FIG. 10A) and anti-idiotype (FIG. 10B) ELISA binding. The anti-idiotype antibody (Type 1 or neutralizing) binds to the antigen binding site of natalizumab, specifically recognizes free natalizumab but not free human alpha-4/beta-1 integrin, and is suitable for pharmacokinetic (PK) studies. FIGS. 11A and B demonstrate that both VERITOPE™ (FIG. 11A) and anti-idiotype (FIG. 11B) bind to the same region of natalizumab, most likely the antigen binding site. In FIG. 11A, the natalizumab-specific VERITOPE™ is attached to the surface of the plate as the capture reagent for natalizumab (TYSABRI™), and natalizumab-specific VERITOPE™ (left side) or the anti-idiotype (right side) at different concentrations is the competitive reagent. In FIG. 11B, the anti-idiotype is attached to the surface of the plate as the capture reagent for bivalent (left side) or bispecific (monovalent; right side) natalizumab (TYSABRI™) and the natalizumab-specific VERITOPE™ is the competitive reagent. FIGS. 12A and B demonstrate that heat-inactivation of natalizumab alters natalizumab binding to its cellular target. Natalizumab was heat-inactivated by incubation at 60° C. for various periods of time. Alteration of the binding capacity of natalizumab to its cellular target was measured by flow cytometry. Jeko-1 cells, expressing CD49d, were used as a model. FIG. 12A is the bivalent natalizumab and FIG. 12B is the arm-exchanged (bispecific) natalizumab.
  • Example 7: VERITOPE™ ELISA Specifically Captures Active Drug Compared to Anti-Idiotype ELISA
  • FIGS. 13A and B depict a comparison between anti-idiotype ELISA (FIG. 13A) and VERITOPE™ ELISA (FIG. 13B), demonstrating that VERITOPE™ ELISA is more sensitive than anti-idiotype ELISA to changes that affect the binding pocket of natalizumab. Natalizumab was heat-inactivated by incubation at 60° C. for various periods of time. The ability of anti-idiotype and VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24) capture reagents to distinguish between active and inactive drug was measured by ELISA. Anti-idiotype ELISA measures both inactive and active drug whereas VERITOPE™ ELISA selectively distinguishes & quantifies active drug.
  • Example 8: Natalizumab VERITOPE™ ELISA Assay Specifications
  • The natalizumab-specific VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24) was characterized by a CLIA-certified/CAP-accredited lab and the assay specifications are presented in FIG. 14 . The Lower Limit of Quantification (LLOQ) is 2.0 ug/mL natalizumab in undiluted serum, which corresponds to 8 ng/mL after applying the minimum required dilution of 1/250; the Upper Limit of Quantification (ULOQ) is 16.0 ug/mL natalizumab in undiluted serum, which corresponds to 64 ng/mL after applying the minimum required dilution of 1/250; the data presented are from 5 independent runs, each sample ran in triplicates (mean+/−SD) (Limit of Blank: 0.6 ug/mL; Limit of Detection: 0.8 ug/mL). Intra-assay accuracy and precision was measured. Natalizumab was spiked in human serum at 2, 8 and 16 ug/mL. The samples were analyzed in 5 independent runs in triplicates (16 ug/mL) or quintuplicates (2-8 ug/mL). Analyte recovery was calculated for each concentration as a measure of accuracy. Recovery was found to be between 80-120% of nominal concentrations, within acceptance criteria (Calibrated value/Nominal Value*100). Intra-assay precision was calculated using the same set of samples. A coefficient of variation (% CV) below 15% was obtained for all concentrations tested (SD/mean*100). The results are presented in Table 4:
  • TABLE 4
    Intra-assay Accuracy and Precision
    Nominal % recovery
    Concentration Run
    1 Run 2 Run 3 Run 4 Run 5 Cumulative
    (ug/mL) Mean SD Mean SD Mean SD Mean SD Mean SD Mean SD
    2 101.7 6.2 108.8 5.8 111.4 5.6 113.8 4.2 121.6 6.3 93.4 7.7
    8 90.5 6.3 99.7 5.6 113.1 9.5 99.0 3.6 82.6 2.4 97.0 4.6
    16 96.8 8.1 92.4 5.7 87.6 7.6 90.5 1.6 99.4 15.4 111.4 5.6
    Nominal
    Concentration % CV
    (ug/mL) Run 1 Run 2 Run 3 Run 4 Run 5 Cumulative
    2 5.5 4.8 4.5 3.3 4.7 4.5
    8 6.2 5.1 7.5 3.2 2.2 4.9
    16 6.9 5.1 7.1 1.5 12.6 6.6
  • Inter-assay accuracy and precision was also measured. Natalizumab was spiked in human serum at 2, 8 and 16 ug/mL. The samples were analyzed in 5 independent runs in triplicates (16 ug/mL) or quintuplicates (2-8 ug/mL). Analyte recovery was calculated for each concentration as a measure of accuracy, and was found to be between 80-120% (Calibrated value/Nominal Value*100). The Coefficient of Variation was calculated for each replicate across all 5 runs. A cumulative % CV for each concentration is shown. (% CV:SD/mean*100). The results are presented in Table 5:
  • TABLE 5
    Inter-assay Accuracy and Precision
    Nominal % recovery
    concentration Rep
    1 Rep 2 Rep 3 Rep 4 Rep 5 Cumulative
    (ug/mL) Mean SD Mean SD Mean SD Mean SD Mean SD Mean SD
    2 114.2 5.8 115.6 6.0 108.5 12.5 112.0 7.8 106.9 8.4 111.4 3.7
    8 99.5 12.1 99.1 10.2 96.6 12.4 96.2 16.4 93.4 11.2 97.0 2.3
    16 96.2 13.9 94.0 6.7 95.2 12.4 95.1 1.1
    Nominal
    concentration % CV
    (ug/mL) Rep 1 Rep 2 Rep 3 Rep 4 Rep 5 Cumulative
    2 4.5 4.6 10.3 6.2 7.0 6.5
    8 10.9 9.2 11.4 15.2 10.7 11.5
    16 12.9 10.3 11.7 11.6
  • Spiked serum sample stability of the natalizumab-specific VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24) was measured and the results presented in FIG. 15 . Serum samples spiked with 8 ug/mL natalizumab were quantified using the VERITOPE™ ELISA after exposure to different storage conditions to determine sample stability. Exposure to room temperature (RT), 4° C. or −80° C. for 1, 2 and 8 days did not affect stability.
  • Example 9: Comparison of First Generation and Second Generation Natalizumab VERITOPES™ ELISA Calibration Curve
  • The sensitivity of different mimetope peptides (NTZ-06-Bio, SEQ ID NO:29; NTZ-07-Bio, SEQ ID NO:30; and NTZ-01-Bio, SEQ ID NO:24) for detection of natalizumab in human serum was determined, and the results are presented in FIG. 16 . Experimental conditions used were similar to those described in Example 3, above. The Limit of Blank for NTZ-07-Bio was 85.6+/−41 ng/ml. The selectivity for the natalizumab-specific VERITOPE™ (NTZ-07-Bio; SEQ ID NO:30) for natalizumab over other therapeutic monoclonal antibodies was measured and the results presented in FIG. 17 . Human serum was spiked with different therapeutic monoclonal antibodies (Nataliz=natalizumab; Ritux=rituximab; Tecen=atezolizumab; Key=pembrolizumab; Soliris=eculizumab) and binding to natalizumab-specific peptide NTZ-07-Bio (SEQ ID NO:30) was evaluated in ELISA. As shown in FIG. 17 , VERITOPE™ selectively captures natalizumab in presence of other circulating human IgG and VERITOPE™ does not capture other mAb drugs.
  • Results from previous quantitation of select clinical samples with anti-idiotype and a First Generation VERITOPE™ (NTZ-01-Bio; SEQ ID NO: 24) were compared with results from quantitation of the same select clinical samples using anew assay with Second Generation VERITOPE™ NTZ-06-Bio (SEQ ID NO: 29) and NTZ-07-Bio (SEQ ID NO: 30) and the data presented in Table 6:
  • TABLE 6
    Comparison of 1st Generation and 2nd Generation Natalizumab
    VERITOPES ™ Quantitation
    Previous Assay New Assay
    Sample # Anti-Id NTZ01 NTZ01 NTZ06 NTZ07
    12 19.1 13.8 15.6 18.8 20.6
    20 33.6 29.5 61.5 71.9 76.4
    24 14.7 1.7 ND 1.4 2
    30 10.3 2.6 2.6 3.4 4.4
    32 46.8 35.9 47.9 62.8 68.8
  • Results from previous quantitation of select clinical samples with anti-idiotype and a First Generation VERITOPE™ (NTZ-01-Bio; SEQ ID NO:24) were compared with results from anew assay with a Second Generation VERITOPE™ NTZ-07-Bio (SEQ ID NO:30) and the data presented in Table 7:
  • TABLE 7
    Comparison of Anti-Idiotype, 1st Generation, and 2nd Generation
    Natalizumab VERITOPES ™ Quantitation
    Previous Assay New Assay
    Sample # Anti-Id NTZ01 NTZ07
    1 28.2 11.3 18.9
    2 N/A 24.9 33.4
    3 4.0 0.0 1.0
    4 8.2 2.1 3.0
    5 4.0 1.1 1.1
    9 23.7 6.2 7.7
    10 25.0 16.6 21.6
    11 18.1 3.1 5.3
    12 14.2 0.0 3.7
    13 2.3 0.0 0.8
    16 29.3 5.7 10.7
    17 11.5 5.6 13.1
    19 20.5 0.9 2.8
    22 21.4 9.6 26.1
    23 3.5 0.0 0.4
    24 6.7 0.6 0.8
    25 9.1 2.7 4.9
    26 29.3 4.1 7.0
    27 5.4 0.2 0.4
    34 17.4 4.6 6.3
    37 16.2 14.0 23.8
    39 19.5 3.5 4.9
    47 20.3 10.0 11.4
    59 5.9 0.2 0.7
  • As shown in Table 7, the Second Generation VERITOPE™ NTZ-07-Bio (SEQ ID NO:30) improves sensitivity, and now measurements that were ‘0’ by NTZ-01-Bio (SEQ ID NO:24) (but not by anti-idiotype) give a value.
  • While preferred embodiments of the present methods, assays, complexes, and assays have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the methods, assays, complexes, and assays described herein. It should be understood that various alternatives to the embodiments of the methods, assays, complexes, and assays described herein may be employed in practice. It is intended that the following claims define the scope of the methods, assays, complexes, and assays and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (20)

What is claimed is:
1. A test device comprising:
(a) a sample pad for receiving a sample from a subject, wherein the sample comprises an antibody or a fragment of the antibody;
(b) a conjugate pad; and
(c) a test membrane comprising a first test line comprising a peptide consisting of any one of SEQ ID NOs: 1, 2, 4, 13, 16, and 18-23.
2. The test device of claim 1, wherein the conjugate pad comprises a detection reagent conjugated to the peptide in step (c).
3. The test device of claim 1, wherein the conjugate pad comprises a detection reagent conjugated to an antibody specific for natalizumab.
4. The test device of claim 1, wherein the conjugate pad comprises a detection reagent conjugated to:
the peptide in step (c); and
an antibody specific for natalizumab.
5. The test device of claim 3, wherein the antibody specific for natalizumab binds natalizumab at a variable region.
6. The test device of claim 3, wherein the antibody specific for natalizumab binds natalizumab at a constant region.
7. The test device of claim 1, wherein the test membrane further comprises a second test line comprising an antibody specific for the peptide in step (c).
8. The test device of claim 7, wherein the antibody is natalizumab.
9. The test device of claim 1, wherein the antibody is free, circulating natalizumab and not complexed to a protein prior to step (b).
10. The test device of claim 1, wherein the first test line measures a bivalent form of the antibody.
11. The test device of claim 7, wherein the second test line measures both bivalent and monovalent forms of the antibody.
12. The test device of claim 1, wherein the test membrane comprises Western blot analysis, dot blot analysis, flow cytometry, enzyme-linked immunosorbent assay (ELISA), lateral flow immunoassay, radioimmunoassay (RIA), competitive immunoassay, dual antibody sandwich assay, chemiluminescent assay, bioluminescent assay, fluorescent assay, or agglutination assay.
13. The test device of claim 1, wherein the first test line comprises a dual antibody sandwich assay.
14. The test device of claim 7, wherein the second test line comprises a competitive immunoassay.
15. The test device of claim 1, wherein the test membrane further comprises a control line.
16. The test device of claim 2, wherein the detection reagent comprises colloidal gold.
17. The test device of claim 1, wherein the sample is a biological fluid.
18. The test device of claim 17, wherein the biological fluid comprises serum, plasma, whole blood, red blood cell concentrates, platelet concentrates, leukocytes concentrates, urine, cerebral spinal fluid, or sputum.
19. The test device of claim 17, wherein the biological fluid contains antibody at a concentration of between about 0.5 mcg/mL to 120 mcg/mL.
20. The test device of claim 1, wherein the test device is a lateral flow immunoassay (LFA) device.
US18/312,915 2016-08-12 2023-05-05 Detection and quantification of natalizumab Pending US20230417761A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/312,915 US20230417761A1 (en) 2016-08-12 2023-05-05 Detection and quantification of natalizumab

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662374217P 2016-08-12 2016-08-12
PCT/US2017/046499 WO2018031887A1 (en) 2016-08-12 2017-08-11 Detection and quantification of natalizumab
US201916324878A 2019-02-11 2019-02-11
US18/312,915 US20230417761A1 (en) 2016-08-12 2023-05-05 Detection and quantification of natalizumab

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2017/046499 Division WO2018031887A1 (en) 2016-08-12 2017-08-11 Detection and quantification of natalizumab
US16/324,878 Division US11680948B2 (en) 2016-08-12 2017-08-11 Detection and quantification of natalizumab

Publications (1)

Publication Number Publication Date
US20230417761A1 true US20230417761A1 (en) 2023-12-28

Family

ID=61163422

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/324,878 Active 2040-07-04 US11680948B2 (en) 2016-08-12 2017-08-11 Detection and quantification of natalizumab
US18/312,915 Pending US20230417761A1 (en) 2016-08-12 2023-05-05 Detection and quantification of natalizumab

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/324,878 Active 2040-07-04 US11680948B2 (en) 2016-08-12 2017-08-11 Detection and quantification of natalizumab

Country Status (3)

Country Link
US (2) US11680948B2 (en)
EP (1) EP3497444A4 (en)
WO (1) WO2018031887A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3497444A4 (en) 2016-08-12 2020-11-11 Abreos Biosciences, Inc. Detection and quantification of natalizumab

Family Cites Families (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5089605A (en) 1987-03-13 1992-02-18 Repligen Corporation Immobilized immunoglobulin-binding proteins
DE3911539A1 (en) 1989-04-08 1990-10-11 Boehringer Mannheim Gmbh TEST CARRIER ANALYSIS SYSTEM
US6596476B1 (en) * 1989-12-22 2003-07-22 Abbott Laboratories Hepatitis C assay
DE4041905A1 (en) 1990-12-27 1992-07-02 Boehringer Mannheim Gmbh TEST CARRIER ANALYSIS SYSTEM
US5567594A (en) 1991-04-26 1996-10-22 Enteron, L.P. Methods and compositions for the detection and treatment of diseases associated with antigens of microorganisms
US5877028A (en) 1991-05-29 1999-03-02 Smithkline Diagnostics, Inc. Immunochromatographic assay device
DE4402756A1 (en) 1994-01-31 1995-08-03 Boehringer Mannheim Gmbh Specific binding substances for antibodies and their use for immunoassays or vaccines
US5885577A (en) 1994-09-21 1999-03-23 Cytogen Corporation Antigen binding peptides (abtides) from peptide libraries
GB9502112D0 (en) 1995-02-03 1995-03-22 British Biocell Int Assay device and method
US7074888B1 (en) 1995-03-17 2006-07-11 The Research Foundation Of State University Of New York Mimotopes and anti-mimotopes of human platelet glycoprotein Ib/IX
JP2002511264A (en) 1998-04-16 2002-04-16 ジェネンテック・インコーポレーテッド Secretion of glycosylated proteins using tissue plasminogen activator prosequence
US7112661B1 (en) 1998-10-30 2006-09-26 The Research Foundation Of State University Of New York Variable heavy chain and variable light chain regions of antibodies to human platelet glycoprotein Ib alpha
AU2001282035B2 (en) 2000-08-03 2006-11-09 Inverness Medical Switzerland Gmbh Peptides capable of functioning as mimotopes for estradiol analytes
US8216797B2 (en) 2001-02-07 2012-07-10 Massachusetts Institute Of Technology Pathogen detection biosensor
JP4424987B2 (en) 2001-09-20 2010-03-03 ボード オブ リージェンツ, ザ ユニバーシティ オブ テキサス システム Measurement of circulating therapeutic antibodies, antigens and antigen / antibody complexes using an ELISA assay
US20030091986A1 (en) 2001-11-09 2003-05-15 Regents Of The University Of California, Office Of Technology Transfer Identification of expressed genes using phage display
US20060286379A1 (en) 2002-08-13 2006-12-21 Yong Gao Magnetic nanoparticle supports
US7459314B2 (en) 2003-02-13 2008-12-02 Inverness Medical Switzerland Gmbh Lateral flow immunoassay controls
US7365154B2 (en) 2003-06-23 2008-04-29 Centocor, Inc. Peptides antagonistic to an anti-angiogenic antibody and uses therefor
EP1677825A4 (en) 2003-09-19 2008-11-05 Scripps Research Inst Peptide that binds to a broadly neutralizing anti-hiv antibody-structure of 4e10 fab fragment complex, uses thereof, compositions therefrom
EP1718972A4 (en) 2004-02-17 2009-05-27 Binax Inc Methods and kits for detection of multiple pathogens
US7781226B2 (en) 2004-02-27 2010-08-24 The Board Of Regents Of The University Of Texas System Particle on membrane assay system
US20060008920A1 (en) 2004-07-09 2006-01-12 Branan Medical Corporation Combination assay for alcohol and drugs of abuse
AU2005263334C1 (en) * 2004-07-20 2011-01-20 Symphogen A/S A procedure for structural characterization of a recombinant polyclonal protein or a polyclonal cell line
US20060068501A1 (en) 2004-09-30 2006-03-30 Nj International Llc Apparatus and methods for steroid hormone testing
AU2006272714A1 (en) 2005-07-25 2007-02-01 Trubion Pharmaceuticals, Inc. Compositions and methods for protein deaggregation
WO2007022557A1 (en) 2005-08-23 2007-03-01 Diatech Pty Ltd Mimotopes of epstein-barr virus (ebv) epitopes
KR101282251B1 (en) 2006-05-23 2013-07-10 삼성전자주식회사 Display apparatus, image processing apparatus and control method thereof
CA2690904C (en) 2007-06-27 2015-11-03 Inbios International, Inc. Lateral flow assay system and methods for its use
USD579459S1 (en) 2007-08-03 2008-10-28 Nec Display Solutions Of America, Inc. Projector screen with an image
US20110117601A1 (en) 2007-08-31 2011-05-19 Markus Haberger Glycosylation Profile Analysis
US8394602B2 (en) 2007-11-12 2013-03-12 Hiroshima University Luminescent method for measuring endotoxin
WO2009121024A2 (en) 2008-03-27 2009-10-01 The Regents Of The University Of California Methods for detecting antibodies
WO2010073647A1 (en) 2008-12-25 2010-07-01 国立大学法人大阪大学 Human anti-human influenza virus antibody
GB0905519D0 (en) 2009-03-31 2009-05-13 Biofortuna Ltd Assay method and device
EP2452197A4 (en) 2009-07-08 2013-01-23 Anp Technologies Inc Immunogenicity assay
CA2776756A1 (en) 2009-10-11 2011-04-14 Biogen Idec Ma Inc. Anti-vla-4 related assays
KR20120096932A (en) 2009-11-17 2012-08-31 아박시스, 인크. Peptides and methods for the detection of lyme disease antibodies
KR100946566B1 (en) 2009-11-18 2010-03-11 주식회사 인포피아 A lateral flow immunoassay device with a more rapid and accurate test result
US9336353B2 (en) 2010-06-25 2016-05-10 Dexcom, Inc. Systems and methods for communicating sensor data between communication devices of a glucose monitoring system
WO2012023053A2 (en) 2010-08-16 2012-02-23 Novimmune S.A. Methods for the generation of multispecific and multivalent antibodies
US20130085349A1 (en) 2011-06-21 2013-04-04 Yofimeter, Llc Analyte testing devices
US20130040401A1 (en) 2011-08-12 2013-02-14 Benedict L. Zin Test strip reader
US9020476B2 (en) 2011-09-12 2015-04-28 Leipzig Technology, Llc System and method for remote care and monitoring using a mobile device
EP3489685B1 (en) 2011-11-21 2023-11-01 Zoetis Services LLC Signal amplification in lateral flow and related immunoassays
US20130143246A1 (en) 2011-12-02 2013-06-06 Lifescan Scotland Ltd. Hand-held test meter with analytical test strip ejection mechanism
USD731528S1 (en) 2012-05-15 2015-06-09 Fujitsu Limited Display with a graphical user interface
USD750115S1 (en) 2012-12-05 2016-02-23 Ivoclar Vivadent Ag Display screen or a portion thereof having an animated graphical user interface
USD732062S1 (en) 2013-02-22 2015-06-16 Samsung Electronics Co., Ltd. Display screen or a portion thereof with graphical user interface
USD701875S1 (en) 2013-02-25 2014-04-01 Pepsico, Inc. Display screen with graphical user interface
US20150293086A1 (en) 2014-04-14 2015-10-15 Abreos Biosciences, Inc. Lateral flow immunoassay
GB2548944B (en) 2014-04-17 2018-11-28 Z Integrated Digital Tech Inc Electronic test device data communication
USD780188S1 (en) 2014-05-12 2017-02-28 Tencent Technology (Shenzhen) Company Limited Display screen or portion thereof with graphical user interface
USD757040S1 (en) 2014-05-29 2016-05-24 Comcast Cable Communications, Llc Display screen with animated graphical user interface
EP2982987A1 (en) 2014-08-08 2016-02-10 Proteomika, S.L. Lateral flow immunoassays for the detection of antibodies against biological drugs
WO2016123105A1 (en) 2015-01-26 2016-08-04 Abreos Biosciences, Inc. Cleavage coupled competitive lateral flow assay
US9927443B2 (en) 2015-04-10 2018-03-27 Conquerab Inc. Risk assessment for therapeutic drugs
USD771072S1 (en) 2015-04-27 2016-11-08 Lutron Electronics Co., Inc. Display screen or portion thereof with graphical user interface
CN108431018A (en) 2015-06-12 2018-08-21 王天欣 The method of protein modification in medicinal application
US11046745B2 (en) 2015-07-14 2021-06-29 BioNTech SE Peptide mimotopes of the CD3 T-cell co-receptor epsilon chain and uses thereof
USD766936S1 (en) 2015-07-29 2016-09-20 Solar Turbines Incorporated Display screen or portion thereof with icon
JP1557056S (en) 2015-09-15 2016-08-29
USD774205S1 (en) 2015-12-24 2016-12-13 Beckman Coulter, Inc. Diagnostic device
USD807380S1 (en) 2016-04-13 2018-01-09 Vudu, Inc. Display screen or portion thereof with graphical user interface
JP1572567S (en) 2016-04-19 2020-03-09
JP1572568S (en) 2016-04-19 2020-03-09
USD805526S1 (en) 2016-05-05 2017-12-19 Chris Steven Ternoey Display screen portion with animated graphical user interface
USD805527S1 (en) 2016-05-05 2017-12-19 Chris Steven Ternoey Display screen portion with animated graphical user interface
JP1594017S (en) 2016-05-13 2018-01-09
USD806091S1 (en) 2016-05-16 2017-12-26 Google Llc Display screen with animated graphical user interface
USD800912S1 (en) 2016-06-17 2017-10-24 Abreos Biosciences, Inc. Point-of-care diagnostic device
EP3497444A4 (en) 2016-08-12 2020-11-11 Abreos Biosciences, Inc. Detection and quantification of natalizumab
USD798316S1 (en) 2016-10-26 2017-09-26 Microsoft Corporation Display screen with graphical user interface
EP3606937A4 (en) 2017-04-04 2020-12-30 Abreos Biosciences, Inc. Immunoaffinity purification of antibodies using mimetopes

Also Published As

Publication number Publication date
EP3497444A4 (en) 2020-11-11
US11680948B2 (en) 2023-06-20
EP3497444A1 (en) 2019-06-19
US20210285959A1 (en) 2021-09-16
WO2018031887A1 (en) 2018-02-15

Similar Documents

Publication Publication Date Title
Eckman et al. Basophil phenotypes in chronic idiopathic urticaria in relation to disease activity and autoantibodies
US20220146535A1 (en) Compounds and methods targeting human tau
Smith et al. Detection of antibodies against therapeutic proteins in the presence of residual therapeutic protein using a solid-phase extraction with acid dissociation (SPEAD) sample treatment prior to ELISA
JP4838140B2 (en) Immunosorbent blood test to assess paroxysmal brain firing
US20090011444A1 (en) Detection and diagnosis of inflammatory disorders
US20230417761A1 (en) Detection and quantification of natalizumab
JP6938372B2 (en) Antibodies that bind to the linear epitope of human p53 and their diagnostic applications
Kuehnl et al. Comparative measurement of CNP and NT-proCNP in human blood samples: a methodological evaluation
JP2023517751A (en) Methods for mitigating target interference in anti-drug antibody assays
US20200239598A1 (en) Products and methods for monitoring adherence to nucleoside reverse transcriptase inhibitor therapy
JP6564435B2 (en) Administration of alpha4beta7 heterodimer specific antibody
JP5518836B2 (en) Method for diagnosing pulmonary arterial hypertension
JP6170644B1 (en) Anti-drug antibody measurement method
WO1997026537A1 (en) Methods for the detection of alzheimer's disease
US11591387B2 (en) Compounds and methods targeting interleukin-19
EP4262881A2 (en) Reagents, methods, and systems for detecting therapeutic agents to monitor adherence to nucleoside reverse transcriptase inhibitor metabolites
JP2014517911A (en) Adiponectin receptor C-terminal fragment (CTF) immunoglobulin
US20200116741A1 (en) Immunoassay for Human Erythroferrone
WO2003104812A2 (en) ASSAY FOR THE DIAGNOSIS OF ALZHEIMER'S DISEASE BASED ON THE DETERMINATION OF THE RATIO OF hAβ42:hAβ40
JP7106810B2 (en) Novel lung cancer marker
Borromeo et al. Heterophile antibody interference in a solid phase sandwich immunoassay for detection of equine growth hormone in plasma
US10627415B2 (en) Compositions and methods for diagnosing, monitoring, and treating an autoimmune disease
WO2018132639A1 (en) Methods and kits for the diagnosis and/or prognosis of ocular cicatricial pemphigoid
CN111919121A (en) Method for determining severity of neonatal hypoxic-ischemic encephalopathy and prognosis prediction method
CN114364984A (en) A method of diagnosing or monitoring renal function or diagnosing renal dysfunction in a pediatric patient

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABREOS BIOSCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MESSMER, BRADLEY T.;UZRI, DINA;FECTEAU, JESSIE-FARAH;SIGNING DATES FROM 20230216 TO 20230316;REEL/FRAME:064726/0341

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION