US20170360926A1 - PREDICTING OUTCOME OF TREATMENT WITH AN ANTI-alpha4beta7 INTEGRIN ANTIBODY - Google Patents

PREDICTING OUTCOME OF TREATMENT WITH AN ANTI-alpha4beta7 INTEGRIN ANTIBODY Download PDF

Info

Publication number
US20170360926A1
US20170360926A1 US15/538,869 US201515538869A US2017360926A1 US 20170360926 A1 US20170360926 A1 US 20170360926A1 US 201515538869 A US201515538869 A US 201515538869A US 2017360926 A1 US2017360926 A1 US 2017360926A1
Authority
US
United States
Prior art keywords
vedolizumab
patient
antibody
concentration
dose
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.)
Abandoned
Application number
US15/538,869
Inventor
Maria Rosario
Timothy L. Wyant
Brihad Abhyankar
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.)
Takeda Development Centre Europe Ltd
Millennium Pharmaceuticals Inc
Original Assignee
Takeda Development Centre Europe Ltd
Millennium Pharmaceuticals 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 Takeda Development Centre Europe Ltd, Millennium Pharmaceuticals Inc filed Critical Takeda Development Centre Europe Ltd
Priority to US15/538,869 priority Critical patent/US20170360926A1/en
Assigned to MILLENNIUM PHARMACEUTICALS, INC. reassignment MILLENNIUM PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROSARIO, MARIA, WYANT, TIMOTHY L.
Assigned to TAKEDA DEVELOPMENT CENTRE EUROPE LTD. reassignment TAKEDA DEVELOPMENT CENTRE EUROPE LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABHYANKAR, Brihad
Assigned to TAKEDA PHARMACEUTICAL COMPANY, LTD. reassignment TAKEDA PHARMACEUTICAL COMPANY, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TAKEDA DEVELOPMENT CENTRE EUROPE LTD.
Assigned to MILLENNIUM PHARMACEUTICALS, INC. reassignment MILLENNIUM PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TAKEDA PHARMACEUTICAL COMPANY, LTD.
Publication of US20170360926A1 publication Critical patent/US20170360926A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • 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
    • G01N2333/52Assays involving cytokines
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • IBD Inflammatory bowel disease
  • IBD treatments have included anti-inflammatory drugs (such as, corticosteroids and sulfasalazine), immunosuppressive drugs (such as, 6-mercaptopurine, cyclosporine and azathioprine) and surgery (such as, colectomy).
  • anti-inflammatory drugs such as, corticosteroids and sulfasalazine
  • immunosuppressive drugs such as, 6-mercaptopurine, cyclosporine and azathioprine
  • surgery such as, colectomy.
  • treatment progresses into regimens that expose patients to progressive risk of side effects and loss of quality of life.
  • Integrin receptors are important for regulating both lymphocyte recirculation and recruitment to sites of inflammation (Carlos, T. M. and Harlan, J. M., Blood, 84:2068-2101 (1994)).
  • the human ⁇ 4 ⁇ 7 integrin has several ligands, one of which is the mucosal vascular addressin MAdCAM-1 (Berlin, C., et al., Cell 74: 185-195 (1993); Erle, D. J., et al., J. Immunol. 153:517-528 (1994)), which is expressed on high endothelial venules in mesenteric lymph nodes and Peyer's patches (Streeter, P.
  • the ⁇ 4 ⁇ 7 integrin acts as a homing receptor that mediates lymphocyte migration to intestinal mucosal lymphoid tissue (Schweighoffer, T., et al., J. Immunol. 151: 717-729 (1993)).
  • Antibodies against human ⁇ 4 ⁇ 7 integrin such as murine monoclonal antibody Act-1 (mAb Act-1), interfere with ⁇ 4 ⁇ 7 integrin binding to mucosal addressin cell adhesion molecule-1 (MAdCAM-1) present on high endothelial venules in mucosal lymph nodes.
  • Act-1 was originally isolated by Lazarovits, A. I., et al., J. Immunol. 133:1857-1862 (1984), from mice immunized with human tetanus toxoid-specific T lymphocytes and was reported to be a mouse IgG1/ ⁇ antibody. Subsequent analysis of the antibody by Schweighoffer, T., et al., J. Immunol.
  • EntyvioTM vedolizumab an anti- ⁇ 4 ⁇ 7 integrin monoclonal antibody (mAb) with structural features derived from Act-1, is indicated for treating ulcerative colitis (UC) and Crohn's disease (CD).
  • UC ulcerative colitis
  • CD Crohn's disease
  • Studies reporting the activity of vedolizumab in treating these disorders showed varying levels of success depending on the disorder and nature of prior therapies. As these were lengthy studies and there are a growing number of treatment options available to patients, there is a need to identify patients who can benefit from vedolizumab therapy early in their treatment. Expedient and accurate treatment decisions lead to effective management of the disease.
  • the invention relates to the identification of patients who can respond to therapy comprising an anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab.
  • an anti- ⁇ 4 ⁇ 7 antibody such as vedolizumab.
  • factors measured from the patient or from biological samples of the patient indicate whether a patient is likely to respond to treatment.
  • pharmacokinetics or pharmacodynamics factors can indicate whether a patient will respond to treatment with an anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab.
  • an anti- ⁇ 4 ⁇ 7 antibody such as vedolizumab.
  • understanding determinants of mAb clearance may optimize dosing regimens.
  • Applicants have identified the problem, and characterized anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab, pharmacokinetic factors in patients with UC and CD, identified clinically relevant determinants of anti- ⁇ 4 ⁇ 7 antibody, such as antibody clearance, and described the pharmacokinetic-pharmacodynamic relationship using population modeling.
  • a pharmacokinetics factor is mean serum trough concentration. In other embodiments, a pharmacokinetics factor is therapeutic antibody clearance. In an embodiment, a pharmacodynamic factor is the amount of antibody bound to ⁇ 4 ⁇ 7 integrin. In another embodiment, a pharmacodynamic factor is the amount of unbound ⁇ 4 ⁇ 7 integrin.
  • a method for treating a patient having inflammatory bowel disease (IBD) with an anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab comprises administering two doses of the anti- ⁇ 4 ⁇ 7 antibody to a patient suffering from IBD, wherein the second dose is administered about two weeks after the first dose is administered to the patient; waiting a period of time about four weeks; measuring the patient's serum concentration of the anti- ⁇ 4 ⁇ 7 antibody; and administering one or more further doses of the anti- ⁇ 4 ⁇ 7 antibody to the patient if the patient's serum concentration, e.g., a trough serum concentration, of the antibody is at least 8 ⁇ g per ml.
  • the waiting period is two weeks or about two to five weeks.
  • the method comprises administering two doses of vedolizumab to a patient suffering from IBD, wherein the second dose is administered about two weeks after the first dose is administered to the patient; waiting a period of time about four weeks; measuring the patient's serum concentration of vedolizumab; and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration, e.g., a trough serum concentration, of the antibody is at least 8 ⁇ g per ml.
  • the patient's serum concentration of the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 ⁇ g per ml.
  • a patient who is likely to respond to the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab has a serum concentration, e.g., a trough serum concentration of greater than 17 ⁇ g/ml, greater than 25 ⁇ g/ml, or greater than 35 mg/mi.
  • the patient's serum concentration of anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab may be selected from the group consisting of 12-25 ⁇ s per ml, 15-17 ⁇ g per ml, 17-25 ⁇ g per ml, 12-40 ⁇ g per ml, and 17-40 ⁇ g ml.
  • a method for treating a patient having inflammatory bowel disease (IBD) with an anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab comprises the steps of administering at least one dose of vedolizumab to the patient suffering from IBD; waiting a period of at least two weeks, measuring the patient's serum concentration of vedolizumab; and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration is at least 8 ⁇ g per ml.
  • the waiting period is about two to five weeks.
  • clinical measures during early treatment can indicate whether a patient will respond to treatment with an anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab.
  • the patient can have mucosal healing.
  • the patient can have deep remission.
  • the method for treating a patient can comprise measuring an endoscopic subscore, e.g., from the Mayo score, wherein the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab is continued with an endoscopic score of less than 3.
  • the endoscopic score can be less than 2.5, less than 2, between 0 and 2, 1, or equal to or less than 1 ( ⁇ 1) or less than 1 (( ⁇ 1).
  • the method for treating a patient can comprise measuring the Crohn's Disease Activity Index (CDAI). In another embodiment, the method for treating a patient can comprise measuring the number, size or severity of fistulae. In an embodiment, the endoscopic score is a simple endoscopic score for Crohn's Disease (SES-CD). In some embodiments, the SES-CD can be in the range of 0 to 6, ⁇ 6, ⁇ 5 or no more than 4 ( ⁇ 4). In some embodiments, the SES-CD is decreased by 25%, 35%, 40%, 45%, 50%, 55%, 60% or 65% from baseline. In one embodiment, the SES-CD is decreased 50% from baseline. In some embodiments, the method for treating a patient can comprise measuring the amount of ulceration in the digestive tract. In some embodiments, the method for treating a patient can comprise measuring mucosal healing. In some embodiments of mucosal healing, ulceration is decreased or absent.
  • CDAI Crohn's Disease Activity Index
  • the method for treating a patient can comprise measuring the number, size or severity
  • measures of inflammation can indicate whether a patient will respond to treatment with an anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab.
  • Measures of inflammation include amounts of fecal calprotectin, amounts of C-reactive protein (CRP) and amount of albumin.
  • the method comprises measuring the fecal calprotectin concentration.
  • Treatment with the antibody, e.g., vedolizumab can be continued with a fecal calprotectin concentration of less than 1500 ⁇ g/g.
  • the fecal calprotectin concentration can be less than 1250 ⁇ g/g, less than 1000 ⁇ g/g, less than 750 ⁇ g/g, less than 500 ⁇ g/g, less than 400 ⁇ g/g, less than 300 ⁇ g/g, less than 250 ⁇ g/g, between 200-1200 ⁇ g/g, between 350 to 800 ⁇ g/g, between 300-1000 ⁇ g/g, ⁇ 50 ⁇ g/g, ⁇ 100 ⁇ g/g, ⁇ 150 ⁇ g/g, ⁇ 200 ⁇ g/g, ⁇ 250-499 ⁇ g/g, or between 500 to 900 ⁇ g/g.
  • treatment with an anti- ⁇ 4 ⁇ 7 antibody e.g., vedolizumab can be continued if the fecal calprotectin is reduced to less than 50% of the baseline or concentration before treatment.
  • the fecal calprotectin can be reduced to less than 45%, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, between 10-55%, between 10-30%, between 15-35%, between 15-45%, or between 20-40% of the baseline or concentration before treatment.
  • methods described herein comprise measuring albumin concentration.
  • An albumin concentration greater than 3.2 g/dL further identifies the patient for continued treatment with an anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab.
  • the albumin concentration can be greater than 3.5 g/dL, greater than 4.0 g/dL, or greater than 4.7 g/dL, in the range of 3.3 to 5.0 g/dL, in the range of 3.5 to 5.0 g/dL, in the range of 3.8 to 5.0 g/dL or in the range of 4.0 to 5.0 g/dL.
  • the invention relates to therapeutic methods which further include the step of continuing, stopping, discontinuing or halting a therapy accordingly where factors measured as described herein indicate whether there is continued benefit of the therapy.
  • a method of identifying a patient for continued treatment with an anti- ⁇ 4 ⁇ 7 antibody comprises the steps of: measuring the concentration of the anti- ⁇ 4 ⁇ 7 antibody in a sample of serum obtained from a patient suffering from inflammatory bowel disease (IBD) and who received at least one dose of an anti- ⁇ 4 ⁇ 7 antibody within the previous two months; and identifying the patient for continued treatment with the anti- ⁇ 4 ⁇ 7 antibody if the serum concentration in the sample is at least 8 ⁇ g per ml.
  • the patient may have received at least one dose of the anti- ⁇ 4 ⁇ 7 antibody within the previous month.
  • the patient received at least two doses of the anti- ⁇ 4 ⁇ 7 antibody within the previous four months; and the method identifies the patient for continued treatment with the anti- ⁇ 4 ⁇ 7 antibody if its serum concentration in the sample is at least 8 ⁇ g per ml four weeks after the second dose, e.g., four weeks after the dose at week 2.
  • the patient may have received at least two doses of the anti- ⁇ 4 ⁇ 7 antibody within the previous three months or the previous two months.
  • the patient's serum concentration of the anti- ⁇ 4 ⁇ 7 antibody may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 ⁇ g per ml.
  • the patient's serum concentration of the anti- ⁇ 4 ⁇ 7 antibody may be selected from the group consisting of 12-25 ⁇ g per ml, 15-17 ⁇ g per ml, 17-25 ⁇ g per ml, 12-40 ⁇ g per ml, and 17-40 ⁇ g ml.
  • a method of identifying a patient for continued treatment with vedolizumab comprises the steps of: measuring the concentration of vedolizumab in a sample of serum obtained from a patient suffering from inflammatory bowel disease (IBD) and who received at least two doses of vedolizumab within the previous four months; and identifying the patient for continued treatment with vedolizumab if the serum concentration in the sample is at least 8 ⁇ g per ml four weeks after the second dose, e.g., four weeks after the dose at week 2.
  • the patient may have received at least two doses of vedolizumab within the previous three months or the previous two months.
  • the patient's serum concentration of vedolizumab may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 ⁇ g per ml. In another embodiment, the patient's serum concentration of vedolizumab may be selected from the group consisting of 12-25 ⁇ g per ml, 15-17 ⁇ g per ml, 17-25 ⁇ g per ml, 12-40 ⁇ g per ml, and 17-40 ⁇ g ml.
  • the invention in another embodiment, relates to a method of identifying a patient for continued treatment with vedolizumab, the method comprising the steps of: measuring the concentration of vedolizumab in a sample of serum obtained from a patient suffering from inflammatory bowel disease (IBD) and who received at least one dose of vedolizumab within the previous two months; and identifying the patient for continued treatment with vedolizumab if the serum concentration in the sample is at least 8 ⁇ g per ml.
  • the patient may have received at least one dose of vedolizumab within the previous month.
  • the patient's serum concentration of vedolizumab may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 ⁇ g per ml.
  • the patient's serum concentration of vedolizumab may be selected from the group consisting of 12-25 ⁇ g per ml, 15-17 ⁇ g per ml, 17-25 ⁇ g per ml, 12-40 ⁇ g per ml, and 17-40 ⁇ g ml.
  • continuing treatment includes maintaining remission of IBD.
  • the invention relates to a method for maintaining remission of inflammatory bowel disease (IBD) in a patient, wherein the patient had received at least one dose of an anti- ⁇ 4 ⁇ 7 antibody in the previous two months, the method comprising: obtaining a serum sample from the patient; measuring the concentration of the anti- ⁇ 4 ⁇ 7 antibody in the sample; administering the anti- ⁇ 4 ⁇ 7 antibody thereafter every eight weeks if the concentration is at least 8 ⁇ g per ml.
  • the patient's serum concentration of the anti- ⁇ 4 ⁇ 7 antibody may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 ⁇ g per ml.
  • the patient's serum concentration of the anti- ⁇ 4 ⁇ 7 antibody may be selected from the group consisting of 12-25 ⁇ g per ml, 15-17 ⁇ g per ml, 17-25 ⁇ g per ml, 12-40 ⁇ s per ml, and 17-40 ⁇ s ml.
  • the invention relates to a method for maintaining remission of inflammatory bowel disease (IBD) in a patient, wherein the patient had received at least one dose of vedolizumab in the previous two months, the method comprising:
  • the patient's serum concentration of vedolizumab may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 ⁇ g per ml.
  • the patient's serum concentration of vedolizumab may be selected from the group consisting of 12-25 ⁇ g per ml, 15-17 ⁇ g per ml, 17-25 ⁇ g per ml, 12-40 ⁇ g per ml, and 17-40 ⁇ g ml.
  • evaluation of a patient early in treatment with an anti- ⁇ 4 ⁇ 7 antibody indicates that the patient is in deep remission.
  • the invention relates to a method for treating a patient having inflammatory bowel disease (IBD) with vedolizumab, comprising: providing two doses of vedolizumab, wherein the second dose is about two weeks after the first; waiting about four weeks; and continuing to treat the patient with vedolizumab if the patient is in deep remission, wherein determining deep remission comprises measuring an endoscopic subscore for the patient.
  • IBD inflammatory bowel disease
  • the patient can be determined to be in deep remission and continue to receive the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab, if the endoscopic subscore is 0 to 1.
  • the method can further comprise determining a patient-reported outcome score, wherein a determination of deep remission comprises 0 to 1 for both the endoscopic subscore and the patient reported outcome score.
  • the patient reported outcome score can comprise a rectal bleeding subscore.
  • the patient reported outcome score can comprise a stool frequency subscore.
  • the method can further comprise measuring the fecal calprotectin concentration.
  • Treatment with an anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab can be continued with a fecal calprotectin concentration of less than 1500 ⁇ g/g.
  • the fecal calprotectin concentration can be less than 1250 ⁇ g/g, less than 1000 ⁇ g/g, less than 750 ⁇ g/g, less than 500 ⁇ g/g, less than 400 ⁇ g/g, less than 300 ⁇ g/g, less than 250 ⁇ g/g, between 200-1200 ⁇ g/g, between 350 to 800 ⁇ g/g, between 300-1000 ⁇ g/g, ⁇ 50 ⁇ g/g, ⁇ 100 ⁇ g/g, ⁇ 150 ⁇ g/g, ⁇ 200 ⁇ g/g, ⁇ 250-499 ⁇ g/g, or between 500 to 900 ⁇ g/g.
  • treatment with the anti- ⁇ 4 ⁇ 7 antibody can be continued if the fecal calprotectin is reduced to less than 50% of the baseline or concentration before treatment.
  • the fecal calprotectin can be reduced to less than 45%, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, between 10-55%, between 10-30%, between 15-35%, between 15-45%, or between 20-40% of the baseline or concentration before treatment.
  • a vedolizumab-treated patient is in deep remission and can continue to be administered vedolizumab if the vedolizumab trough serum concentration is at least 25 ⁇ g/ml, at least 27 ⁇ g/ml, at least 30 ⁇ g/ml, at least 32 ⁇ g/ml or at least 34 ⁇ g/ml.
  • the invention relates to a method for identifying a patient for continued treatment with an anti- ⁇ 4 ⁇ 7 antibody, the method comprising the steps of: measuring the clearance of the anti- ⁇ 4 ⁇ 7 antibody in a biological sample obtained from a patient suffering from inflammatory bowel disease (IBD) and who was administered at least two doses of the anti- ⁇ 4 ⁇ 7 antibody within the previous four months; and identifying the patient for continued treatment with the anti- ⁇ 4 ⁇ 7 antibody if the clearance in the patient is less than 0.25 L/day.
  • IBD inflammatory bowel disease
  • the patient was administered at least one dose of the anti- ⁇ 4 ⁇ 7 antibody within the previous two months, the patient was administered at least two doses of the anti- ⁇ 4 ⁇ 7 antibody within the previous three months or the previous two months, or, the patient was administered at least one dose of the anti- ⁇ 4 ⁇ 7 antibody within the previous two months or within the previous month.
  • the clearance may be less than 0.20 L/day or between 0.1 and 0.2 L/day.
  • the invention relates to a method for identifying a patient for continued treatment with vedolizumab, the method comprising the steps of: measuring the clearance of vedolizumab in a biological sample obtained from a patient suffering from inflammatory bowel disease (IBD) and who was administered at least two doses of vedolizumab within the previous four months; and identifying the patient for continued treatment with vedolizumab if the clearance in the patient is less than 0.25 L/day.
  • the clearance may be less than 0.20 L/day or between 0.1 and 0.2 L/day.
  • the patient was administered at least two doses of vedolizumab within the previous three months or the previous two months.
  • the invention in another aspect, relates to a method for identifying a patient for continued treatment with vedolizumab, the method comprising the steps of: measuring the clearance of vedolizumab in a biological sample obtained from a patient suffering from inflammatory bowel disease (IBD) and who was administered at least one dose of vedolizumab within the previous two months; and identifying the patient for continued treatment with vedolizumab if the clearance in the patient is less than 0.25 L/day.
  • the clearance may be less than 0.20 L/day or between 0.1 and 0.2 L/day.
  • the patient was administered at least one dose of vedolizumab within the previous month.
  • the patient was administered at least two doses of vedolizumab within the previous month.
  • the invention further relates to assays for use in measuring the factors described herein for identifying a patient who will respond to treatment with an anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab.
  • the assay is a pharmacokinetic assay for circulating anti- ⁇ 4 ⁇ 7 antibody.
  • the assay may measure high or sustained positive levels of anti- ⁇ 4 ⁇ 7 antibody, such as at least 8, 10, 12, 14, 17, 20, 25, 30, 35, or 40 ⁇ g per ml in a serum sample from a patient, e.g., for predicting the ability to respond or maintain a response or remission of the IBD afflicting the patient.
  • the serum concentration of the anti- ⁇ 4 ⁇ 7 antibody may be measured by a sandwich ELISA assay.
  • the serum concentration of the anti- ⁇ 4 ⁇ 7 antibody may be measured in an antibody bridging assay.
  • the assay for use in measuring the factors described herein for identifying a patient who will respond to treatment with an anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab is a pharmacodynamic assay for ⁇ 4 ⁇ 7 integrin, e.g., on circulating lymphocytes.
  • low or minimum levels of free ⁇ 4 ⁇ 7 integrin such as less than 10%, less than 7%, less than 5% or less than 3% free ⁇ 4 ⁇ 7 integrin, may predict the effectiveness of the anti- ⁇ 4 ⁇ 7 antibody.
  • the free ⁇ 4 ⁇ 7 integrin may be measured by the amount of MAdCAM binding to the ⁇ 4 ⁇ 7 integrin.
  • the free ⁇ 4 ⁇ 7 integrin may be measured by the amount of anti- ⁇ 4 ⁇ 7 antibody binding to the ⁇ 4 ⁇ 7 integrin.
  • the assay measures immune response to the anti- ⁇ 4 ⁇ 7 antibody.
  • a low or absent immune response to anti- ⁇ 4 ⁇ 7 antibody may predict the ability to respond or maintain a response or remission of the IBD afflicting the patient.
  • the method may further comprise measuring the fecal calprotectin concentration.
  • Treatment with the anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab may be continued with a fecal calprotectin concentration of less than 1500 ⁇ g/g.
  • the fecal calprotectin concentration may be less than 1250 ⁇ g/g, less than 1000 ⁇ g/g, less than 750 ⁇ g/g, less than 500 ⁇ g/g, less than 400 ⁇ g/g, less than 300 ⁇ g/g, less than 250 ⁇ g/g, between 200-1200 ⁇ g/g, between 350 to 800 ⁇ g/g between 300-1000 ⁇ g/g, ⁇ 50 ⁇ g/g, ⁇ 100 ⁇ g/g, ⁇ 150 ⁇ g/g, ⁇ 200 ⁇ g/g, ⁇ 250-499 ⁇ g/g, or between 500 to 900 ⁇ g/g, in a sample from a patient who is likely to respond to the anti- ⁇ 4 ⁇ 7 antibody,
  • treatment with the anti- ⁇ 4 ⁇ 7 antibody, such as vedolizumab can be continued if the fecal calprotectin is reduced to less than 50% of the baseline or concentration before treatment.
  • the fecal calprotectin can be reduced to less than 45%, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, between 10-55%, between 10-30%, between 15-35%, between 15-45%, or between 20-40% of the baseline or concentration before treatment.
  • a method for treating a human patient having inflammatory bowel disease comprises: selecting a human patient having IBD and having a serum concentration of vedolizumab which is at least 8 ⁇ g per ml at a time point that is four weeks after a second dose of vedolizumab, wherein a first dose of vedolizumab was administered to the subject two weeks prior to the second dose of vedolizumab; and administering vedolizumab to the human patient having IBD, thereby treating the human patient having IBD.
  • the first dose of vedolizumab comprises 300 mg.
  • the second dose of vedolizumab comprises 300 mg.
  • the patient received the first and the second dose intravenously.
  • the patient had an inadequate response with, lost response to, or was intolerant to a TNF blocker.
  • One embodiment provided herein is an in vitro method for determining the responsiveness of a human patient having Inflammatory Bowel Disease (IBD) to treatment with vedolizumab, the method comprising measuring the concentration of vedolizumab in a blood sample from the patient by contacting the blood sample with an anti-vedolizumab antibody, wherein the sample is obtained about four weeks following administration of a second dose of vedolizumab, wherein a first dose of vedolizumab was administered to the subject two weeks prior to the second dose of vedolizumab, and wherein a vedolizumab concentration of at least 8 ⁇ g per ml in the blood sample indicates that the patient is responsive to treatment with vedolizumab, and a concentration of less than 8 ⁇ g per ml in the blood sample indicates that the patient is not responsive to treatment with vedolizumab.
  • IBD Inflammatory Bowel Disease
  • the method further comprises administering vedolizumab to the patient.
  • the first dose of vedolizumab comprises 300 mg.
  • the second dose of vedolizumab comprises 300 mg.
  • the patient received the first and the second dose intravenously. In some embodiments, the patient had an inadequate response with, lost response to, or was intolerant to a TNF blocker.
  • FIG. 1 shows the median (interquartile range) of observed vedolizumab trough serum concentration versus nominal sampling time in patients with UC (GEMINI 1) and patients with CD (GEMINI 2) during maintenance treatment with vedolizumab 300 mg Q4W or Q8W or placebo. All patients (including those in the placebo group) received 2 doses of vedolizumab 300 mg during induction (at weeks 0 and 2).
  • FIG. 2 is a diagramatic representation of the population pharmacokinetic model of vedolizumab.
  • Conc concentration
  • K m Michaelis-Menton constant
  • V max maximum rate.
  • FIG. 3 shows distribution of individual vedolizumab linear clearance (CL L ) estimates from the final population pharmacokinetic model in patients with UC and patients with CD.
  • FIG. 4 is a goodness-of-fit plot: observed vedolizumab concentration versus predicted vedolizumab concentration from the final population pharmacokinetic model. Values are indicated by closed circles with a dashed black loss trend line through the data. The line of identity (solid white) is included as reference.
  • FIG. 5 shows the effect of covariates on vedolizumab linear clearance (CL L ).
  • Each point and line represents the median and 95% credible interval (CDI), respectively, of the Bayesian posterior distribution of normalized samples of vedolizumab CL L adjusted for the covariate.
  • the reference individual weighs 70 kg, is 40 years old, has an albumin level of 4 g/dL, fecal calprotectin level of 700 mg/kg, CDAI score of 300 (for patient with CD), partial Mayo score of 6 (for patient with UC), ADA negative, and no concomitant therapy use, and is TNF- ⁇ antagonist therapy na ⁇ ve.
  • Albumin 2.7, 3.2, 3.7, 4.0, 4.2, and 4.7 g/dL represent the 6th, 18th, 50th, 70th, 85th, and 98.5th percentiles, respectively, of baseline albumin levels for patients in GEMINI 1, 2, and 3.
  • the vertical black line is drawn at the reference point estimate, and the shaded region is ⁇ 25% of the reference point estimate chosen to represent an uncertainty range of clinical unimportance.
  • FIG. 6 shows individual vedolizumab linear clearance (CL L ) estimates at the end of induction (week 6) by Mayo clinic endoscopic subscore for patients with UC (GEMINI 1). Midlines represent medians. Box limits represent 25th and 75th percentiles. Whiskers (error bars) represent highest and lowest points within 1.5 ⁇ interquartile range. Individual points represent outliers.
  • FIG. 7 is a receptor ( ⁇ 4 ⁇ 7 ) saturation plot: observed MAdCAM-1 versus observed vedolizumab concentration in patients with UC (GEMINI 1) and patients with CD (GEMINI 2). Baseline data are excluded.
  • FIG. 8 shows observed MAdCAM-1 versus time for patients with UC (GEMINI 1) and patients with CD (GEMINI 2).
  • FIG. 9 is a goodness-of-fit plot for the vedolizumab final population pharmacokinetic model: CWRESI and residual versus time and population predicted vedolizumab concentration.
  • FIG. 10 shows a predictive check for the vedolizumab final population pharmacokinetic model: induction therapy.
  • FIG. 11 shows a predictive check for the vedolizumab final population pharmacokinetic model: maintenance therapy—every 4 weeks.
  • FIG. 12 is a predictive check for the vedolizumab final population pharmacokinetic model: maintenance therapy—every 8 weeks.
  • FIG. 13 is a goodness-of-fit model for MAdCAM-1.
  • FIG. 14 is Percent of Baseline Calprotectin at Week 6 by Treatment or Response Status.
  • FIGS. 15A-15D show clinical, and FIGS. 15E and 15F show HRQoL, outcomes at week 52 in patients in or not in deep remission (definition 1) at week 6.
  • FIGS. 16A-16D show clinical, and FIGS. 16 E and 16 F show HRQoL, outcomes at week 52 in patients in or not in deep remission (definition 2) at week 6.
  • the invention relates to methods for treating with an anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab, a patient having inflammatory bowel disease (IBD), methods for identifying a patient for continued treatment with the antibody, such as vedolizumab, and methods for maintaining remission of IBD in a patient.
  • an anti- ⁇ 4 ⁇ 7 antibody e.g., vedolizumab
  • IBD inflammatory bowel disease
  • Vedolizumab a humanized monoclonal antibody that binds specifically to the ⁇ 4 ⁇ 7 integrin, is indicated for the treatment of patients with moderately to severely active ulcerative colitis (UC) and Crohn's disease (CD).
  • Vedolizumab has a novel gut-selective mechanism of action that differs from that of other currently marketed biologic agents for the treatment for inflammatory bowel disease (IBD), including natalizumab and tumor necrosis factor- ⁇ (TNF- ⁇ ) antagonists.
  • IBD inflammatory bowel disease
  • TNF- ⁇ tumor necrosis factor- ⁇
  • vedolizumab By binding to cell surface—expressed ⁇ 4 ⁇ 7 , vedolizumab blocks the interaction of a subset of memory gut-homing T lymphocytes with mucosal addressin cell adhesion molecule-1 (MAdCAM-1) expressed on endothelial cells. Consequently, migration of these cells into inflamed intestinal tissue is inhibited.
  • MAdCAM-1 mucosal addressin cell adhesion molecule-1
  • Vedolizumab pharmacokinetics was generally linear following an IV infusion over the dose range of 2 to 10 mg/kg in patients with UC. After multiple-dose administration, rapid and near complete ⁇ 4 ⁇ 7 receptor saturation was achieved following the first dose of vedolizumab.
  • vedolizumab induction and maintenance therapy were demonstrated in patients with UC in the GEMINI 1 trial (ClinicalTrials.gov number, NCT00783718) and in patients with CD in the GEMINI 2 (ClinicalTrials.gov number, NCT00783692) and GEMINI 3 (ClinicalTrials.gov number, NCT01224171) trials.
  • the exposure-response (efficacy) relationships of vedolizumab in patients with UC and CD for induction and maintenance therapy have been presented elsewhere.
  • the “trough” serum concentration of an antibody refers to the concentration just before the next dose.
  • CDAI Crohn's Disease Activity Index
  • HBI Hard-Bradshaw Index
  • Endoscopic remission refers to a condition with a low endoscopic score.
  • An example of a method to assess the endoscopic score in ulcerative colitis is flexible sigmoidoscopy.
  • the endoscopic score in ulcerative colitis can be the Mayo subscore.
  • An example of a method to assess the endoscopic score in Crohn's disease is ileocolonoscopy.
  • the endoscopic score in Crohn's disease can be the simple endoscopic score for Crohn's Disease (SES-CD).
  • the SES-CD can include measures such as the size of ulcers, the amount of ulcerated surface, the amount of affected surface and whether and to what extent the alimentary canal is narrowed.
  • a “clinical response” as used herein with reference to ulcerative colitis subjects refers to a reduction in complete Mayo score of 3 or greater points and 30% from baseline, (or a partial Mayo score of 2 or greater points and 25% or greater from baseline, if the complete Mayo score was not performed at the visit) with an accompanying decrease in rectal bleeding subscore of 1 or greater points ( ⁇ 1) or absolute rectal bleeding score of 1 or less point ( ⁇ 1).
  • a “clinical response” as used herein with reference to Crohn's disease subjects refers to a 70 point or greater decrease in CDAI score from baseline (week 0).
  • the terms “clinical response” and “response” e.g., alone without any adjective, are used interchangeably herein.
  • Endoscopic response refers to a percentage decrease in an endoscopic score from baseline (e.g., at screening or just prior to initial dose). In Crohn's disease, endoscopic response can be assessed by a simple endoscopic score for Crohn's Disease (SES-CD).
  • Baseline as used herein describes a value of a parameter which is measured prior to the initial dose of a treatment. It can refer to a measurement of a sample obtained the same day, the day before, during the week before initial treatment, i.e., at a time period before the first dose when little change is expected until after the first dose and values of the measurement obtained after the first dose can be compared to this baseline value to represent the change caused by the dose.
  • Mucosal healing refers to a Mayo endoscopic subscore of less than or equal to 1.
  • “fistula healing” results in closure or elimination of fistulae.
  • mucosal healing refers to an improvement in the amount or severity of wounding in mucosae, e.g., the digestive tract.
  • mucosal healing can refer to a decrease in the amount, size or severity of one or more than one ulcer in the digestive tract.
  • mucosal healing refers to a decrease in one or more parameters selected from the group consisting of wall thickness, enhanced bowel wall contrast, mural edema, ulceration and perienteric vascularity.
  • Such mucosal healing can be expressed as an SES-CD score, or a Magnetic Resonance Index of Activity (MaRIA) score.
  • Complete mucosal healing in Crohn's disease includes absence of ulceration.
  • the “MaRIA score” is the sum of the scores, e.g., as measured by magnetic resonance enterography, of various mucosal healing parameters for each segment of colon and the terminal ileum (e.g., ileum, ascending colon, transverse colon, descending colon, sigmoid, and rectum).
  • Corticosteroid (CS)-free remission refers to patients using oral corticosteroids at baseline who have discontinued corticosteroid use and are in clinical remission at week 52.
  • EQ-5D European Quality of Life-5 Dimension
  • VAS visual analogue scale
  • a composite EQ-5D score can be calculated from the individual scores to assess overall HRQOL.
  • the EQ-5D Visual Analog Scale (VAS) score is a self-assigned rating of overall health using a 20 cm visual, vertical scale, with a score of 0 as the worst and 100 as best possible health.
  • the EQ-5D and EQ-5D VAS have been shown in many studies to be valid and reliable instruments for measuring HRQOL in patients with GI diseases.
  • a decrease of ⁇ 0.3 points in the EQ-5D score represents a clinically meaningful improvement in HRQOL for patients.
  • An increase of greater than or equal to 7 points in the EQ-5D VAS score represents a clinically meaningful improvement in HRQOL for patients.
  • the “Inflammatory Bowel Disease Questionnaire” (IBDQ) questionnaire” (Irvine Journal of Pediatric Gastroenterology & Nutrition 28:S23-27 (1999)) is used to assess quality of life in adult patients with inflammatory bowel disease, ulcerative colitis, or Crohn's Disease and includes 32 questions on four areas of HRQOL: Bowel Systems (10 questions), Emotional Function (12 questions), Social Function (5 questions), and Systemic Function (5 questions). Patients are asked to recall symptoms and quality of life from the last 2 weeks and rate each item on a 7-point Likert scale (higher scores equate to higher quality of life). A total IBDQ score is calculated by summing the scores from each domain; the total IBDQ score ranges from 32 to 224. An IBDQ total score greater than 170 is characteristic of the health related quality of life (HRQoL) of patients in remission.
  • HRQoL health related quality of life
  • induction therapy is an initial stage of therapy, wherein a patient is administered a relatively intensive dosing regimen of a therapeutic agent.
  • the therapeutic agent e.g., antibody
  • the therapeutic agent is administered in a way that quickly provides an effective amount of the agent suitable for certain purposes, such as inducing immune tolerance to the agent or for inducing a clinical response and ameliorating disease symptoms (see WO 2012/151247 and WO 2012/151248, incorporated herein by reference).
  • maintenance therapy is after induction therapy and is administered in a way that continues the response achieved by induction therapy with a stable level of therapeutic agent, e.g., antibody.
  • a maintenance regimen can prevent return of symptoms or relapse of disease, e.g., IBD (see WO 2012/151247 and WO 2012/151248, incorporated herein by reference).
  • a maintenance regimen can provide convenience to the patient, e.g., be a simple dosing regimen or require infrequent trips for treatment.
  • ⁇ 4 ⁇ 7 integrin is a heterodimer of an ⁇ 4 chain (CD49D, ITGA4) and a ⁇ 7 chain (ITGB7). Each chain can form a heterodimer with an alternative integrin chain, to form ⁇ 4 ⁇ 1 or ⁇ E ⁇ 7 .
  • Human ⁇ 4 and ⁇ 7 genes (GenBank (National Center for Biotechnology Information, Bethesda, Md.) RefSeq Accession numbers NM_000885 and NM_000889, respectively) are expressed by B and T lymphocytes, particularly memory CD4+ lymphocytes. Typical of many integrins, ⁇ 4 ⁇ 7 can exist in either a resting or activated state.
  • Ligands for ⁇ 4 ⁇ 7 include vascular cell adhesion molecule (VCAM), fibronectin and mucosal addressin (MAdCAM (e.g., MAdCAM-1)).
  • VCAM vascular cell adhesion molecule
  • MAdCAM mucosal addressin
  • the ⁇ 4 ⁇ 7 integrin mediates lymphocyte trafficking to GI mucosa and gut-associated lymphoid tissue (GALT) through adhesive interaction with mucosal addressin cell adhesion molecule-1 (MAdCAM-1), which is expressed on the endothelium of mesenteric lymph nodes and GI mucosa.
  • antibody herein is used in the broadest sense and specifically covers full length monoclonal antibodies, immunoglobulins, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two full length antibodies, e.g., each to a different antigen or epitope, and individual antigen binding fragments, including dAbs, scFv, Fab, F(ab)′ 2 , Fab′, including human, humanized and antibodies from non-human species and recombinant antigen binding forms such as monobodies and diabodies.
  • multispecific antibodies e.g. bispecific antibodies
  • individual antigen binding fragments including dAbs, scFv, Fab, F(ab)′ 2 , Fab′, including human, humanized and antibodies from non-human species and recombinant antigen binding forms such as monobodies and diabodies.
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991), for example.
  • Antigen binding fragments of an antibody comprise at least the variable regions of the heavy and/or light chains of an anti- ⁇ 4 ⁇ 7 antibody.
  • an antigen binding fragment of vedolizumab comprises amino acid residues 20-131 of the humanized light chain sequence of SEQ ID NO:2.
  • antigen binding fragments include Fab fragments, Fab′ fragments, scFv and F(ab′) 2 fragments of a humanized antibody known in the art.
  • Antigen binding fragments of the humanized antibody of the invention can be produced by enzymatic cleavage or by recombinant techniques. For instance, papain or pepsin cleavage can be used to generate Fab or F(ab′) 2 fragments, respectively.
  • Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site.
  • a recombinant construct encoding the heavy chain of an F(ab′) 2 fragment can be designed to include DNA sequences encoding the CH 1 domain and hinge region of the heavy chain.
  • antigen binding fragments inhibit binding of ⁇ 4 ⁇ 7 integrin to one or more of its ligands (e.g. the mucosal addressin MAdCAM (e.g., MAdCAM-1), fibronectin).
  • Fc receptor or “FcR” are used to describe a receptor that binds to the Fc region of an antibody.
  • the FcR is a native sequence human FcR.
  • the FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc- ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc ⁇ RII receptors include Fc ⁇ RIIA (an “activating receptor”) and Fc ⁇ RIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor Fc ⁇ RIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain.
  • Inhibiting receptor Fc ⁇ RIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain.
  • ITAM immunoreceptor tyrosine-based activation motif
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • FcR neonatal receptor
  • FcRn the neonatal receptor
  • IgG immunoglobulin G
  • albumin albumin
  • hypervariable region when used herein refers to the amino acid residues of an antibody which are responsible for antigen binding and are found in the “variable domain” of each chain.
  • the hypervariable region generally comprises amino acid residues from a “complementarity determining region” or “CDR” (e.g. residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.
  • CDR complementarity determining region
  • “hypervariable loop” e.g. residues 26-32 (L1), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)).
  • “Framework Region” or “FR” residues are those variable domain residues other than the hypervariable region residues as herein defined.
  • the hypervariable region or the CDRs thereof can be transferred from one antibody chain to another or to another protein to confer antigen binding specificity to the resulting (composite) antibody or binding protein.
  • an “isolated” antibody is one which has been identified and separated and/or recovered from a component of its natural environment.
  • the antibody will be purified (1) to greater than 95% by weight of protein as determined by the Lowry method, and alternatively, more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disease as well as those in which the disease or its recurrence is to be prevented. Hence, the patient to be treated herein may have been diagnosed as having the disease or may be predisposed or susceptible to the disease.
  • patient and “subject” are used interchangeably herein.
  • the invention relates to a method of treating IBD in a subject comprising administering to the subject an anti- ⁇ 4 ⁇ 7 antibody described herein in an amount effective to treat IBD, e.g., in humans.
  • the human patient or subject may be an adult (e.g., 18 years or older), an adolescent, or a child.
  • a pharmaceutical composition comprising an anti- ⁇ 4 ⁇ 7 antibody can be used as described herein for treating IBD in a subject suffering therefrom.
  • the anti- ⁇ 4 ⁇ 7 antibody can bind to an epitope on the ⁇ 4 chain (e.g., humanized MAb 21.6 (Bendig et al., U.S. Pat. No. 5,840,299), on the ⁇ 7 chain (e.g., FIB504 or a humanized derivative (e.g., Fong et al., U.S. Pat. No. 7,528,236)), or to a combinatorial epitope formed by the association of the ⁇ 4 chain with the ⁇ 7 chain.
  • an epitope on the ⁇ 4 chain e.g., humanized MAb 21.6 (Bendig et al., U.S. Pat. No. 5,840,299)
  • FIB504 or a humanized derivative e.g., Fong et al., U.S. Pat. No. 7,528,236
  • the antibody binds a combinatorial epitope on the ⁇ 4 ⁇ 7 complex, but does not bind an epitope on the ⁇ 4 chain or the ⁇ 7 chain unless the chains are in association with each other.
  • the association of ⁇ 4 integrin with ⁇ 7 integrin can create a combinatorial epitope for example, by bringing into proximity residues present on both chains which together comprise the epitope or by conformationally exposing on one chain, e.g., the ⁇ 4 integrin chain or the ⁇ 7 integrin chain, an epitopic binding site that is inaccessible to antibody binding in the absence of the proper integrin partner or in the absence of integrin activation.
  • the anti- ⁇ 4 ⁇ 7 antibody binds both the ⁇ 4 integrin chain and the ⁇ 7 integrin chain, and thus, is specific for the ⁇ 4 ⁇ 7 integrin complex.
  • Such antibodies can bind ⁇ 4 ⁇ 7 but not bind ⁇ 4 ⁇ 1, and/or not bind ⁇ E ⁇ 7, for example.
  • the anti- ⁇ 4 ⁇ 7 antibody binds to the same or substantially the same epitope as the Act-1 antibody (Lazarovits, A. I. et al., J. Immunol., 133(4): 1857-1862 (1984), Schweighoffer et al., J. Immunol., 151(2): 717-729, 1993; Bednarczyk et al., J.
  • Murine ACT-1 Hybridoma cell line which produces the murine Act-1 monoclonal antibody, was deposited under the provisions of the Budapest Treaty on Aug. 22, 2001, on behalf of Millennium Pharmaceuticals, Inc., 40 Landsdowne Street, Cambridge, Mass. 02139, U.S.A., at the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209, U.S.A., under Accession No. PTA-3663.
  • the anti- ⁇ 4 ⁇ 7 antibody is a human antibody or an ⁇ 4 ⁇ 7 binding protein using the CDRs provided in U.S. Patent Application Publication No. 2010/0254975.
  • the anti- ⁇ 4 ⁇ 7 antibody inhibits binding of ⁇ 4 ⁇ 7 to one or more of its ligands (e.g. the mucosal addressin, e.g., MAdCAM (e.g., MAdCAM-1), fibronectin, and/or vascular addressin (VCAM)).
  • MAdCAM mucosal addressin
  • VCAM vascular addressin
  • Primate MAdCAMs are described in the PCT publication WO 96/24673, the entire teachings of which are incorporated herein by this reference.
  • the anti- ⁇ 4 ⁇ 7 antibody inhibits binding of ⁇ 4 ⁇ 7 to MAdCAM (e.g., MAdCAM-1) and/or fibronectin without inhibiting the binding of VCAM.
  • the anti- ⁇ 4 ⁇ 7 antibodies for use in the treatments are humanized versions of the mouse Act-1 antibody. Suitable methods for preparing humanized antibodies are well-known in the art.
  • the humanized anti- ⁇ 4 ⁇ 7 antibody will contain a heavy chain that contains the 3 heavy chain complementarity determining regions (CDRs, CDR1, SEQ ID NO:4, CDR2, SEQ ID NO:5 and CDR3, SEQ ID NO:6) of the mouse Act-1 antibody and suitable human heavy chain framework regions; and also contain a light chain that contains the 3 light chain CDRs (CDR1, SEQ ID NO:7, CDR2, SEQ ID NO:8 and CDR3, SEQ ID NO:9) of the mouse Act-1 antibody and suitable human light chain framework regions.
  • CDRs 3 heavy chain complementarity determining regions
  • the humanized Act-1 antibody can contain any suitable human framework regions, including consensus framework regions, with or without amino acid substitutions.
  • one or more of the framework amino acids can be replaced with another amino acid, such as the amino acid at the corresponding position in the mouse Act-1 antibody.
  • the human constant region or portion thereof if present, can be derived from the ⁇ or ⁇ light chains, and/or the ⁇ (e.g., ⁇ 1, ⁇ 2, ⁇ 3, ⁇ 4), ⁇ , ⁇ (e.g., ⁇ 1, ⁇ 2), ⁇ or ⁇ heavy chains of human antibodies, including allelic variants.
  • a particular constant region (e.g., IgG1), variant or portions thereof can be selected in order to tailor effector function.
  • a mutated constant region can be incorporated into a fusion protein to minimize binding to Fc receptors and/or ability to fix complement (see e.g., Winter et al., GB 2,209,757 B; Morrison et al., WO 89/07142; Morgan et al., WO 94/29351, Dec. 22, 1994).
  • Humanized versions of Act-1 antibody were described in PCT publications nos. WO98/06248 and WO07/61679, the entire teachings of each of which are incorporated herein by this reference.
  • the anti- ⁇ 4 ⁇ 7 antibody is vedolizumab.
  • Vedolizumab also called MLN0002, ENTYVIOTM or KYNTELESTM
  • MLN0002, ENTYVIOTM or KYNTELESTM is a humanized immunoglobulin (Ig) G1 mAb directed against the human lymphocyte integrin ⁇ 4 ⁇ 7.
  • Vedolizumab binds the ⁇ 4 ⁇ 7 integrin, antagonizes its adherence to MAdCAM-1 and as such, impairs the migration of gut homing leukocytes into GI mucosa.
  • Vedolizumab is an integrin receptor antagonist indicated for adult patients with moderately to severely active UC or CD who have had an inadequate response with, lost response to, or were intolerant to a tumor necrosis factor (TNF) blocker or immunomodulator, or had an inadequate response with, were intolerant to, or demonstrated dependence on corticosteroids.
  • TNF tumor necrosis factor
  • vedolizumab is for inducing and maintaining clinical response, inducing and maintaining clinical remission, improving endoscopic appearance of the mucosa, and/or achieving corticosteroid-free remission.
  • vedolizumab is for achieving clinical response, achieving clinical remission, and/or achieving corticosteroid-free remission.
  • corticosteroid-free remission is achieved through a tapering regimen during continued treatment with vedolizumab.
  • the humanized anti- ⁇ 4 ⁇ 7 antibody for use in the treatment comprises a heavy chain variable region comprising amino acids 20 to 140 of SEQ ID NO:1, and a light chain variable region comprising amino acids 20 to 131 of SEQ ID NO:2 or amino acids 21 to 132 of SEQ ID NO:3.
  • a suitable human constant region(s) can be present.
  • the humanized anti- ⁇ 4 ⁇ 7 antibody can comprise a heavy chain that comprises amino acids 20 to 470 of SEQ ID NO:1 and a light chain comprising amino acids 21 to 239 of SEQ ID NO:3.
  • the humanized anti- ⁇ 4 ⁇ 7 antibody can comprise a heavy chain that comprises amino acids 20 to 470 of SEQ ID NO:1 and a light chain comprising amino acids 20 to 238 of SEQ ID NO:2.
  • the humanized light chain of vedolizumab e.g., Chemical Abstract Service (CAS, American Chemical Society) Registry number 943609-66-3
  • CAS American Chemical Society
  • LDP-02 has the somewhat hydrophobic, flexible alanine 114 and a hydrophilic site (Aspartate 115) that is replaced in vedolizumab with the slightly hydrophilic hydroxyl-containing threonine 114 and hydrophobic, potentially inward facing valine 115 residue.
  • substitutions to the humanized anti- ⁇ 4 ⁇ 7 antibody sequence can be, for example, mutations to the heavy and light chain framework regions, such as a mutation of isoleucine to valine on residue 2 of SEQ ID NO:10; a mutation of methionine to valine on residue 4 of SEQ ID NO:10; a mutation of alanine to glycine on residue 24 of SEQ ID NO:11; a mutation of arginine to lysine at residue 38 of SEQ ID NO:11; a mutation of alanine to arginine at residue 40 of SEQ ID NO:11; a mutation of methionine to isoleucine on residue 48 of SEQ ID NO:11; a mutation of isoleucine to leucine on residue 69 of SEQ ID NO:11; a mutation of arginine to valine on residue 71 of SEQ ID NO:11; a mutation of threonine to isoleucine on residue 73 of SEQ ID NO:11; or any combination thereof
  • the present invention provides, in a first aspect, a method for treating a patient having inflammatory bowel disease (IBD) with an anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab.
  • the method comprises determining pharmacokinetic factors of the patient.
  • the method comprises selecting for treatment a patient who has low clearance of the antibody.
  • the method comprises measuring the concentration of the anti- ⁇ 4 ⁇ 7 antibody in a biological sample from the patient, e.g., blood, serum, plasma, stool, bowel fluid, saliva, inflammatory exudate, at a time, e.g., at least one, two, three, four, five or six weeks, after receiving at least one prior dose of the antibody.
  • the measurement provides an indication of clearance, for which “low” is defined by the various methods, and results criteria thereof described herein, of obtaining a clearance indication.
  • An indication of clearance may result from the measurement of the concentration of anti- ⁇ 4 ⁇ 7 antibody in the sample, which may be used alone or used in a calculation, as described herein, of the rate the anti- ⁇ 4 ⁇ 7 antibody is eliminated from the body, e.g., the blood volume.
  • the calculation may be based on knowledge of the amount of anti- ⁇ 4 ⁇ 7 antibody in the prior dose, the size of the patient or blood volume, the number of days between the day of the prior dose of the anti- ⁇ 4 ⁇ 7 antibody and the day of obtaining the sample for measurement or from a pharmacokinetics model, e.g., a model as described herein.
  • measurement of serum concentration of anti- ⁇ 4 ⁇ 7 antibody may be an indicator of clearance. Clearance may be affected or further illustrated by other parameters, such as pharmacodynamic factors, clinical factors, inflammation or immune response factors, whose measurement may be used in combination with the measurement of anti- ⁇ 4 ⁇ 7 antibody.
  • An indication of clearance alone or in combination with measurements of one or more other parameters, may be used to predict response to anti- ⁇ 4 ⁇ 7 antibody treatment, identify a patient who is responding to anti- ⁇ 4 ⁇ 7 antibody treatment, select a patient for further treatment with anti- ⁇ 4 ⁇ 7 antibody or monitor the effectiveness of the anti- ⁇ 4 ⁇ 7 antibody during treatment.
  • Low clearance or sufficient amounts of anti- ⁇ 4 ⁇ 7 antibody in the patient indicates that further dosing with anti- ⁇ 4 ⁇ 7 antibody will provide benefit in the treatment of IBD.
  • a method wherein low clearance is indicated, measured or calculated at end of induction comprises further treatment of the patient for maintenance of response or remission, achieving corticosteroid-free remission, improving endoscopic appearance of the mucosa.
  • a patient who has low clearance of the antibody may be characterized, a) by a rate of antibody, e.g., vedolizumab, clearance that is less than about 0.25 L/day, less than 0.20 L/day, between 0.1 to 0.2 L/day, less than 0.15 L/day or less than 0.1.
  • a rate of antibody e.g., vedolizumab
  • a serum concentration of antibody e.g., vedolizumab, that is at least 8 ⁇ g per ml, 10 ⁇ g per ml, 12 ⁇ g per ml, 14 ⁇ g per ml, 17 ⁇ g per ml, 20 ⁇ g per ml, 25 ⁇ g per ml, 30 ⁇ g per ml; 35 ⁇ g per ml, or 40 ⁇ g per ml or has a range of 12-25 ⁇ g per ml, 15-17 ⁇ g per ml, 17-25 ⁇ g per ml, 12-40 ⁇ g per ml, and 17-40 ⁇ g ml.
  • the method for treating a patient having inflammatory bowel disease (IBD) with an anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab comprises the steps of selecting a human patient having IBD from a group of two or more patients having or suffering from IBD that has, at a time point of four weeks after receiving a second dose of vedolizumab, where the first dose of vedolizumab was administered to the subject two weeks prior to the second dose, a serum concentration of at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 ⁇ g per ml.
  • IBD inflammatory bowel disease
  • the patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 ⁇ g per ml.
  • the patient's serum concentration e.g., a trough serum concentration, may be greater than 17 ⁇ g/ml, greater than 25 ⁇ g/ml, or greater than 35 ⁇ g/ml.
  • the patient received the prior dose of vedolizumab about two weeks, about three weeks, about four weeks, about five weeks or about six weeks prior to the sampling for serum vedolizumab measurement. Once such a patient is selected from a group of patients, he or she is administered vedolizumab to treat the IBD.
  • the present invention provides a method for treating a patient having inflammatory bowel disease (IBD) with an anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab.
  • the method using vedolizumab comprises the steps of administering two doses of vedolizumab to a patient suffering from IBD, wherein the second dose is administered about two weeks after the first dose is administered to the patient; waiting a period of time of at least two weeks, at least three weeks, about four weeks or five weeks; measuring the patient's serum concentration of vedolizumab; and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 ⁇ g per ml.
  • the patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 ⁇ g per ml.
  • the patient's serum concentration e.g., a trough serum concentration, may be greater than 17 ⁇ g/ml, greater than 25 ⁇ g/ml, or greater than 35 ⁇ g/ml.
  • At least one dose of the anti- ⁇ 4 ⁇ 7 antibody may be administered to a patient suffering from IBD, waiting at least about two weeks, or optionally, a period of two to five weeks, and then measuring the patient's serum concentration of vedolizumab and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 ⁇ g per ml.
  • the patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 ⁇ s per ml.
  • the patient's serum concentration, e.g., a trough serum concentration may be greater than 17 ⁇ g/ml, greater than 25 ⁇ g/ml, or greater than 35 ⁇ g/ml.
  • the present invention provides a method of identifying a patient (e.g., a patient having mucosal healing) for continued treatment with vedolizumab.
  • the method may comprise the steps of measuring the concentration of vedolizumab in a sample of serum obtained from a patient suffering from IBD and who received at least two doses of vedolizumab within the previous four months (e.g, within the previous three months, within the previous two months), and identifying the patient for continued treatment with vedolizumab if the serum concentration in the sample is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 ⁇ g per ml.
  • the patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 ⁇ g per ml.
  • the patient's serum concentration e.g., a trough serum concentration, may be greater than 17 ⁇ g/ml, greater than 25 ⁇ g/ml, or greater than 35 ⁇ g/ml.
  • the patient received the last prior dose, e.g., the second dose, of vedolizumab about four weeks prior to the sampling for serum vedolizumab measurement. In other embodiments, the patient received the last prior dose 3 to 8 weeks prior to the sampling for serum vedolizumab measurement.
  • the method of identifying a patient for continued treatment with an anti- ⁇ 4 ⁇ 7 antibody may comprise the steps of measuring the concentration of vedolizumab in a sample of serum obtained from a patient suffering from IBD and who received at least one dose of vedolizumab within the previous one or two months, and identifying the patient for continued treatment with vedolizumab if the serum concentration in the sample is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 ⁇ s per ml.
  • the patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 ⁇ g per ml.
  • the patient's serum concentration e.g., a trough serum concentration
  • the patient's serum concentration may be greater than 17 ⁇ g/ml, greater than 25 ⁇ g/ml, or greater than 35 ⁇ g/ml.
  • the patient received the prior dose of vedolizumab about two weeks, about three weeks, about four weeks, about five weeks or about six weeks prior to the sampling for serum vedolizumab measurement.
  • At least one dose of the anti- ⁇ 4 ⁇ 7 antibody may be administered to a patient suffering from IBD, waiting at least about two weeks, or optionally, a period of two to five weeks, and then measuring the patient's serum concentration of vedolizumab and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 ⁇ g per ml.
  • the patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 ⁇ g per ml.
  • the patient's serum concentration, e.g., a trough serum concentration may be greater than 17 ⁇ g/ml, greater than 25 ⁇ g/ml, or greater than 35 ⁇ g/ml.
  • Vedolizumab may be administered by any suitable method, such as by one or more of intravenous injection, subcutaneous injection, or infusion. In some embodiments, vedolizumab is administered at a dose of 50 mg, 100 mg, 180 mg, 300 mg, or 600 mg. In some embodiments, the vedolizumab is administered, for example subcutaneously, at a dose of 0.5 mg/kg, 1.0 mg/kg, 1.5 mg/kg, 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg. 4.0 mg/kg, or 5.0 mg/kg, at a dose of 108 mg, 216 mg, 160 mg or 165 mg. The vedolizumab may be administered once per day, per week, per month, or per year.
  • a vedolizumab dosing regimen may have an initial or induction phase and a maintenance phase.
  • An induction phase may be one or more than one, e.g., two, three or four doses, of high amounts or without long times, such as only one week, two weeks, three weeks or four weeks between each dose.
  • an induction regimen may have two doses, one at day (week) zero and one at week 2 (day 14).
  • a maintenance phase e.g., to maintain remission of the IBD, may have lower doses or doses further apart than in the induction phase.
  • the vedolizumab is administered at zero, two and six weeks (induction), and then every four weeks or every eight weeks thereafter (maintenance).
  • vedolizumab is administered intravenously at zero, two and six weeks, then every eight weeks thereafter. In some embodiments, vedolizumab is administered one or more times, and then at least one month, at least six months, or at least one year later, vedolizumab is again administered one or more times. In some embodiments, 300 mg vedolizumab may be administered by intravenous infusion at zero, two, and six weeks, and then at four weeks intervals or eight week intervals thereafter.
  • 300 mg vedolizumab may be administered by intravenous infusion at zero, two, and six weeks, and then at two-, three- or four-week intervals, 108 mg of vedolizumab may be administered subcutaneously.
  • Treatment methods using anti- ⁇ 4 ⁇ 7 integrin antibodies are described in publication nos. U.S. 2005/0095238, WO2012151248 and WO 2012/151247.
  • the method of identifying a patient for continued treatment with an anti- ⁇ 4 ⁇ 7 antibody may comprise a clinical measure.
  • a clinical measure may be mucosal healing.
  • an endoscopic score of less than 4, less than 3, ⁇ 1, or between 0 and 2 may help identify a patient for continued treatment.
  • a patient with mucosal healing and thus a candidate for continued treatment with an anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab, may have an assessment selected from the group consisting of endoscopic response, endoscopic remission, improvement in the MaRIA score, decrease in ulceration, and improvement in a mucosal healing parameter, e.g., an MREn parameter.
  • an anti- ⁇ 4 ⁇ 7 antibody e.g., vedolizumab
  • RCE Relative Contrast Enhancement
  • endoscopic response is achieved by about a 25% decrease, about a 40% decrease, about a 50% decrease, about a 60% decrease or about a 75% decrease in SES-CD score from baseline.
  • endoscopic remission is achieved by an SES-CD score of ⁇ 6, ⁇ 5, ⁇ 4 or ⁇ 3.
  • mucosal healing is achieved by a MaRIA score of ⁇ 14, ⁇ 13, ⁇ 12, ⁇ 11, ⁇ 10, ⁇ 9, ⁇ 8, ⁇ 7, ⁇ 6, ⁇ 5 or ⁇ 4.
  • a decrease in ulceration is selected from the group consisting of a decrease in the size of ulcers, a decrease in the percent of ulcerated surface, a decrease in the percent of affected surface and decrease in the narrowings of the canal.
  • the size of ulcers is less than 2 cm in diameter, 0.5 to 2 cm in diameter, 0.1 to 0.5 cm in diameter or ⁇ 0.2 cm diameter.
  • the ulcerated surface is less than 30%, 10-30%, less than 10%, or 0.
  • the affected surface is less than 75%, 50%-75%, less than 50%, less than 25% or unaffected.
  • the wall thickness is decreased by about 10%, about 15%, about 20%, about 25%, about 30%, about 20%-40% or more than 45% from baseline.
  • the bowel wall contrast (RCE) is decreased about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 20%-40% or more than 40% from baseline.
  • the mural edema is decreased by about 25%, about 40%, about 50%, about 60%, about 70%, more than 75%, or about 70%-90% from baseline.
  • the perienteric vascularity is decreased by about 25%, about 30%, about 40%, about 50%, about 60%, about 50%-70%, about 75% or more than 75% from baseline.
  • a Crohn's disease patient who is responsive to vedolizumab has an SES-CD of ⁇ 4 at week 6, 10, 12, 14, 22 or 26 after initiating treatment. In an embodiment, a Crohn's disease patient who is responsive to vedolizumab has an endoscopic response or ⁇ 50% reduction of SES-CD at week 6, 10, 12, 14, 22 or 26 after initiating treatment. In an embodiment, a Crohn's disease patient who is responsive to vedolizumab has a clinical remission or ⁇ 70 or ⁇ 100 point reduction of CDAI score at week 6, 10, 12, or 14 after initiating treatment.
  • a Crohn's disease patient who is responsive to vedolizumab has a MaRIA score of ⁇ 15, ⁇ 12, ⁇ 10 or ⁇ 7 at week 6, 10, 12, 14, respectively, globally or on a per segment basis, after initiating treatment.
  • a Crohn's disease patient who is responsive to vedolizumab has no ulceration at week 14, 22 or 26 after initiating treatment.
  • a Crohn's disease patient who is responsive to vedolizumab has no fistulae at week 14, 22, 26 or 30 after initiating treatment.
  • a patient who is responsive to vedolizumab may continue to be treated, e.g., may continue a maintenance regimen, with vedolizumab.
  • a maintenance regimen comprises a dose of vedolizumab once every 2 weeks. In one embodiment, a maintenance regimen comprises a dose of vedolizumab once every 4 weeks. In one embodiment, a maintenance regimen comprises a dose of vedolizumab once every 6 weeks. In one embodiment, a maintenance regimen comprises a dose of vedolizumab once every 8 weeks.
  • the present invention relates to a method for maintaining remission of inflammatory bowel disease in a patient.
  • the patient may have received at least one dose of vedolizumab in the previous two months, the previous three months or the previous four months.
  • the method may comprise the steps of obtaining a serum sample from the patient, measuring the concentration of vedolizumab in the sample, and administering vedolizumab thereafter every eight weeks if the concentration is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 ⁇ g per ml.
  • the patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 ⁇ g per ml.
  • the patient's serum concentration e.g., a trough serum concentration
  • the patient's serum concentration may be greater than 17 ⁇ g/ml, greater than 25 ⁇ g/ml, or greater than 35 ⁇ g/mi.
  • the patient received the prior dose of vedolizumab about two weeks, about three weeks, about four weeks, about five weeks or about six weeks prior to the sampling for serum vedolizumab measurement.
  • a method for continuing to treat an IBD patient with vedolizumab comprises the steps of providing two doses of vedolizumab, wherein the second dose is about two weeks after the first; waiting about four weeks; and continuing to treat the patient with vedolizumab if the patient is in deep remission.
  • Deep remission may be determined by measuring an endoscopic subscore for the patient and is defined as having an endoscopic subscore of 0 to 1.
  • the method may further comprise the step of determining a patient-reported outcome score (e.g., a subscore of 0 to 1).
  • the patient-reported outcome may comprise a rectal bleeding subscore (e.g., 0) and/or a stool frequency subscore (e.g., decrease or no change; 0 or 1).
  • the invention also relates to a method for identifying a patient for continued treatment with vedolizumab comprising the steps of measuring the clearance of vedolizumab in a biological sample obtained from a patient suffering from IBD and who was administered at least two doses of vedolizumab within the previous four months (e.g., within the previous three months, within the previous two months), and identifying the patient for continued treatment with vedolizumab if the clearance in the patient is less than 0.25 L/day, less than 0.20 L/day, or between 0.1 to 0.2 L/day.
  • the biological sample may be any biological sample, for example, serum, plasma, saliva, urine, or feces.
  • the method may further comprise measuring anti- ⁇ 4 ⁇ 7 antibody antibodies.
  • a low or absent immune response to the anti- ⁇ 4 ⁇ 7 antibody e.g., anti-vedolizumab antibody, e.g., a titer of ⁇ 50, ⁇ 125 or ⁇ 575 may further identify the patient for continued treatment with vedolizumab.
  • anti-vedolizumab antibody e.g., a titer of ⁇ 50, ⁇ 125 or ⁇ 575
  • Methods to determine whether a patient will respond to treatment with an anti- ⁇ 4 ⁇ 7 antibody or whether to continue treating a patient with an anti- ⁇ 4 ⁇ 7 antibody may further comprise measuring albumin concentration. In therapeutic antibody therapy, this can be a reflection of clearance activity, such as ability to bind the neonate FcR. In cases of low serum albumin levels, the anti- ⁇ 4 ⁇ 7 antibody can have a high clearance. Consequently, a patient with high serum albumin levels may not respond or may take longer to respond to treatment with anti- ⁇ 4 ⁇ 7 antibody.
  • An albumin concentration greater than about 3.0 g/dL, about 3.2 g/dL, about 4.0 g/dL, about 4.7 g/dL, or about 5.0 g/dL, or in the range of 3.3 to 5.0 g/dL, in the range of 3.5 to 5.0 g/dL, in the range of 3.8 to 5.0 g/dL or in the range of 4.0 to 5.0 g/dL may further identify the patient for continued treatment with the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab.
  • the albumin concentration measurement may be accompanied by measurement of patient weight.
  • a high weight patient e.g., greater than 90 kg, greater than 100 kg, greater than 110 kg, or greater than 120 kg, with low albumin levels, e.g., less than 4.2 g/dL, less than 4.0 g/dL, less than 3.5 g/dL or less than 3.2 g/dL, may have high anti- ⁇ 4 ⁇ 7 antibody clearance and thus may not respond to therapy with the anti- ⁇ 4 ⁇ 7 antibody or may need a higher or more frequent dose of the anti- ⁇ 4 ⁇ 7 antibody for continued treatment.
  • Clearance e.g., linear clearance, e.g., the volume of blood which is cleared of drug per unit time
  • linear clearance e.g., the volume of blood which is cleared of drug per unit time
  • clearance may be estimated by population approach or Bayesian methods, e.g., the full Bayesian Markov Chain Monte Carlo (MCMC) method.
  • MCMC Bayesian Markov Chain Monte Carlo
  • the anti- ⁇ 4 ⁇ 7 antibody exposure metric such as trough serum concentration, e.g., serum concentration of anti- ⁇ 4 ⁇ 7 antibody prior, e.g., 1 day, 2 days, 3 days, 4 days or up to a week prior, to administering a new dose, peak serum concentration, average serum concentration measured at more than one sampling or area under the concentration time curve, is inputted into the model to determine clearance.
  • trough serum concentration e.g., serum concentration of anti- ⁇ 4 ⁇ 7 antibody prior, e.g., 1 day, 2 days, 3 days, 4 days or up to a week prior
  • the method for identifying a patient for continued treatment with vedolizumab comprising the steps of measuring the clearance of vedolizumab in a biological sample obtained from a patient suffering from IBD can be performed on a patient who was administered at least one dose of vedolizumab within the previous one or two months, and identifying the patient for continued treatment with vedolizumab if the clearance in the patient is less than 0.25 L/day, less than 0.20 L/day, between 0.1 to 0.2 L/day, less than 0.15 L/day or less than 0.1. L/day.
  • the biological sample may be any biological sample, for example, serum, plasma, saliva, urine, or feces.
  • the method may further comprise measuring an endoscopic subscore.
  • Anti- ⁇ 4 ⁇ 7 antibody e.g., vedolizumab treatment may be continued with an endoscopic subscore of less than about 3, less than about 2.5, less than about 2, between about 0-2, or less than or equal to 1.
  • a non-diseased subject will have a fecal calprotectin level of less than 50 ⁇ g/g.
  • a fecal calprotectin level greater than 50 but less than 150 ⁇ g/g may be a sign of possible mucosal inflammation, whereas fecal calprotectin levels greater than 150 ⁇ g/g is usually a sign of active inflammation.
  • the methods described herein may further comprise measuring the fecal calprotectin concentration. Higher levels of fecal calprotectin are associated with a greater risk of relapse.
  • Vedolizumab treatment may be continued with a fecal calprotectin concentration of less than 1500 ⁇ g/g, less 1250 ⁇ g/g, less than 1000 ⁇ g/g, less than 750 ⁇ g/g, less than 500 ⁇ g/g, less than 400 ⁇ g/g, less than 300 ⁇ g/g, less than 250 ⁇ g/g, between 200-1200 ⁇ g/g, between 350 to 800 ⁇ g/g, between 300-1000 ⁇ g/g, ⁇ 50 ⁇ g/g, ⁇ 100 ⁇ g/g, ⁇ 150 ⁇ g/g, ⁇ 200 ⁇ g/g, ⁇ 250-499 ⁇ g/g, or between 500 to 900 ⁇ g/g.
  • a fecal calprotectin concentration of less than 1500 ⁇ g/g, less 1250 ⁇ g/g, less than 1000 ⁇ g/g, less than 750 ⁇ g/g, less than 500 ⁇ g/g, less than 400 ⁇ g/g,
  • fecal calprotectin may be reduced to less than about 50%, 45%, 40%, 35%, 30%, 25%, 20%, between 10-55%, between 10-30%, between 15-35%, between 15-45% or between 20-40% of the baseline or concentration before treatment.
  • Fecal calprotectin in a stool sample can be measured using the PHICAL test kit (Calpro, Lysaker Norway).
  • the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab, serum concentration may be measured by any appropriate means known by those skilled in the art.
  • the vedolizumab serum concentration is measured by a sandwich enzyme-linked immunosorbent assay (ELISA) assay.
  • ELISA sandwich enzyme-linked immunosorbent assay
  • use of a pharmacodynamic assay inhibition of MAdCAM-1-Fc binding to ⁇ 4 ⁇ 7 -expressing peripheral blood cells by the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab in the blood is used as a measure of the extent of ⁇ 4 ⁇ 7 saturation by the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab.
  • the anti- ⁇ 4 ⁇ 7 antibody amount in serum can be measured in a pharmacokinetic assay.
  • An immobilized phase such as a microtiter plate, vessel or bead is coated with a reagent which specifically binds to the anti- ⁇ 4 ⁇ 7 antibody.
  • the immobilized reagent is contacted with a patient sample, e.g., serum, which may or may not comprise the anti- ⁇ 4 ⁇ 7 antibody.
  • a patient sample e.g., serum
  • the anti- ⁇ 4 ⁇ 7 antibody complexed to the coating reagent is contacted with a reagent which binds to the captured antibody and may be detected, e.g., using a label such as horseradish peroxidase (HRP).
  • HRP horseradish peroxidase
  • the binding reagent may be an anti-human antibody, e.g., polyclonal or monoclonal, which binds to the Fc portion of the anti- ⁇ 4 ⁇ 7 antibody.
  • Addition of an HRP substrate such as 3,3′,5,5′-tetramethylbenzidine (TMB)
  • TMB 3,3′,5,5′-tetramethylbenzidine
  • the coating reagent is an anti-idiotypic antibody which specifically binds to the anti- ⁇ 4 ⁇ 7 antibody, e.g., its variable region or a portion thereof comprising one or more CDRs, such as heavy chain CDR3, SEQ ID NO:6.
  • the anti-idiotypic anti- ⁇ 4 ⁇ 7 antibody for use in the assay can be specific for, and thus bind, the ⁇ 4 ⁇ 7 integrin-binding portion of the anti- ⁇ 4 ⁇ 7 antibody but is not specific for the Fc portion of the anti- ⁇ 4 ⁇ 7 antibody and thus does not bind the Fc portion of the anti- ⁇ 4 ⁇ 7 antibody.
  • the anti-idiotypic anti- ⁇ 4 ⁇ 7 antibody for use in the assay can be specific for, and thus bind, a variable region of the heavy and/or light chain of anti- ⁇ 4 ⁇ 7 antibody, e.g., selected from the group consisting of amino acids 20 to 140 of SEQ ID NO:1, amino acids 20 to 131 of SEQ ID NO:2 and amino acids 21 to 132 of SEQ ID NO:3.
  • the anti-idiotypic anti- ⁇ 4 ⁇ 7 antibody for use in the assay can be specific for, and thus bind, an antigen-binding fragment of the anti- ⁇ 4 ⁇ 7 antibody.
  • the anti-idiotypic antibody can be isolated from an immunization process using the anti- ⁇ 4 ⁇ 7 antibody or an ⁇ 4 ⁇ 7 integrin-binding portion thereof, such as an antibody fragment comprising one or more CDRs, and used as isolated or produced by a recombinant method.
  • the anti-idiotypic anti- ⁇ 4 ⁇ 7 antibody is raised against an immunogen comprising heavy chain CDR3, SEQ ID NO:6.
  • the anti-idiotypic anti- ⁇ 4 ⁇ 7 antibody is raised against an immunogen comprising a variable region of the heavy and/or light chain of anti- ⁇ 4 ⁇ 7 antibody, e.g., selected from the group consisting of amino acids 20 to 140 of SEQ ID NO:1, amino acids 20 to 131 of SEQ ID NO:2 and amino acids 21 to 132 of SEQ ID NO:3.
  • the anti-idiotypic antibody is a monoclonal antibody.
  • an scFv fragment of the anti-idiotypic antibody is used in the assay.
  • the intact anti-idiotypic antibody is used in the assay.
  • an anti-idiotypic anti- ⁇ 4 ⁇ 7 antibody can proceed in the following general methods. Immunization of a suitable animal (e.g., mouse, rat, rabbit or sheep) with protein, e.g., anti- ⁇ 4 ⁇ 7 antibody or an ⁇ 4 ⁇ 7 integrin binding portion thereof, or fusion protein comprising the portion, can be performed with the immunogen prepared for injection in a manner to induce a response, e.g., with adjuvant, e.g., complete Freund's adjuvant.
  • adjuvants include TITERMAX GOLD® adjuvant (CYTRX Corporation, Los Angeles, Calif.) and alum.
  • Small peptide immunogens such as a fragment comprising a CDR, such as CDR3 of the heavy chain can be linked to a larger molecule, such as keyhole limpet hemocyanin.
  • Mice can be injected in a number of manners, e.g., subcutaneous, intravenous or intramuscular at a number of sites, e.g., in the peritoneum (i.p.), base of the tail, or foot pad, or a combination of sites, e.g., i.p. and base of tail.
  • Booster injections can include the same or a different immunogen and can additionally include adjuvant, e.g., incomplete Freund's adjuvant.
  • a hybridoma is produced by fusing a suitable cell from an immortal cell line (e.g., a myeloma cell line such as SP2/0, P3X63Ag8.653 or a heteromyeloma) with antibody-producing cells.
  • an immortal cell line e.g., a myeloma cell line such as SP2/0, P3X63Ag8.653 or a heteromyeloma
  • Antibody-producing cells can be obtained from the peripheral blood or, preferably the spleen or lymph nodes, of animals immunized with the antigen of interest.
  • Cells that produce antibodies can be produced using suitable methods, for example, fusion of a human antibody-producing cell and a heteromyeloma or trioma, or immortalization of an activated human B cell via infection with Epstein Barr virus.
  • the fused or immortalized antibody-producing cells can be isolated using selective culture conditions, and cloned by limiting dilution. Cells which produce antibodies with the desired specificity can be identified using a suitable assay (e.g., ELISA (e.g., with immunogen immobilized on the microtiter well).
  • a suitable assay e.g., ELISA (e.g., with immunogen immobilized on the microtiter well).
  • the anti- ⁇ 4 ⁇ 7 antibody or the anti-idiotypic anti- ⁇ 4 ⁇ 7 antibody may be produced by expression of nucleic acid sequences encoding each chain in living cells, e.g., cells in culture.
  • a variety of host-expression vector systems may be utilized to express the antibody molecules of the invention.
  • host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an anti- ⁇ 4 ⁇ 7 antibody in situ. These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia ) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3, NSO cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter)
  • mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
  • CHO Chinese hamster ovary cells
  • a vector such as the major intermediate early gene promoter element from human cytomegalovirus
  • a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed.
  • vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • Such vectors include, but are not limited to, the E. coli expression vector pUR278 (Ruther et al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • the antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • the coating reagent is a ligand of the antibody, such as MAdCAM or an ⁇ 4 ⁇ 7 integrin-binding fragment thereof or fusion protein comprising an ⁇ 4 ⁇ 7-integrin binding fragment of MAdCAM fused with a non-MAdCAM protein, such as an immunoglobulin G constant domain.
  • a ligand of the antibody such as MAdCAM or an ⁇ 4 ⁇ 7 integrin-binding fragment thereof or fusion protein comprising an ⁇ 4 ⁇ 7-integrin binding fragment of MAdCAM fused with a non-MAdCAM protein, such as an immunoglobulin G constant domain.
  • the human anti-anti- ⁇ 4 ⁇ 7 antibody activity can be determined by detecting and/or measuring anti-drug antibodies (ADAs) or antibodies specific to the anti- ⁇ 4 ⁇ 7 antibody (anti-vedolizumab antibodies).
  • ADAs anti-drug antibodies
  • anti-vedolizumab antibodies antibodies specific to the anti- ⁇ 4 ⁇ 7 antibody
  • a screening assay can use a bridging ELISA in which the plate is coated with the anti- ⁇ 4 ⁇ 7 antibody.
  • the immobilized anti- ⁇ 4 ⁇ 7 antibody captures the ADA in the test sample which is bound by an anti- ⁇ 4 ⁇ 7 antibody conjugated to biotin, which is tagged by horseradish peroxidase (HRP)-labeled streptavidin, then detected with an enzymatic substrate, such as TMB.
  • HRP horseradish peroxidase
  • TMB horseradish peroxidase
  • a positive color development e.g., as measured in a microplate reader, such as Spectramax, with analytical software, such as SOFTMAX Pro3.1.2, indicates the presence of ADAs in the sample.
  • the assay cut point e.g., in biotin-avidin-HRP based bridging assay, can be determined by using normal human serum samples as negative controls.
  • the mean absorbance values of the 10 negative control serums can be added to 1.65 times the standard deviation of the negative controls to determine the cut point.
  • the cut point can allow for approximately a 5% false positive rate.
  • low titer responses are interfered with such that they may become undetectable, although high levels of immunogenicity are detectable at vedolizumab concentrations greater than 1 ⁇ g/mL.
  • the standard assay sensitivity can be 0.44 ng/ml
  • the sensitivity of the assay can be 180 ng/ml.
  • serum samples can be taken greater than 4 weeks, greater than 8 weeks, greater than 12 weeks or greater than 16 weeks after the final dose of anti- ⁇ 4 ⁇ 7 antibody. With a longer time period between the prior dose and the sampling, serum drug levels typically can be below the interference level.
  • Another assay method uses streptavidin coated plates, biotin-labeled anti- ⁇ 4 ⁇ 7 antibody anchored to streptavidin coated vessels, beads or microtiter plates for the immobilized side of the bridge and heavy metal, such as ruthenium, osmium or rhenium-labeled (e.g., via a sulfo tag) anti- ⁇ 4 ⁇ 7 antibody for the other side of the bridge.
  • the bridged complex can be built on the plate by stepwise additions and washes between or in solution, with both sides of the bridge contacting diluted serum sample, then transferred to the plate.
  • An example of an assay using this method has a sensitivity of 3.90 ng/ml anti-anti- ⁇ 4 ⁇ 7 antibody.
  • Detection of the heavy metal labeled bridge complex e.g., a ruthenium-labeled complex, by electrochemiluminescence (ECL), e.g., in a Meso Scale Discovery Sector Imager 6000 (Rockville, Md.), may be more sensitive than an HRP method and/or have higher tolerance to the amount of anti- ⁇ 4 ⁇ 7 antibody in the serum. Thus there would not be a need to wait for a delayed sample after the serum drug level lowers.
  • ECL electrochemiluminescence
  • pretreatment of the serum sample with acid e.g., acetic acid or low pH glycine
  • acid e.g., acetic acid or low pH glycine
  • pretreatment of the serum sample with acid e.g., acetic acid or low pH glycine
  • to release the anti- ⁇ 4 ⁇ 7 antibody from the patient-derived anti-anti- ⁇ 4 ⁇ 7 antibodies prior to contacting with the bridging anti- ⁇ 4 ⁇ 7 antibodies can reduce the interference from the drug in the serum.
  • acid e.g., acetic acid or low pH glycine
  • an assay to detect anti-vedolizumab antibodies in a sample of serum from a patient comprises diluting serum by a standard dilution factor, such as 1:5, 1:25, 1:50, and/or 1:125; treating with acetic acid; combining the acid treated diluted sample with an assay composition comprising a high pH reagent, such as high concentration TRIS buffer for neutralizing the acid, a biotin-labeled vedolizumab and a ruthenium-labeled vedolizumab for a time sufficient to form a bridge with serum-derived anti-vedolizumab antibodies between the two tagged versions of vedolizumab; transferring the complexes to a streptavidin-coated plate; washing the plate so only ruthenium complexed by the antibody bridge is present.
  • a standard dilution factor such as 1:5, 1:25, 1:50, and/or 1:125
  • an assay composition comprising a high pH reagent, such as high concentration
  • Detection of the bound ruthenium-labeled complex and measuring the sample by electrochemiluminescence in the microplate reader can be achieved by adding a read solution such as tripropylamine and applying voltage to stimulate the ruthenium label complexed to the plate via the antibody bridge.
  • a read solution such as tripropylamine
  • samples can be further tested in a confirmatory assay that uses excess unlabeled anti- ⁇ 4 ⁇ 7 antibody to demonstrate specificity. Confirmed positive samples can be further assessed for the ability of the HAHA to neutralize the binding of the anti- ⁇ 4 ⁇ 7 antibody, e.g., vedolizumab to cells.
  • a competitive flow cytometry-based assay was designed to determine the ability of the immune serum to inhibit the binding of labeled vedolizumab to an ⁇ 4 ⁇ 7 integrin-expressing cell line, RPMI8866, and detection by flow cytometry.
  • the results can indicate categories of immunogenicity status: Negative: no positive HAHA sample; Positive: at least 1 positive HAHA sample; Transiently positive: at least 1 positive HAHA sample and no consecutive positive HAHA samples; and Persistently positive: at least 2 or more consecutive positive HAHA samples.
  • Negative patients are likely to respond to anti- ⁇ 4 ⁇ 7 antibody and can continue being treated with the antibody.
  • Persistently positive patients are likely to have high clearance of anti- ⁇ 4 ⁇ 7 antibody and may not respond to anti- ⁇ 4 ⁇ 7 antibody treatment.
  • Positive patients may have high clearance of anti- ⁇ 4 ⁇ 7 antibody and may not respond to anti- ⁇ 4 ⁇ 7 antibody.
  • Positive patients can have an additional serum sample 2, 3, 4, 5 or 6 weeks after another dose of anti- ⁇ 4 ⁇ 7 antibody to determine if they are persistently positive or transiently positive. Transiently positive patients are likely to respond to anti- ⁇ 4 ⁇ 7 antibody treatment and treatment of these patients can be continued.
  • Titers of immunogenicity levels also may be determined. Titer categories include ⁇ 5 (low), ⁇ 50, ⁇ 125, ⁇ 625 and ⁇ 3125 (high). A patient with a high titer in a positive sample may have high clearance of anti- ⁇ 4 ⁇ 7 antibody and may not respond to anti- ⁇ 4 ⁇ 7 antibody treatment. A patient with a low titer in a positive sample may respond to anti- ⁇ 4 ⁇ 7 antibody treatment.
  • Vedolizumab serum concentrations were determined using a sandwich enzyme-linked immunosorbent assay (ELISA), with a lower limit of detection of 0.00125 ⁇ g/mL at a 1:100 dilution.
  • the accuracy of the assay was 10.1% coefficient of variation (CV), intra-sample precision ranged from 1.8% to 3.0% CV, and inter-sample precision ranged from 4.0% to 16.0% CV.
  • a MAdCAM-1-Fc binding interference flow cytometry assay was developed.
  • inhibition of MAdCAM-1-Fc binding to ⁇ 4 ⁇ 7 -expressing peripheral blood cells by vedolizumab in the blood is used as a measure of the extent of ⁇ 4 ⁇ 7 saturation by vedolizumab [1].
  • the assay which was developed by Millennium Pharmaceuticals, Inc. (d/b/a Takeda Pharmaceuticals International, Co.), demonstrated an overall intra-sample variability of 6% CV and an intra-subject variability of 20% CV.
  • ADAs anti-drug antibodies
  • anti-vedolizumab antibodies antibodies specific to the anti- ⁇ 4 ⁇ 7 antibody
  • the plate is coated with the anti- ⁇ 4 ⁇ 7 antibody.
  • the immobilized anti- ⁇ 4 ⁇ 7 antibody captures the ADA in the test sample.
  • the bound complex captures an anti- ⁇ 4 ⁇ 7 antibody conjugated to biotin, which after another wash, captures HRP-labeled streptavidin.
  • TMB substrate a positive color development indicates the presence of ADAs in the sample.
  • vedolizumab serum concentration measurements that were missing, or any values with unknown or missing associated observation times, dose times, dose amounts, or dosing intervals, were excluded from the analysis.
  • BLQ limit of quantification
  • Covariates present in the population pharmacokinetic dataset were serum C-reactive protein (CRP), serum albumin, fecal calprotectin, body weight, disease activity (Crohn's Disease Activity Index [CDAI], complete Mayo score, partial Mayo score), Mayo endoscopic subscore, age, sex, ADA status (positive or negative), prior TNF- ⁇ antagonist therapy status (na ⁇ ve or failed), body mass index (BMI), serum globulin, diagnosis (CD or UC), lymphocyte count, and concomitant therapy use (methotrexate, azathioprine, mercaptopurine, or aminosalicylates).
  • CRP C-reactive protein
  • CDAI Crohn's Disease Activity Index
  • the start date and end date of concomitant therapy was populated in the dataset to evaluate the time-dependent effects of concomitant treatments.
  • Covariates with missing data were imputed using different imputation methods, based on the remaining available data (e.g. median of the remaining values). No covariates present in the population pharmacokinetic dataset were missing more than 10% of values.
  • the population pharmacokinetic analysis for repeated measures was conducted using a nonlinear mixed effects modeling approach (NONMEM 7, Version 7.2; ICON Development Solutions, Ellicott City, Md., USA) [13].
  • the base population pharmacokinetic model was developed using the first-order conditional estimation with the ⁇ - ⁇ interaction (FOCEI) method and extensively sampled phase 1 and 2 data. Results from the base model were subsequently used as prior information to selectively inform a subset of population pharmacokinetic model parameters in the full covariate model, which was fit to sparse phase 3 data from GEMINI 1, 2, and 3.
  • the full covariate model was fit to the phase 3 data using the full Bayesian Markov Chain Monte Carlo (MCMC) method.
  • predefined covariate parameter relationships were identified based on exploratory graphics, scientific interest, and mechanistic plausibility.
  • a full covariate model was constructed with care to avoid correlation or collinearity in predictors; covariates with correlation coefficients greater than ⁇ 0.35 were not simultaneously included as potential predictors. Construction of the full model was also guided by evaluating the adequacy of the study design and covariate data to support quantification of the covariate effects of interest.
  • TPV exp ⁇ ( ⁇ n + ⁇ l m ⁇ log ⁇ ( cov m ⁇ ⁇ i ref m ) ⁇ ⁇ ( m + n ) + ⁇ l p ⁇ ⁇ ( p + m + n ) cov ⁇ ⁇ pi )
  • the final population pharmacokinetic model and parameter estimates were investigated with a predictive check method. This method is similar to the posterior predictive check, but assumes that parameter uncertainty is negligible relative to interindividual and residual variance.
  • Five hundred Monte Carlo simulation replicates of the original dataset were compared with the distribution of the same exposure metric in the observed vedolizumab dataset, using quantile-quantile plots.
  • the population pharmacokinetic-pharmacodynamic analysis for repeated measures was conducted using the nonlinear mixed effects modeling approach (NONMEM, Version 7.2).
  • the pharmacokinetic-pharmacodynamic data were modeled using a sequential approach, where individual predicted vedolizumab serum concentrations from the population pharmacokinetic model were used to drive the pharmacodynamic response.
  • the pharmacodynamic evaluations were based on percentage of MAdCAM-1 binding by lymphocytes expressing high levels of ⁇ 4 ⁇ 7 integrin (CD4 + CD45RO high ).
  • a direct effect sigmoid E max model was selected to characterize the exposure-response relationship for the effect of vedolizumab on MAdCAM-1 binding to ⁇ 4 ⁇ 7 . No formal covariate modeling or model evaluation was conducted.
  • the study population consisted of 2554 individuals who contributed 18427 evaluable vedolizumab serum samples, including 87 healthy volunteers from the phase 1 study, 46 patients from the phase 2 study (UC), and 891, 1115, and 415 patients from the phase 3 GEMINI 1 (UC), GEMINI 2 (CD), and GEMINI 3 (CD) studies, respectively.
  • Demographics and other characteristics of the pharmacokinetic analysis population are summarized in Table 1.
  • the analysis population consisted of 1290 men and 1264 women with ages ranging from 18 to 78 years and baseline body weights ranging from 28 to 170 kg. A total of 1530 individuals had CD and 937 had UC; 87 were healthy volunteers.
  • the median (interquartile range) vedolizumab serum trough concentration-time profiles for patients with UC from GEMINI 1 and patients with CD from GEMINI 2 are shown in FIG. 1 .
  • Vedolizumab pharmacokinetics was described by a 2-compartment model with parallel linear and nonlinear elimination.
  • a 2-compartment model resulted in a significant improvement in goodness-of-fit criteria over a 1-compartment model, as did a parallel linear and nonlinear elimination model over a linear model.
  • the population pharmacokinetic model of vedolizumab is represented diagrammatically in FIG. 2 .
  • Body weight was chosen to represent changes in vedolizumab pharmacokinetics as a function of body size and was described using an allometric model with a reference weight of 70 kg.
  • the other continuous covariates of albumin, fecal calprotectin, partial Mayo score, age, and CDAI score entered the model as power functions normalized by a reference value (typically near the observed median value of the data).
  • the categorical covariates of prior TNF- ⁇ antagonist therapy status, ADA status, concomitant therapy use, and IBD diagnosis entered the model as power functions, with a separate dichotomous (0, 1) covariate serving as an on-off switch for each effect.
  • Time-dependent covariates were body weight, albumin, fecal calprotectin, and concomitant therapy use.
  • the effect of IBD diagnosis on linear clearance (CL L ) was investigated by modeling separate CL L parameters for patients with UC and those with CD, while the effect of IBD diagnosis on central compartment volume of distribution (V c ) was evaluated by including IBD as a predictor of V c in the covariate model.
  • the typical values of CL L were 0.159 L/day for patients with UC and 0.155 L/day for patients with CD.
  • the individual estimates of CL L for patients with UC and CD were distributed over a wide range as represented in FIG. 3 .
  • the proportional residual variance estimate (unexplained random residual variability in the model) was relatively small.
  • the estimates of interindividual variances and correlations are presented in Table S2, and the standard deviations and shrinkage estimates of interindividual random effects are presented in Table S3.
  • FIGS. 4 and 9 Goodness-of-fit plots from the final population pharmacokinetic model are presented in FIGS. 4 and 9 . Diagnostic plots indicated that the full covariate pharmacokinetic model was consistent with the observed data and no systematic bias was evident.
  • the relative changes in CL L for a reference individual with various covariate values are illustrated in FIG. 5 .
  • the point estimates and 95% CDIs for the effects of covariates on CL L are presented in Table S4.
  • a patient of 120 kg with a serum albumin concentration of 4.0 g/dL had a 19% probability of having clearance greater than the pre-specified criterion for clinical significance.
  • the effects of fecal calprotectin, CDAI score, partial Mayo score, age, prior TNF- ⁇ antagonist therapy status, ADA status, and concomitant therapy use on vedolizumab CL L were not considered clinically relevant since the covariate effect sizes were less than ⁇ 25% from the typical reference values (Table S4).
  • the 95% CDIs for these covariate effects were generally narrow and contained the null effect value.
  • the final population pharmacokinetic model and parameter estimates were evaluated with a predictive check method and Bayesian 95% CDIs derived from the posterior probability distributions.
  • the basic premise of a predictive check is that a model and parameters derived from an observed dataset should produce simulated data that are similar to the original observed data.
  • the predictive check plots demonstrated overall good agreement between the observed and simulated data ( FIGS. 10, 11, and 12 ).
  • the precision of the parameters estimates was assessed by evaluating the Bayesian 95% CDIs (Tables 2, S2, and S4). Overall, the structural pharmacokinetic model parameters, covariates effects, and variance parameters were estimated with good precision.
  • the vedolizumab population pharmacokinetic-pharmacodynamic dataset was composed of 593 individuals contributing a total of 2442 evaluable MAdCAM-1 observations.
  • the analysis population consisted of 297 patients with UC and 296 patients with CD (from the phase 2 study and GEMINI 1 and 2).
  • MAdCAM - 1 E 0 * ( 1 - E max * Conc ⁇ EC 50 + Conc ⁇ )
  • Eo is the baseline MAdCAM-1 percent binding
  • Emax is the maximum effect
  • Conc is the vedolizumab serum concentration
  • EC50 is the vedolizumab serum concentration at half-maximum effect
  • is the Hill-coefficient or slope factor.
  • the model was parameterized in terms of baseline MAdCAM-1 inhibition (E 0 ), maximum effect (E max ), vedolizumab serum concentration at half-maximum effect (EC 50 ), and Hill-coefficient or slope factor ( ⁇ ).
  • E 0 baseline MAdCAM-1 inhibition
  • E max maximum effect
  • EC 50 vedolizumab serum concentration at half-maximum effect
  • Hill-coefficient or slope factor
  • the base model provided a reasonable description of the data as judged by visual inspection of diagnostic plots ( FIG. 13 ) but some deficiencies in the model were noted. Variance parameter estimates were indicative of moderate to large unexplained interindividual and residual variability, with estimates of 41.8% CV for E 0 , 0.551 (SD logistic distribution) for E max , and 78.3% CV for the exponential residual error variance ( ⁇ 2 exp ) (Table 3).
  • the nonlinear pathway is thought to be due to clearance by saturable, target-mediated mechanisms such as receptor-mediated endocytosis.
  • the linear pathway represents components that are non-saturable at therapeutic concentrations, such as Fc-mediated elimination.
  • Parallel elimination is typical of monoclonal antibodies with disposition that is affected by binding to the target, in this case the ⁇ 4 ⁇ 7 integrin on circulating T lymphocytes.
  • TNF- ⁇ antagonists are best described by a linear elimination process.
  • the elimination of infliximab in patients with UC or ankylosing spondylitis and of golimumab in patients with rheumatoid arthritis follows a 2-compartment model with linear elimination.
  • TNF- ⁇ exists in both soluble and membrane-bound forms and is present in abnormally high concentrations in serum and gut mucosa in patients with IBD.
  • the localization of TNF- ⁇ in inflammatory tissues may make it difficult to rapidly achieve target saturation because of slow redistribution of the drug from plasma to the target sites.
  • TNF- ⁇ concentrations in different compartments results in drug redistribution, with possible effects on pharmacokinetic and pharmacokinetic-efficacy/safety relationships of TNF- ⁇ antagonists. The clinical implications of these differences in elimination pathways are not currently understood.
  • vedolizumab CL L for a 40-year-old, 70-kg patient with a serum albumin concentration of 4 g/dL was 0.159 L/day for a patient with UC and 0.155 L/day for a patient with CD.
  • FcRn Fc receptor
  • the second important covariate identified was body weight, which was positively correlated with the clearance of vedolizumab.
  • a patient of 120 kg with a serum albumin concentration of 4.0 g/dL had a 19% probability of having clearance greater than the pre-specified criterion for clinical significance.
  • Measures of body size are the most commonly identified covariates influencing the pharmacokinetics of therapeutic monoclonal antibodies.
  • the impact of body weight on vedolizumab pharmacokinetics is consistent with that reported in population analyses of other therapeutic monoclonal antibodies.
  • ADAs have been reported to increase infliximab clearance.
  • the presence of ADAs was estimated to increase vedolizumab clearance by only 12%.
  • the small impact may be a result of the low incidence of ADAs observed in the GEMINI trials.
  • vedolizumab trough concentrations were below the limit of quantification. We believe that sensitization of patients to monoclonal antibodies is an important cause of treatment failure and that vedolizumab is not unique in this regard.
  • thiopurines and methotrexate on the pharmacokinetics of vedolizumab differs from TNF- ⁇ antagonists in both IBD and rheumatoid arthritis patients where co-administration of these agents is associated with higher trough concentrations and lower drug clearance.
  • modulation of Fc ⁇ expression is one possible explanation.
  • methotrexate is known to down-regulate Fc ⁇ receptors on monocytes and other Fc-receptor subtypes.
  • prevention of the ADA development increases drug exposure by reducing immune-mediated drug clearance.
  • ⁇ 4 ⁇ 7 receptor saturation was maintained at vedolizumab concentrations considered subtherapeutic, raising the question as to whether receptor saturation is necessary but not sufficient for clinical efficacy.
  • the EC 50 estimate from population pharmacokinetic-pharmacodynamic model was 0.093 ⁇ g/mL, suggesting that full saturation is reached at vedolizumab serum concentration of approximately 1 ⁇ g/mL.
  • the exposure-efficacy results indicated that vedolizumab concentrations below 17 and 15 mg/mL at induction were associated with efficacy similar to placebo in patients with UC and CD, respectively.
  • MAdCAM-1 assay measures ⁇ 4 ⁇ 7 saturation in circulating T-cells or may be due to a slow onset of action of the drug.
  • the MAdCAM-1 assay is insensitive to dose and should not be used for dose selection. Further studies to evaluate the relative pharmacodynamic contributions of ⁇ 4 ⁇ 7 receptor blockade in peripheral blood in distinction to effects on cell-cell interactions in the tissue compartment are a research priority.
  • the aim is to determine if induction treatment with VDZ reduces fecal calprotectin levels in patients with moderately to severely active UC.
  • the Percent of Baseline Calprotectin at Week 6 by Treatment or Response Status is shown in FIG. 14 .
  • mean fCAL levels were 2370 and 2552 ⁇ g/g in PBO and VDZ-treated patients, respectively.
  • VDZ induction therapy for UC reduced fCal levels at week 6, a marker of mucosal inflammation, significantly more than PBO. Response and remission rates were higher with VDZ regardless of baseline fCal concentration.
  • Deep remission a combination of endoscopic and patient-reported outcomes, is an emerging treatment goal for patients with ulcerative colitis (UC).
  • UC ulcerative colitis
  • Statistically significant and clinically meaningful improvement in deep remission endpoints at weeks 6 and 52 were previously reported in the GEMINI 1 population using varying combinations of Mayo subscores of endoscopic and patient-reported outcomes.
  • HRQoL health-related quality of life
  • VDZ-treated patients in deep remission had improved IBDQ and EQ-5D VAS. Even among patients not in deep remission at week 6, maintenance treatment with VDZ resulted in more patients with improved clinical and HRQoL outcomes than those re-randomised to PBO.

Abstract

The invention provides methods for treating patients with anti-α4β7 antibody comprising predicting outcome of the antibody therapy. The invention relates to the identification of patients who can respond to therapy comprising an anti-a4β7 antibody.

Description

    RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application 62/096,636 filed on Dec. 24, 2014, U.S. Provisional Application 62/117,750 filed on Feb. 18, 2015 and U.S. Provisional Application 62/132,917 filed on Mar. 13, 2015. The entire contents of the foregoing applications are incorporated herein by reference.
  • BACKGROUND
  • Inflammatory bowel disease (IBD), such as ulcerative colitis and Crohn's disease, for example, can be a debilitating and progressive disease involving inflammation of the gastrointestinal tract. IBD treatments have included anti-inflammatory drugs (such as, corticosteroids and sulfasalazine), immunosuppressive drugs (such as, 6-mercaptopurine, cyclosporine and azathioprine) and surgery (such as, colectomy). Podolsky, New Engl. J. Med., 325:928-937 (1991) and Podolsky, New Engl. J. Med., 325:1008-1016 (1991). As the disease progresses, treatment progresses into regimens that expose patients to progressive risk of side effects and loss of quality of life.
  • Integrin receptors are important for regulating both lymphocyte recirculation and recruitment to sites of inflammation (Carlos, T. M. and Harlan, J. M., Blood, 84:2068-2101 (1994)). The human α4β7 integrin has several ligands, one of which is the mucosal vascular addressin MAdCAM-1 (Berlin, C., et al., Cell 74: 185-195 (1993); Erle, D. J., et al., J. Immunol. 153:517-528 (1994)), which is expressed on high endothelial venules in mesenteric lymph nodes and Peyer's patches (Streeter, P. R., et al., Nature 331:41-46 (1998)). As such, the α4β7 integrin acts as a homing receptor that mediates lymphocyte migration to intestinal mucosal lymphoid tissue (Schweighoffer, T., et al., J. Immunol. 151: 717-729 (1993)).
  • Antibodies against human α4β7 integrin, such as murine monoclonal antibody Act-1 (mAb Act-1), interfere with α4β7 integrin binding to mucosal addressin cell adhesion molecule-1 (MAdCAM-1) present on high endothelial venules in mucosal lymph nodes. Act-1 was originally isolated by Lazarovits, A. I., et al., J. Immunol. 133:1857-1862 (1984), from mice immunized with human tetanus toxoid-specific T lymphocytes and was reported to be a mouse IgG1/κ antibody. Subsequent analysis of the antibody by Schweighoffer, T., et al., J. Immunol. 151:717-729 (1993) demonstrated that it can bind to a subset of human memory CD4+T lymphocytes which selectively express the α4β7 integrin. Entyvio™ vedolizumab, an anti-α4β7 integrin monoclonal antibody (mAb) with structural features derived from Act-1, is indicated for treating ulcerative colitis (UC) and Crohn's disease (CD). Studies reporting the activity of vedolizumab in treating these disorders (Feagen et al. NEJM 369:699-710 (2013) and Sandborn et al. NEJM369:711-721 (2013)) showed varying levels of success depending on the disorder and nature of prior therapies. As these were lengthy studies and there are a growing number of treatment options available to patients, there is a need to identify patients who can benefit from vedolizumab therapy early in their treatment. Expedient and accurate treatment decisions lead to effective management of the disease.
  • SUMMARY OF THE INVENTION
  • The invention relates to the identification of patients who can respond to therapy comprising an anti-α4β7 antibody, such as vedolizumab. Early in the course of treatment, e.g., with vedolizumab, factors measured from the patient or from biological samples of the patient indicate whether a patient is likely to respond to treatment.
  • In one aspect, pharmacokinetics or pharmacodynamics factors can indicate whether a patient will respond to treatment with an anti-α4β7 antibody, such as vedolizumab. As higher therapeutic mAb trough concentrations have been associated with greater efficacy, understanding determinants of mAb clearance may optimize dosing regimens. Applicants have identified the problem, and characterized anti-α4β7 antibody, such as vedolizumab, pharmacokinetic factors in patients with UC and CD, identified clinically relevant determinants of anti-α4β7 antibody, such as antibody clearance, and described the pharmacokinetic-pharmacodynamic relationship using population modeling.
  • In some embodiments, a pharmacokinetics factor is mean serum trough concentration. In other embodiments, a pharmacokinetics factor is therapeutic antibody clearance. In an embodiment, a pharmacodynamic factor is the amount of antibody bound to α4β7 integrin. In another embodiment, a pharmacodynamic factor is the amount of unbound α4β7 integrin.
  • In one embodiment, a method for treating a patient having inflammatory bowel disease (IBD) with an anti-α4β7 antibody, such as vedolizumab, comprises administering two doses of the anti-α4β7 antibody to a patient suffering from IBD, wherein the second dose is administered about two weeks after the first dose is administered to the patient; waiting a period of time about four weeks; measuring the patient's serum concentration of the anti-α4β7 antibody; and administering one or more further doses of the anti-α4β7 antibody to the patient if the patient's serum concentration, e.g., a trough serum concentration, of the antibody is at least 8 μg per ml. In some embodiments, the waiting period is two weeks or about two to five weeks. In an embodiment, the method comprises administering two doses of vedolizumab to a patient suffering from IBD, wherein the second dose is administered about two weeks after the first dose is administered to the patient; waiting a period of time about four weeks; measuring the patient's serum concentration of vedolizumab; and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration, e.g., a trough serum concentration, of the antibody is at least 8 μg per ml. In some embodiments, the patient's serum concentration of the anti-α4β7 antibody, e.g., vedolizumab, may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 μg per ml. In an embodiment, a patient who is likely to respond to the anti-α4β7 antibody, e.g., vedolizumab has a serum concentration, e.g., a trough serum concentration of greater than 17 μg/ml, greater than 25 μg/ml, or greater than 35 mg/mi. In another embodiment, the patient's serum concentration of anti-α4β7 antibody, e.g., vedolizumab, may be selected from the group consisting of 12-25 μs per ml, 15-17 μg per ml, 17-25 μg per ml, 12-40 μg per ml, and 17-40 μg ml.
  • In another embodiment, a method for treating a patient having inflammatory bowel disease (IBD) with an anti-α4β7 antibody, such as vedolizumab comprises the steps of administering at least one dose of vedolizumab to the patient suffering from IBD; waiting a period of at least two weeks, measuring the patient's serum concentration of vedolizumab; and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration is at least 8 μg per ml. In some embodiments, the waiting period is about two to five weeks.
  • In another aspect, clinical measures during early treatment can indicate whether a patient will respond to treatment with an anti-α4β7 antibody, such as vedolizumab. In one embodiment, the patient can have mucosal healing. In another embodiment, the patient can have deep remission. In an embodiment, the method for treating a patient can comprise measuring an endoscopic subscore, e.g., from the Mayo score, wherein the anti-α4β7 antibody, e.g., vedolizumab is continued with an endoscopic score of less than 3. The endoscopic score can be less than 2.5, less than 2, between 0 and 2, 1, or equal to or less than 1 (≦1) or less than 1 ((<1). In another embodiment, the method for treating a patient can comprise measuring the Crohn's Disease Activity Index (CDAI). In another embodiment, the method for treating a patient can comprise measuring the number, size or severity of fistulae. In an embodiment, the endoscopic score is a simple endoscopic score for Crohn's Disease (SES-CD). In some embodiments, the SES-CD can be in the range of 0 to 6, ≦6, ≦5 or no more than 4 (≦4). In some embodiments, the SES-CD is decreased by 25%, 35%, 40%, 45%, 50%, 55%, 60% or 65% from baseline. In one embodiment, the SES-CD is decreased 50% from baseline. In some embodiments, the method for treating a patient can comprise measuring the amount of ulceration in the digestive tract. In some embodiments, the method for treating a patient can comprise measuring mucosal healing. In some embodiments of mucosal healing, ulceration is decreased or absent.
  • In another aspect, measures of inflammation can indicate whether a patient will respond to treatment with an anti-α4β7 antibody, such as vedolizumab. Measures of inflammation include amounts of fecal calprotectin, amounts of C-reactive protein (CRP) and amount of albumin. In some embodiments, the method comprises measuring the fecal calprotectin concentration. Treatment with the antibody, e.g., vedolizumab can be continued with a fecal calprotectin concentration of less than 1500 μg/g. The fecal calprotectin concentration can be less than 1250 μg/g, less than 1000 μg/g, less than 750 μg/g, less than 500 μg/g, less than 400 μg/g, less than 300 μg/g, less than 250 μg/g, between 200-1200 μg/g, between 350 to 800 μg/g, between 300-1000 μg/g, <50 μg/g, <100 μg/g, <150 μg/g, <200 μg/g, ≦250-499 μg/g, or between 500 to 900 μg/g.
  • In some embodiments, treatment with an anti-α4β7 antibody, e.g., vedolizumab can be continued if the fecal calprotectin is reduced to less than 50% of the baseline or concentration before treatment. The fecal calprotectin can be reduced to less than 45%, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, between 10-55%, between 10-30%, between 15-35%, between 15-45%, or between 20-40% of the baseline or concentration before treatment.
  • In some embodiments, methods described herein comprise measuring albumin concentration. An albumin concentration greater than 3.2 g/dL further identifies the patient for continued treatment with an anti-α4β7 antibody, e.g., vedolizumab. The albumin concentration can be greater than 3.5 g/dL, greater than 4.0 g/dL, or greater than 4.7 g/dL, in the range of 3.3 to 5.0 g/dL, in the range of 3.5 to 5.0 g/dL, in the range of 3.8 to 5.0 g/dL or in the range of 4.0 to 5.0 g/dL.
  • In one aspect, the invention relates to therapeutic methods which further include the step of continuing, stopping, discontinuing or halting a therapy accordingly where factors measured as described herein indicate whether there is continued benefit of the therapy.
  • In one embodiment, a method of identifying a patient for continued treatment with an anti-α4β7 antibody comprises the steps of: measuring the concentration of the anti-α4β7 antibody in a sample of serum obtained from a patient suffering from inflammatory bowel disease (IBD) and who received at least one dose of an anti-α4β7 antibody within the previous two months; and identifying the patient for continued treatment with the anti-α4β7 antibody if the serum concentration in the sample is at least 8 μg per ml. The patient may have received at least one dose of the anti-α4β7 antibody within the previous month. In another embodiment, the patient received at least two doses of the anti-α4β7 antibody within the previous four months; and the method identifies the patient for continued treatment with the anti-α4β7 antibody if its serum concentration in the sample is at least 8 μg per ml four weeks after the second dose, e.g., four weeks after the dose at week 2. The patient may have received at least two doses of the anti-α4β7 antibody within the previous three months or the previous two months. In some embodiments, the patient's serum concentration of the anti-α4β7 antibody may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 μg per ml. In another embodiment, the patient's serum concentration of the anti-α4β7 antibody may be selected from the group consisting of 12-25 μg per ml, 15-17 μg per ml, 17-25 μg per ml, 12-40 μg per ml, and 17-40 μg ml.
  • In one embodiment, a method of identifying a patient for continued treatment with vedolizumab comprises the steps of: measuring the concentration of vedolizumab in a sample of serum obtained from a patient suffering from inflammatory bowel disease (IBD) and who received at least two doses of vedolizumab within the previous four months; and identifying the patient for continued treatment with vedolizumab if the serum concentration in the sample is at least 8 μg per ml four weeks after the second dose, e.g., four weeks after the dose at week 2. The patient may have received at least two doses of vedolizumab within the previous three months or the previous two months. In some embodiments, the patient's serum concentration of vedolizumab may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 μg per ml. In another embodiment, the patient's serum concentration of vedolizumab may be selected from the group consisting of 12-25 μg per ml, 15-17 μg per ml, 17-25 μg per ml, 12-40 μg per ml, and 17-40 μg ml.
  • In another embodiment, the invention relates to a method of identifying a patient for continued treatment with vedolizumab, the method comprising the steps of: measuring the concentration of vedolizumab in a sample of serum obtained from a patient suffering from inflammatory bowel disease (IBD) and who received at least one dose of vedolizumab within the previous two months; and identifying the patient for continued treatment with vedolizumab if the serum concentration in the sample is at least 8 μg per ml. The patient may have received at least one dose of vedolizumab within the previous month. In some embodiments, the patient's serum concentration of vedolizumab may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 μg per ml. In another embodiment, the patient's serum concentration of vedolizumab may be selected from the group consisting of 12-25 μg per ml, 15-17 μg per ml, 17-25 μg per ml, 12-40 μg per ml, and 17-40 μg ml.
  • In another aspect, continuing treatment includes maintaining remission of IBD.
  • In one embodiment, the invention relates to a method for maintaining remission of inflammatory bowel disease (IBD) in a patient, wherein the patient had received at least one dose of an anti-α4β7 antibody in the previous two months, the method comprising: obtaining a serum sample from the patient; measuring the concentration of the anti-α4β7 antibody in the sample; administering the anti-α4β7 antibody thereafter every eight weeks if the concentration is at least 8 μg per ml. In some embodiments, the patient's serum concentration of the anti-α4β7 antibody may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 μg per ml. In another embodiment, the patient's serum concentration of the anti-α4β7 antibody may be selected from the group consisting of 12-25 μg per ml, 15-17 μg per ml, 17-25 μg per ml, 12-40 μs per ml, and 17-40 μs ml.
  • In one embodiment, the invention relates to a method for maintaining remission of inflammatory bowel disease (IBD) in a patient, wherein the patient had received at least one dose of vedolizumab in the previous two months, the method comprising:
  • obtaining a serum sample from the patient; measuring the concentration of vedolizumab in the sample; administering vedolizumab thereafter every eight weeks if the concentration is at least 8 μg per ml. In some embodiments, the patient's serum concentration of vedolizumab may be at least 10, 12, 14, 17, 20, 25, 30, 35, or 40 μg per ml. In another embodiment, the patient's serum concentration of vedolizumab may be selected from the group consisting of 12-25 μg per ml, 15-17 μg per ml, 17-25 μg per ml, 12-40 μg per ml, and 17-40 μg ml.
  • In one aspect, evaluation of a patient early in treatment with an anti-α4β7 antibody, such as vedolizumab, indicates that the patient is in deep remission. In an embodiment, the invention relates to a method for treating a patient having inflammatory bowel disease (IBD) with vedolizumab, comprising: providing two doses of vedolizumab, wherein the second dose is about two weeks after the first; waiting about four weeks; and continuing to treat the patient with vedolizumab if the patient is in deep remission, wherein determining deep remission comprises measuring an endoscopic subscore for the patient. The patient can be determined to be in deep remission and continue to receive the anti-α4β7 antibody, e.g., vedolizumab, if the endoscopic subscore is 0 to 1. The method can further comprise determining a patient-reported outcome score, wherein a determination of deep remission comprises 0 to 1 for both the endoscopic subscore and the patient reported outcome score. The patient reported outcome score can comprise a rectal bleeding subscore. The patient reported outcome score can comprise a stool frequency subscore. The method can further comprise measuring the fecal calprotectin concentration. Treatment with an anti-α4β7 antibody, e.g., vedolizumab, can be continued with a fecal calprotectin concentration of less than 1500 μg/g. The fecal calprotectin concentration can be less than 1250 μg/g, less than 1000 μg/g, less than 750 μg/g, less than 500 μg/g, less than 400 μg/g, less than 300 μg/g, less than 250 μg/g, between 200-1200 μg/g, between 350 to 800 μg/g, between 300-1000 μg/g, <50 μg/g, <100 μg/g, <150 μg/g, <200 μg/g, ≦250-499 μg/g, or between 500 to 900 μg/g.
  • In some embodiments, treatment with the anti-α4β7 antibody, e.g., vedolizumab, can be continued if the fecal calprotectin is reduced to less than 50% of the baseline or concentration before treatment. The fecal calprotectin can be reduced to less than 45%, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, between 10-55%, between 10-30%, between 15-35%, between 15-45%, or between 20-40% of the baseline or concentration before treatment.
  • In an embodiment, a vedolizumab-treated patient is in deep remission and can continue to be administered vedolizumab if the vedolizumab trough serum concentration is at least 25 μg/ml, at least 27 μg/ml, at least 30 μg/ml, at least 32 μg/ml or at least 34 μg/ml.
  • In one aspect, the invention relates to a method for identifying a patient for continued treatment with an anti-α4β7 antibody, the method comprising the steps of: measuring the clearance of the anti-α4β7 antibody in a biological sample obtained from a patient suffering from inflammatory bowel disease (IBD) and who was administered at least two doses of the anti-α4β7 antibody within the previous four months; and identifying the patient for continued treatment with the anti-α4β7 antibody if the clearance in the patient is less than 0.25 L/day. In some embodiments, the patient was administered at least one dose of the anti-α4β7 antibody within the previous two months, the patient was administered at least two doses of the anti-α4β7 antibody within the previous three months or the previous two months, or, the patient was administered at least one dose of the anti-α4β7 antibody within the previous two months or within the previous month. The clearance may be less than 0.20 L/day or between 0.1 and 0.2 L/day.
  • In one aspect, the invention relates to a method for identifying a patient for continued treatment with vedolizumab, the method comprising the steps of: measuring the clearance of vedolizumab in a biological sample obtained from a patient suffering from inflammatory bowel disease (IBD) and who was administered at least two doses of vedolizumab within the previous four months; and identifying the patient for continued treatment with vedolizumab if the clearance in the patient is less than 0.25 L/day. The clearance may be less than 0.20 L/day or between 0.1 and 0.2 L/day.
  • In some embodiments, the patient was administered at least two doses of vedolizumab within the previous three months or the previous two months.
  • In another aspect, the invention relates to a method for identifying a patient for continued treatment with vedolizumab, the method comprising the steps of: measuring the clearance of vedolizumab in a biological sample obtained from a patient suffering from inflammatory bowel disease (IBD) and who was administered at least one dose of vedolizumab within the previous two months; and identifying the patient for continued treatment with vedolizumab if the clearance in the patient is less than 0.25 L/day. The clearance may be less than 0.20 L/day or between 0.1 and 0.2 L/day. In some embodiments, the patient was administered at least one dose of vedolizumab within the previous month. In some embodiments, the patient was administered at least two doses of vedolizumab within the previous month.
  • The invention further relates to assays for use in measuring the factors described herein for identifying a patient who will respond to treatment with an anti-α4β7 antibody, such as vedolizumab. In some embodiments the assay is a pharmacokinetic assay for circulating anti-α4β7 antibody. In an embodiment, the assay may measure high or sustained positive levels of anti-α4β7 antibody, such as at least 8, 10, 12, 14, 17, 20, 25, 30, 35, or 40 μg per ml in a serum sample from a patient, e.g., for predicting the ability to respond or maintain a response or remission of the IBD afflicting the patient. In an embodiment, the serum concentration of the anti-α4β7 antibody may be measured by a sandwich ELISA assay. In an embodiment, the serum concentration of the anti-α4β7 antibody may be measured in an antibody bridging assay.
  • In some embodiments, the assay for use in measuring the factors described herein for identifying a patient who will respond to treatment with an anti-α4β7 antibody, such as vedolizumab, is a pharmacodynamic assay for α4β7 integrin, e.g., on circulating lymphocytes. In an embodiment, low or minimum levels of free α4β7 integrin, such as less than 10%, less than 7%, less than 5% or less than 3% free α4β7 integrin, may predict the effectiveness of the anti-α4β7 antibody. In an embodiment, the free α4β7 integrin may be measured by the amount of MAdCAM binding to the α4β7 integrin. In another embodiment, the free α4β7 integrin may be measured by the amount of anti-α4β7 antibody binding to the α4β7 integrin.
  • In some embodiments, for use in measuring the factors described herein for identifying a patient who will respond to treatment with an anti-α4β7 antibody, such as vedolizumab, the assay measures immune response to the anti-α4β7 antibody. In an embodiment, a low or absent immune response to anti-α4β7 antibody may predict the ability to respond or maintain a response or remission of the IBD afflicting the patient.
  • The method may further comprise measuring the fecal calprotectin concentration. Treatment with the anti-α4β7 antibody, such as vedolizumab may be continued with a fecal calprotectin concentration of less than 1500 μg/g. The fecal calprotectin concentration may be less than 1250 μg/g, less than 1000 μg/g, less than 750 μg/g, less than 500 μg/g, less than 400 μg/g, less than 300 μg/g, less than 250 μg/g, between 200-1200 μg/g, between 350 to 800 μg/g between 300-1000 μg/g, <50 μg/g, <100 μg/g, <150 μg/g, <200 μg/g, ≦250-499 μg/g, or between 500 to 900 μg/g, in a sample from a patient who is likely to respond to the anti-α4β7 antibody, such as vedolizumab.
  • In some embodiments, treatment with the anti-α4β7 antibody, such as vedolizumab can be continued if the fecal calprotectin is reduced to less than 50% of the baseline or concentration before treatment. The fecal calprotectin can be reduced to less than 45%, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, between 10-55%, between 10-30%, between 15-35%, between 15-45%, or between 20-40% of the baseline or concentration before treatment.
  • In one aspect, a method for treating a human patient having inflammatory bowel disease (IBD), the method comprises: selecting a human patient having IBD and having a serum concentration of vedolizumab which is at least 8 μg per ml at a time point that is four weeks after a second dose of vedolizumab, wherein a first dose of vedolizumab was administered to the subject two weeks prior to the second dose of vedolizumab; and administering vedolizumab to the human patient having IBD, thereby treating the human patient having IBD. In one embodiment, the first dose of vedolizumab comprises 300 mg. In one embodiment, the second dose of vedolizumab comprises 300 mg. In one embodiment, the patient received the first and the second dose intravenously. In some embodiments, the patient had an inadequate response with, lost response to, or was intolerant to a TNF blocker.
  • One embodiment provided herein is an in vitro method for determining the responsiveness of a human patient having Inflammatory Bowel Disease (IBD) to treatment with vedolizumab, the method comprising measuring the concentration of vedolizumab in a blood sample from the patient by contacting the blood sample with an anti-vedolizumab antibody, wherein the sample is obtained about four weeks following administration of a second dose of vedolizumab, wherein a first dose of vedolizumab was administered to the subject two weeks prior to the second dose of vedolizumab, and wherein a vedolizumab concentration of at least 8 μg per ml in the blood sample indicates that the patient is responsive to treatment with vedolizumab, and a concentration of less than 8 μg per ml in the blood sample indicates that the patient is not responsive to treatment with vedolizumab. In one embodiment, the method further comprises administering vedolizumab to the patient. In one embodiment, the first dose of vedolizumab comprises 300 mg. In one embodiment, the second dose of vedolizumab comprises 300 mg. In one embodiment, the patient received the first and the second dose intravenously. In some embodiments, the patient had an inadequate response with, lost response to, or was intolerant to a TNF blocker.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the median (interquartile range) of observed vedolizumab trough serum concentration versus nominal sampling time in patients with UC (GEMINI 1) and patients with CD (GEMINI 2) during maintenance treatment with vedolizumab 300 mg Q4W or Q8W or placebo. All patients (including those in the placebo group) received 2 doses of vedolizumab 300 mg during induction (at weeks 0 and 2).
  • FIG. 2 is a diagramatic representation of the population pharmacokinetic model of vedolizumab. Conc, concentration; Km, Michaelis-Menton constant; Vmax, maximum rate.
  • FIG. 3 shows distribution of individual vedolizumab linear clearance (CLL) estimates from the final population pharmacokinetic model in patients with UC and patients with CD.
  • FIG. 4 is a goodness-of-fit plot: observed vedolizumab concentration versus predicted vedolizumab concentration from the final population pharmacokinetic model. Values are indicated by closed circles with a dashed black loss trend line through the data. The line of identity (solid white) is included as reference.
  • FIG. 5 shows the effect of covariates on vedolizumab linear clearance (CLL). Each point and line represents the median and 95% credible interval (CDI), respectively, of the Bayesian posterior distribution of normalized samples of vedolizumab CLL adjusted for the covariate. The reference individual weighs 70 kg, is 40 years old, has an albumin level of 4 g/dL, fecal calprotectin level of 700 mg/kg, CDAI score of 300 (for patient with CD), partial Mayo score of 6 (for patient with UC), ADA negative, and no concomitant therapy use, and is TNF-α antagonist therapy naïve. Albumin: 2.7, 3.2, 3.7, 4.0, 4.2, and 4.7 g/dL represent the 6th, 18th, 50th, 70th, 85th, and 98.5th percentiles, respectively, of baseline albumin levels for patients in GEMINI 1, 2, and 3. Weight: 40, 60, 80, 100, and 120 kg represent the 1.5th, 30th, 71st, 92nd, and 98th percentiles, respectively, of baseline weight values for patients in GEMINI 1, 2, and 3. The vertical black line is drawn at the reference point estimate, and the shaded region is ±25% of the reference point estimate chosen to represent an uncertainty range of clinical unimportance.
  • FIG. 6 shows individual vedolizumab linear clearance (CLL) estimates at the end of induction (week 6) by Mayo clinic endoscopic subscore for patients with UC (GEMINI 1). Midlines represent medians. Box limits represent 25th and 75th percentiles. Whiskers (error bars) represent highest and lowest points within 1.5×interquartile range. Individual points represent outliers.
  • FIG. 7 is a receptor (α4β7) saturation plot: observed MAdCAM-1 versus observed vedolizumab concentration in patients with UC (GEMINI 1) and patients with CD (GEMINI 2). Baseline data are excluded.
  • FIG. 8 shows observed MAdCAM-1 versus time for patients with UC (GEMINI 1) and patients with CD (GEMINI 2).
  • FIG. 9 is a goodness-of-fit plot for the vedolizumab final population pharmacokinetic model: CWRESI and residual versus time and population predicted vedolizumab concentration.
  • FIG. 10 shows a predictive check for the vedolizumab final population pharmacokinetic model: induction therapy.
  • FIG. 11 shows a predictive check for the vedolizumab final population pharmacokinetic model: maintenance therapy—every 4 weeks.
  • FIG. 12 is a predictive check for the vedolizumab final population pharmacokinetic model: maintenance therapy—every 8 weeks.
  • FIG. 13 is a goodness-of-fit model for MAdCAM-1.
  • FIG. 14 is Percent of Baseline Calprotectin at Week 6 by Treatment or Response Status.
  • FIGS. 15A-15D show clinical, and FIGS. 15E and 15F show HRQoL, outcomes at week 52 in patients in or not in deep remission (definition 1) at week 6.
  • FIGS. 16A-16D show clinical, and FIGS. 16 E and 16F show HRQoL, outcomes at week 52 in patients in or not in deep remission (definition 2) at week 6.
  • DETAILED DESCRIPTION
  • The invention relates to methods for treating with an anti-α4β7 antibody, e.g., vedolizumab, a patient having inflammatory bowel disease (IBD), methods for identifying a patient for continued treatment with the antibody, such as vedolizumab, and methods for maintaining remission of IBD in a patient.
  • Not all patients with inflammatory bowel diseases who receive treatment with an anti-α4β7 antibody, such as vedolizumab, respond to the treatment and some do not respond fully to treatment. Due to the morbidity of these diseases, there is a need to quickly identify those patients who respond to anti-α4β7 antibody therapy and those who do not respond. The responders can then continue on the therapy, and the non-responders can be started on alternative therapies. This application relates to the surprising discovery that a patient's serum concentration and/or clearance rate of the anti-α4β7 antibody early during a course of anti-α4β7 antibody treatment can be determined and used to identify or select patients who are responding to treatment and will continue to respond to additional doses of the anti-α4β7 antibody, and non-responders.
  • Vedolizumab, a humanized monoclonal antibody that binds specifically to the α4β7 integrin, is indicated for the treatment of patients with moderately to severely active ulcerative colitis (UC) and Crohn's disease (CD). Vedolizumab has a novel gut-selective mechanism of action that differs from that of other currently marketed biologic agents for the treatment for inflammatory bowel disease (IBD), including natalizumab and tumor necrosis factor-α (TNF-α) antagonists. By binding to cell surface—expressed α4β7, vedolizumab blocks the interaction of a subset of memory gut-homing T lymphocytes with mucosal addressin cell adhesion molecule-1 (MAdCAM-1) expressed on endothelial cells. Consequently, migration of these cells into inflamed intestinal tissue is inhibited.
  • The pharmacokinetics of other therapeutic monoclonal antibodies used for the treatment of UC and CD have been previously reported. Several factors are associated with accelerated clearance of these antibodies including the presence of anti-drug antibodies, sex, body size, concomitant immunosuppressant use, disease type, albumin concentration, and degree of systemic inflammation. Furthermore, a consistent relationship between efficacy and exposure, in distinction to drug dose, has been observed for many of these agents, such that higher trough drug concentrations are associated with greater efficacy. Differences in drug clearance may be an important explanation for this observation. Therefore, a better understanding of the determinants of clearance for therapeutic antibodies may result in optimization of drug regimens.
  • In previous studies, single-dose pharmacokinetics, pharmacodynamics (α4β7 receptor saturation), safety, and tolerability of vedolizumab were investigated over a dose range of 0.2 to 10 mg/kg in healthy volunteers (intravenous [IV] infusion) (unpublished data). After reaching peak concentrations, vedolizumab serum concentrations fell in a generally biexponential fashion until concentrations reached approximately 1 to 10 ng/mL. Thereafter, concentrations appeared to fall in a nonlinear fashion. The multiple-dose pharmacokinetics and pharmacodynamics of vedolizumab have been investigated following IV infusions of 0.5 and 2 mg/kg in patients with CD and infusion of 2, 6, and 10 mg/kg in patients with UC. Vedolizumab pharmacokinetics was generally linear following an IV infusion over the dose range of 2 to 10 mg/kg in patients with UC. After multiple-dose administration, rapid and near complete α4β7 receptor saturation was achieved following the first dose of vedolizumab.
  • The efficacy and safety of vedolizumab induction and maintenance therapy were demonstrated in patients with UC in the GEMINI 1 trial (ClinicalTrials.gov number, NCT00783718) and in patients with CD in the GEMINI 2 (ClinicalTrials.gov number, NCT00783692) and GEMINI 3 (ClinicalTrials.gov number, NCT01224171) trials. The exposure-response (efficacy) relationships of vedolizumab in patients with UC and CD for induction and maintenance therapy have been presented elsewhere.
  • Definitions
  • As used herein, the “trough” serum concentration of an antibody refers to the concentration just before the next dose.
  • “Clinical remission” or “remission” as used herein with reference to ulcerative colitis subjects, refers to a complete Mayo score of less than or equal to 2 points and no individual subscore greater than 1 point. Crohn's disease “clinical remission” refers to a Crohn's Disease Activity Index (CDAI) score of 150 points or less. The “Harvey-Bradshaw Index” (HBI) is a simpler version of the CDAI for data collection purposes. It consists of only clinical parameters including general well-being, abdominal pain, number of liquid stools per day, abdominal mass, hematocrit, body weight, medications to control diarrhea and presence of complications, and requires only a single day's worth of diary entries. Magnetic resonance enterography (MREn) is being evaluated as a method to measure remission.
  • “Endoscopic remission” as used herein, refers to a condition with a low endoscopic score. An example of a method to assess the endoscopic score in ulcerative colitis is flexible sigmoidoscopy. The endoscopic score in ulcerative colitis can be the Mayo subscore. An example of a method to assess the endoscopic score in Crohn's disease is ileocolonoscopy. The endoscopic score in Crohn's disease can be the simple endoscopic score for Crohn's Disease (SES-CD). The SES-CD can include measures such as the size of ulcers, the amount of ulcerated surface, the amount of affected surface and whether and to what extent the alimentary canal is narrowed.
  • A “clinical response” as used herein with reference to ulcerative colitis subjects refers to a reduction in complete Mayo score of 3 or greater points and 30% from baseline, (or a partial Mayo score of 2 or greater points and 25% or greater from baseline, if the complete Mayo score was not performed at the visit) with an accompanying decrease in rectal bleeding subscore of 1 or greater points (≧1) or absolute rectal bleeding score of 1 or less point (≦1). A “clinical response” as used herein with reference to Crohn's disease subjects refers to a 70 point or greater decrease in CDAI score from baseline (week 0). The terms “clinical response” and “response” e.g., alone without any adjective, are used interchangeably herein.
  • “Endoscopic response” as used herein, refers to a percentage decrease in an endoscopic score from baseline (e.g., at screening or just prior to initial dose). In Crohn's disease, endoscopic response can be assessed by a simple endoscopic score for Crohn's Disease (SES-CD).
  • “Baseline” as used herein describes a value of a parameter which is measured prior to the initial dose of a treatment. It can refer to a measurement of a sample obtained the same day, the day before, during the week before initial treatment, i.e., at a time period before the first dose when little change is expected until after the first dose and values of the measurement obtained after the first dose can be compared to this baseline value to represent the change caused by the dose.
  • “Mucosal healing” as used herein as used herein with reference to ulcerative colitis subjects, refers to a Mayo endoscopic subscore of less than or equal to 1. In reference to Crohn's disease, “fistula healing” results in closure or elimination of fistulae. In another reference to Crohn's disease, mucosal healing refers to an improvement in the amount or severity of wounding in mucosae, e.g., the digestive tract. For example, mucosal healing can refer to a decrease in the amount, size or severity of one or more than one ulcer in the digestive tract. In another example, mucosal healing refers to a decrease in one or more parameters selected from the group consisting of wall thickness, enhanced bowel wall contrast, mural edema, ulceration and perienteric vascularity. Such mucosal healing can be expressed as an SES-CD score, or a Magnetic Resonance Index of Activity (MaRIA) score. Complete mucosal healing in Crohn's disease includes absence of ulceration.
  • The “MaRIA score” is the sum of the scores, e.g., as measured by magnetic resonance enterography, of various mucosal healing parameters for each segment of colon and the terminal ileum (e.g., ileum, ascending colon, transverse colon, descending colon, sigmoid, and rectum).
  • “Corticosteroid (CS)-free remission” as used herein, refers to patients using oral corticosteroids at baseline who have discontinued corticosteroid use and are in clinical remission at week 52.
  • “European Quality of Life-5 Dimension (EQ-5D) visual analogue scale (VAS)” as used herein, refers to a questionnaire which is a validated (ahrq.gov/rice/eq5dproj.htm, “U.S. Valuation of the EuroQol EQ-5D™ Health States”, accessed 8 Aug. 2012, Bastida et al. BMC Gastroenterology 10:26-(2010), Konig et al. European Journal of Gastroenterology & Hepatology 14:1205-1215 (2002)) instrument used to measure general health-related quality of life (HRQOL) in patients and includes five domains—mobility, self-care, usual activities, pain/discomfort, and anxiety/depression. Patients choose the level of health problems they currently have on each item as “None”, “Moderate”, or “Extreme” and are scored a 1, 2, or 3, respectively. A composite EQ-5D score can be calculated from the individual scores to assess overall HRQOL. The EQ-5D Visual Analog Scale (VAS) score is a self-assigned rating of overall health using a 20 cm visual, vertical scale, with a score of 0 as the worst and 100 as best possible health. The EQ-5D and EQ-5D VAS have been shown in many studies to be valid and reliable instruments for measuring HRQOL in patients with GI diseases. A decrease of ≧0.3 points in the EQ-5D score represents a clinically meaningful improvement in HRQOL for patients. An increase of greater than or equal to 7 points in the EQ-5D VAS score represents a clinically meaningful improvement in HRQOL for patients.
  • The “Inflammatory Bowel Disease Questionnaire” ((IBDQ) questionnaire” (Irvine Journal of Pediatric Gastroenterology & Nutrition 28:S23-27 (1999)) is used to assess quality of life in adult patients with inflammatory bowel disease, ulcerative colitis, or Crohn's Disease and includes 32 questions on four areas of HRQOL: Bowel Systems (10 questions), Emotional Function (12 questions), Social Function (5 questions), and Systemic Function (5 questions). Patients are asked to recall symptoms and quality of life from the last 2 weeks and rate each item on a 7-point Likert scale (higher scores equate to higher quality of life). A total IBDQ score is calculated by summing the scores from each domain; the total IBDQ score ranges from 32 to 224. An IBDQ total score greater than 170 is characteristic of the health related quality of life (HRQoL) of patients in remission.
  • As used herein, “induction therapy” is an initial stage of therapy, wherein a patient is administered a relatively intensive dosing regimen of a therapeutic agent. The therapeutic agent, e.g., antibody, is administered in a way that quickly provides an effective amount of the agent suitable for certain purposes, such as inducing immune tolerance to the agent or for inducing a clinical response and ameliorating disease symptoms (see WO 2012/151247 and WO 2012/151248, incorporated herein by reference).
  • As used herein, “maintenance therapy” is after induction therapy and is administered in a way that continues the response achieved by induction therapy with a stable level of therapeutic agent, e.g., antibody. A maintenance regimen can prevent return of symptoms or relapse of disease, e.g., IBD (see WO 2012/151247 and WO 2012/151248, incorporated herein by reference). A maintenance regimen can provide convenience to the patient, e.g., be a simple dosing regimen or require infrequent trips for treatment.
  • The cell surface molecule, “α4β7 integrin,” or “α4β7,” is a heterodimer of an α4 chain (CD49D, ITGA4) and a β7 chain (ITGB7). Each chain can form a heterodimer with an alternative integrin chain, to form α4β1 or αEβ7. Human α4 and β7 genes (GenBank (National Center for Biotechnology Information, Bethesda, Md.) RefSeq Accession numbers NM_000885 and NM_000889, respectively) are expressed by B and T lymphocytes, particularly memory CD4+ lymphocytes. Typical of many integrins, α4β7 can exist in either a resting or activated state. Ligands for α4β7 include vascular cell adhesion molecule (VCAM), fibronectin and mucosal addressin (MAdCAM (e.g., MAdCAM-1)). The α4β7 integrin mediates lymphocyte trafficking to GI mucosa and gut-associated lymphoid tissue (GALT) through adhesive interaction with mucosal addressin cell adhesion molecule-1 (MAdCAM-1), which is expressed on the endothelium of mesenteric lymph nodes and GI mucosa.
  • The term “antibody” herein is used in the broadest sense and specifically covers full length monoclonal antibodies, immunoglobulins, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two full length antibodies, e.g., each to a different antigen or epitope, and individual antigen binding fragments, including dAbs, scFv, Fab, F(ab)′2, Fab′, including human, humanized and antibodies from non-human species and recombinant antigen binding forms such as monobodies and diabodies.
  • The term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. The modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567). The “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991), for example.
  • “Antigen binding fragments” of an antibody comprise at least the variable regions of the heavy and/or light chains of an anti-α4β7 antibody. For example, an antigen binding fragment of vedolizumab comprises amino acid residues 20-131 of the humanized light chain sequence of SEQ ID NO:2. Examples of such antigen binding fragments include Fab fragments, Fab′ fragments, scFv and F(ab′)2 fragments of a humanized antibody known in the art. Antigen binding fragments of the humanized antibody of the invention can be produced by enzymatic cleavage or by recombinant techniques. For instance, papain or pepsin cleavage can be used to generate Fab or F(ab′)2 fragments, respectively. Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site. For example, a recombinant construct encoding the heavy chain of an F(ab′)2 fragment can be designed to include DNA sequences encoding the CH1 domain and hinge region of the heavy chain. In one aspect, antigen binding fragments inhibit binding of α4β7 integrin to one or more of its ligands (e.g. the mucosal addressin MAdCAM (e.g., MAdCAM-1), fibronectin).
  • The terms “Fc receptor” or “FcR” are used to describe a receptor that binds to the Fc region of an antibody. In one aspect, the FcR is a native sequence human FcR. In another aspect, the FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the FcγRI, Fc-γRII, and FcγRIII subclasses, including allelic variants and alternatively spliced forms of these receptors. FcγRII receptors include FcγRIIA (an “activating receptor”) and FcγRIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof. Activating receptor FcγRIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain. Inhibiting receptor FcγRIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain. (See review in M. Daeron, Annu. Rev. Immunol. 15:203-234 (1997)). FcRs are reviewed in Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991); Capel et al., Immunomethods 4:25-34 (1994); and de Haas et al., J. Lab. Clin. Med. 126:33-41 (1995). Other FcRs, including those to be identified in the future, are encompassed by the term “FcR” herein. The term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., J. Immunol. 117:587 (1976) and Kim et al., J. Immunol. 24:249 (1994)) and for regulating the persistence of immunoglobulin G (IgG) and albumin in the serum (reviewed by Rath et al., J. Clin. Immunol. 33 Suppl 1:S9-17 (2013)).
  • The term “hypervariable region” when used herein refers to the amino acid residues of an antibody which are responsible for antigen binding and are found in the “variable domain” of each chain. The hypervariable region generally comprises amino acid residues from a “complementarity determining region” or “CDR” (e.g. residues 24-34 (L1), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)) and/or those residues from a “hypervariable loop” (e.g. residues 26-32 (L1), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)). “Framework Region” or “FR” residues are those variable domain residues other than the hypervariable region residues as herein defined. The hypervariable region or the CDRs thereof can be transferred from one antibody chain to another or to another protein to confer antigen binding specificity to the resulting (composite) antibody or binding protein.
  • An “isolated” antibody is one which has been identified and separated and/or recovered from a component of its natural environment. In certain embodiments, the antibody will be purified (1) to greater than 95% by weight of protein as determined by the Lowry method, and alternatively, more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using Coomassie blue or silver stain. Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • “Treatment” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disease as well as those in which the disease or its recurrence is to be prevented. Hence, the patient to be treated herein may have been diagnosed as having the disease or may be predisposed or susceptible to the disease. The terms “patient” and “subject” are used interchangeably herein.
  • Treatment of IBD with Anti-α4β7 Antibodies
  • In one aspect, the invention relates to a method of treating IBD in a subject comprising administering to the subject an anti-α4β7 antibody described herein in an amount effective to treat IBD, e.g., in humans. The human patient or subject may be an adult (e.g., 18 years or older), an adolescent, or a child. A pharmaceutical composition comprising an anti-α4β7 antibody can be used as described herein for treating IBD in a subject suffering therefrom.
  • The anti-α4β7 antibody can bind to an epitope on the α4 chain (e.g., humanized MAb 21.6 (Bendig et al., U.S. Pat. No. 5,840,299), on the β7 chain (e.g., FIB504 or a humanized derivative (e.g., Fong et al., U.S. Pat. No. 7,528,236)), or to a combinatorial epitope formed by the association of the α4 chain with the β7 chain. In one aspect, the antibody binds a combinatorial epitope on the α4β7 complex, but does not bind an epitope on the α4 chain or the β7 chain unless the chains are in association with each other. The association of α4 integrin with β7 integrin can create a combinatorial epitope for example, by bringing into proximity residues present on both chains which together comprise the epitope or by conformationally exposing on one chain, e.g., the α4 integrin chain or the β7 integrin chain, an epitopic binding site that is inaccessible to antibody binding in the absence of the proper integrin partner or in the absence of integrin activation. In another aspect, the anti-α4β7 antibody binds both the α4 integrin chain and the β7 integrin chain, and thus, is specific for the α4β7 integrin complex. Such antibodies can bind α4β7 but not bind α4β1, and/or not bind αEβ7, for example. In another aspect, the anti-α4β7 antibody binds to the same or substantially the same epitope as the Act-1 antibody (Lazarovits, A. I. et al., J. Immunol., 133(4): 1857-1862 (1984), Schweighoffer et al., J. Immunol., 151(2): 717-729, 1993; Bednarczyk et al., J. Biol. Chem., 269(11): 8348-8354, 1994). Murine ACT-1 Hybridoma cell line, which produces the murine Act-1 monoclonal antibody, was deposited under the provisions of the Budapest Treaty on Aug. 22, 2001, on behalf of Millennium Pharmaceuticals, Inc., 40 Landsdowne Street, Cambridge, Mass. 02139, U.S.A., at the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209, U.S.A., under Accession No. PTA-3663. In another aspect, the anti-α4β7 antibody is a human antibody or an α4β7 binding protein using the CDRs provided in U.S. Patent Application Publication No. 2010/0254975.
  • In one aspect, the anti-α4β7 antibody inhibits binding of α4β7 to one or more of its ligands (e.g. the mucosal addressin, e.g., MAdCAM (e.g., MAdCAM-1), fibronectin, and/or vascular addressin (VCAM)). Primate MAdCAMs are described in the PCT publication WO 96/24673, the entire teachings of which are incorporated herein by this reference. In another aspect, the anti-α4β7 antibody inhibits binding of α4β7 to MAdCAM (e.g., MAdCAM-1) and/or fibronectin without inhibiting the binding of VCAM.
  • In one aspect, the anti-α4β7 antibodies for use in the treatments are humanized versions of the mouse Act-1 antibody. Suitable methods for preparing humanized antibodies are well-known in the art. Generally, the humanized anti-α4β7 antibody will contain a heavy chain that contains the 3 heavy chain complementarity determining regions (CDRs, CDR1, SEQ ID NO:4, CDR2, SEQ ID NO:5 and CDR3, SEQ ID NO:6) of the mouse Act-1 antibody and suitable human heavy chain framework regions; and also contain a light chain that contains the 3 light chain CDRs (CDR1, SEQ ID NO:7, CDR2, SEQ ID NO:8 and CDR3, SEQ ID NO:9) of the mouse Act-1 antibody and suitable human light chain framework regions. The humanized Act-1 antibody can contain any suitable human framework regions, including consensus framework regions, with or without amino acid substitutions. For example, one or more of the framework amino acids can be replaced with another amino acid, such as the amino acid at the corresponding position in the mouse Act-1 antibody. The human constant region or portion thereof, if present, can be derived from the κ or λ light chains, and/or the γ (e.g., γ1, γ2, γ3, γ4), μ, α (e.g., α1, α2), δ or ε heavy chains of human antibodies, including allelic variants. A particular constant region (e.g., IgG1), variant or portions thereof can be selected in order to tailor effector function. For example, a mutated constant region (variant) can be incorporated into a fusion protein to minimize binding to Fc receptors and/or ability to fix complement (see e.g., Winter et al., GB 2,209,757 B; Morrison et al., WO 89/07142; Morgan et al., WO 94/29351, Dec. 22, 1994). Humanized versions of Act-1 antibody were described in PCT publications nos. WO98/06248 and WO07/61679, the entire teachings of each of which are incorporated herein by this reference.
  • In one aspect, the anti-α4β7 antibody is vedolizumab. Vedolizumab (also called MLN0002, ENTYVIO™ or KYNTELES™) is a humanized immunoglobulin (Ig) G1 mAb directed against the human lymphocyte integrin α4β7. Vedolizumab binds the α4β7 integrin, antagonizes its adherence to MAdCAM-1 and as such, impairs the migration of gut homing leukocytes into GI mucosa. Vedolizumab is an integrin receptor antagonist indicated for adult patients with moderately to severely active UC or CD who have had an inadequate response with, lost response to, or were intolerant to a tumor necrosis factor (TNF) blocker or immunomodulator, or had an inadequate response with, were intolerant to, or demonstrated dependence on corticosteroids. For UC, vedolizumab is for inducing and maintaining clinical response, inducing and maintaining clinical remission, improving endoscopic appearance of the mucosa, and/or achieving corticosteroid-free remission. For CD, vedolizumab is for achieving clinical response, achieving clinical remission, and/or achieving corticosteroid-free remission. In some embodiments, corticosteroid-free remission is achieved through a tapering regimen during continued treatment with vedolizumab.
  • In another aspect, the humanized anti-α4β7 antibody for use in the treatment comprises a heavy chain variable region comprising amino acids 20 to 140 of SEQ ID NO:1, and a light chain variable region comprising amino acids 20 to 131 of SEQ ID NO:2 or amino acids 21 to 132 of SEQ ID NO:3. If desired, a suitable human constant region(s) can be present. For example, the humanized anti-α4β7 antibody can comprise a heavy chain that comprises amino acids 20 to 470 of SEQ ID NO:1 and a light chain comprising amino acids 21 to 239 of SEQ ID NO:3. In another example, the humanized anti-α4β7 antibody can comprise a heavy chain that comprises amino acids 20 to 470 of SEQ ID NO:1 and a light chain comprising amino acids 20 to 238 of SEQ ID NO:2. The humanized light chain of vedolizumab (e.g., Chemical Abstract Service (CAS, American Chemical Society) Registry number 943609-66-3), with two mouse residues switched for human residues, is more human than the light chain of LDP-02, another humanized anti-α4β7 antibody. In addition, LDP-02 has the somewhat hydrophobic, flexible alanine 114 and a hydrophilic site (Aspartate 115) that is replaced in vedolizumab with the slightly hydrophilic hydroxyl-containing threonine 114 and hydrophobic, potentially inward facing valine 115 residue.
  • Further substitutions to the humanized anti-α4β7 antibody sequence can be, for example, mutations to the heavy and light chain framework regions, such as a mutation of isoleucine to valine on residue 2 of SEQ ID NO:10; a mutation of methionine to valine on residue 4 of SEQ ID NO:10; a mutation of alanine to glycine on residue 24 of SEQ ID NO:11; a mutation of arginine to lysine at residue 38 of SEQ ID NO:11; a mutation of alanine to arginine at residue 40 of SEQ ID NO:11; a mutation of methionine to isoleucine on residue 48 of SEQ ID NO:11; a mutation of isoleucine to leucine on residue 69 of SEQ ID NO:11; a mutation of arginine to valine on residue 71 of SEQ ID NO:11; a mutation of threonine to isoleucine on residue 73 of SEQ ID NO:11; or any combination thereof; and replacement of the heavy chain CDRs with the CDRs (CDR1, SEQ ID NO:4, CDR2, SEQ ID NO:5 and CDR3, SEQ ID NO:6) of the mouse Act-1 antibody; and replacement of the light chain CDRs with the light chain CDRs (CDR1, SEQ ID NO:7, CDR2, SEQ ID NO:8 and CDR3, SEQ ID NO:9) of the mouse Act-1 antibody.
  • The present invention provides, in a first aspect, a method for treating a patient having inflammatory bowel disease (IBD) with an anti-α4β7 antibody, e.g., vedolizumab. In this aspect, the method comprises determining pharmacokinetic factors of the patient. In some embodiments, the method comprises selecting for treatment a patient who has low clearance of the antibody. The method comprises measuring the concentration of the anti-α4β7 antibody in a biological sample from the patient, e.g., blood, serum, plasma, stool, bowel fluid, saliva, inflammatory exudate, at a time, e.g., at least one, two, three, four, five or six weeks, after receiving at least one prior dose of the antibody. The measurement provides an indication of clearance, for which “low” is defined by the various methods, and results criteria thereof described herein, of obtaining a clearance indication. An indication of clearance may result from the measurement of the concentration of anti-α4β7 antibody in the sample, which may be used alone or used in a calculation, as described herein, of the rate the anti-α4β7 antibody is eliminated from the body, e.g., the blood volume. The calculation may be based on knowledge of the amount of anti-α4β7 antibody in the prior dose, the size of the patient or blood volume, the number of days between the day of the prior dose of the anti-α4β7 antibody and the day of obtaining the sample for measurement or from a pharmacokinetics model, e.g., a model as described herein. In some embodiments, measurement of serum concentration of anti-α4β7 antibody may be an indicator of clearance. Clearance may be affected or further illustrated by other parameters, such as pharmacodynamic factors, clinical factors, inflammation or immune response factors, whose measurement may be used in combination with the measurement of anti-α4β7 antibody. An indication of clearance, alone or in combination with measurements of one or more other parameters, may be used to predict response to anti-α4β7 antibody treatment, identify a patient who is responding to anti-α4β7 antibody treatment, select a patient for further treatment with anti-α4β7 antibody or monitor the effectiveness of the anti-α4β7 antibody during treatment. Low clearance or sufficient amounts of anti-α4β7 antibody in the patient, e.g., at the time of sampling, indicates that further dosing with anti-α4β7 antibody will provide benefit in the treatment of IBD. In some embodiments, a method wherein low clearance is indicated, measured or calculated at end of induction comprises further treatment of the patient for maintenance of response or remission, achieving corticosteroid-free remission, improving endoscopic appearance of the mucosa. Preferably, a patient who has low clearance of the antibody may be characterized, a) by a rate of antibody, e.g., vedolizumab, clearance that is less than about 0.25 L/day, less than 0.20 L/day, between 0.1 to 0.2 L/day, less than 0.15 L/day or less than 0.1. L/day; and/or b) by a serum concentration of antibody, e.g., vedolizumab, that is at least 8 μg per ml, 10 μg per ml, 12 μg per ml, 14 μg per ml, 17 μg per ml, 20 μg per ml, 25 μg per ml, 30 μg per ml; 35 μg per ml, or 40 μg per ml or has a range of 12-25 μg per ml, 15-17 μg per ml, 17-25 μg per ml, 12-40 μg per ml, and 17-40 μg ml.
  • In one embodiment, the method for treating a patient having inflammatory bowel disease (IBD) with an anti-α4β7 antibody, e.g., vedolizumab comprises the steps of selecting a human patient having IBD from a group of two or more patients having or suffering from IBD that has, at a time point of four weeks after receiving a second dose of vedolizumab, where the first dose of vedolizumab was administered to the subject two weeks prior to the second dose, a serum concentration of at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 μg per ml. Specifically, the patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 μg per ml. The patient's serum concentration, e.g., a trough serum concentration, may be greater than 17 μg/ml, greater than 25 μg/ml, or greater than 35 μg/ml. In some embodiments, the patient received the prior dose of vedolizumab about two weeks, about three weeks, about four weeks, about five weeks or about six weeks prior to the sampling for serum vedolizumab measurement. Once such a patient is selected from a group of patients, he or she is administered vedolizumab to treat the IBD.
  • In another aspect, the present invention provides a method for treating a patient having inflammatory bowel disease (IBD) with an anti-α4β7 antibody, e.g., vedolizumab. In some embodiments, the method using vedolizumab comprises the steps of administering two doses of vedolizumab to a patient suffering from IBD, wherein the second dose is administered about two weeks after the first dose is administered to the patient; waiting a period of time of at least two weeks, at least three weeks, about four weeks or five weeks; measuring the patient's serum concentration of vedolizumab; and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 μg per ml. The patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 μg per ml. The patient's serum concentration, e.g., a trough serum concentration, may be greater than 17 μg/ml, greater than 25 μg/ml, or greater than 35 μg/ml.
  • Alternatively, at least one dose of the anti-α4β7 antibody, e.g., vedolizumab may be administered to a patient suffering from IBD, waiting at least about two weeks, or optionally, a period of two to five weeks, and then measuring the patient's serum concentration of vedolizumab and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 μg per ml. The patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 μs per ml. The patient's serum concentration, e.g., a trough serum concentration, may be greater than 17 μg/ml, greater than 25 μg/ml, or greater than 35 μg/ml.
  • In another aspect, the present invention provides a method of identifying a patient (e.g., a patient having mucosal healing) for continued treatment with vedolizumab. The method may comprise the steps of measuring the concentration of vedolizumab in a sample of serum obtained from a patient suffering from IBD and who received at least two doses of vedolizumab within the previous four months (e.g, within the previous three months, within the previous two months), and identifying the patient for continued treatment with vedolizumab if the serum concentration in the sample is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 μg per ml. The patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 μg per ml. The patient's serum concentration, e.g., a trough serum concentration, may be greater than 17 μg/ml, greater than 25 μg/ml, or greater than 35 μg/ml. In some embodiments, the patient received the last prior dose, e.g., the second dose, of vedolizumab about four weeks prior to the sampling for serum vedolizumab measurement. In other embodiments, the patient received the last prior dose 3 to 8 weeks prior to the sampling for serum vedolizumab measurement.
  • Alternatively, the method of identifying a patient for continued treatment with an anti-α4β7 antibody, e.g., vedolizumab may comprise the steps of measuring the concentration of vedolizumab in a sample of serum obtained from a patient suffering from IBD and who received at least one dose of vedolizumab within the previous one or two months, and identifying the patient for continued treatment with vedolizumab if the serum concentration in the sample is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 μs per ml. The patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 μg per ml. The patient's serum concentration, e.g., a trough serum concentration, may be greater than 17 μg/ml, greater than 25 μg/ml, or greater than 35 μg/ml. In some embodiments, the patient received the prior dose of vedolizumab about two weeks, about three weeks, about four weeks, about five weeks or about six weeks prior to the sampling for serum vedolizumab measurement.
  • Alternatively, at least one dose of the anti-α4β7 antibody, e.g., vedolizumab may be administered to a patient suffering from IBD, waiting at least about two weeks, or optionally, a period of two to five weeks, and then measuring the patient's serum concentration of vedolizumab and administering one or more further doses of vedolizumab to the patient if the patient's serum concentration is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 μg per ml. The patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 μg per ml. The patient's serum concentration, e.g., a trough serum concentration, may be greater than 17 μg/ml, greater than 25 μg/ml, or greater than 35 μg/ml.
  • Vedolizumab may be administered by any suitable method, such as by one or more of intravenous injection, subcutaneous injection, or infusion. In some embodiments, vedolizumab is administered at a dose of 50 mg, 100 mg, 180 mg, 300 mg, or 600 mg. In some embodiments, the vedolizumab is administered, for example subcutaneously, at a dose of 0.5 mg/kg, 1.0 mg/kg, 1.5 mg/kg, 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg. 4.0 mg/kg, or 5.0 mg/kg, at a dose of 108 mg, 216 mg, 160 mg or 165 mg. The vedolizumab may be administered once per day, per week, per month, or per year. A vedolizumab dosing regimen may have an initial or induction phase and a maintenance phase. An induction phase may be one or more than one, e.g., two, three or four doses, of high amounts or without long times, such as only one week, two weeks, three weeks or four weeks between each dose. For example, an induction regimen may have two doses, one at day (week) zero and one at week 2 (day 14). A maintenance phase, e.g., to maintain remission of the IBD, may have lower doses or doses further apart than in the induction phase. In some embodiments, the vedolizumab is administered at zero, two and six weeks (induction), and then every four weeks or every eight weeks thereafter (maintenance). Patients with IBD refractory to other therapies may need longer induction periods, e.g., 8, 10 or 12 weeks, before beginning maintenance therapy. In an embodiment, vedolizumab is administered intravenously at zero, two and six weeks, then every eight weeks thereafter. In some embodiments, vedolizumab is administered one or more times, and then at least one month, at least six months, or at least one year later, vedolizumab is again administered one or more times. In some embodiments, 300 mg vedolizumab may be administered by intravenous infusion at zero, two, and six weeks, and then at four weeks intervals or eight week intervals thereafter. In some embodiments, 300 mg vedolizumab may be administered by intravenous infusion at zero, two, and six weeks, and then at two-, three- or four-week intervals, 108 mg of vedolizumab may be administered subcutaneously. Treatment methods using anti-α4β7 integrin antibodies are described in publication nos. U.S. 2005/0095238, WO2012151248 and WO 2012/151247.
  • In some embodiments, the method of identifying a patient for continued treatment with an anti-α4β7 antibody, e.g., vedolizumab may comprise a clinical measure. A clinical measure may be mucosal healing. An endoscopic score decreased from baseline, e.g. 25% less, 35% less, 40% less, 45% less, 50% less, 55% less, 60% less, 65% less, or 75% less than baseline, may indicate mucosal healing. In ulcerative colitis, an endoscopic score of less than 4, less than 3, ≦1, or between 0 and 2 may help identify a patient for continued treatment. In Crohn's disease, a patient with mucosal healing, and thus a candidate for continued treatment with an anti-α4β7 antibody, e.g., vedolizumab, may have an assessment selected from the group consisting of endoscopic response, endoscopic remission, improvement in the MaRIA score, decrease in ulceration, and improvement in a mucosal healing parameter, e.g., an MREn parameter. In one embodiment, a MaRIA score is the sum of each segment calculation, wherein a sum that one would calculate for each segment=(1.56×wall thickness (mm))+(0.02×Relative Contrast Enhancement (RCE), e.g., after administration of intravenous contrast with gadolinium)+(5×edema)+(10×ulceration)). In some embodiments endoscopic response is achieved by about a 25% decrease, about a 40% decrease, about a 50% decrease, about a 60% decrease or about a 75% decrease in SES-CD score from baseline. In some embodiments, endoscopic remission is achieved by an SES-CD score of ≦6, ≦5, ≦4 or ≦3. In some embodiments, mucosal healing is achieved by a MaRIA score of <14, <13, <12, <11, <10, <9, <8, <7, <6, <5 or <4. In some embodiments a decrease in ulceration is selected from the group consisting of a decrease in the size of ulcers, a decrease in the percent of ulcerated surface, a decrease in the percent of affected surface and decrease in the narrowings of the canal. In some embodiments, the size of ulcers is less than 2 cm in diameter, 0.5 to 2 cm in diameter, 0.1 to 0.5 cm in diameter or <0.2 cm diameter. In some embodiments, the ulcerated surface is less than 30%, 10-30%, less than 10%, or 0. In some embodiments the affected surface is less than 75%, 50%-75%, less than 50%, less than 25% or unaffected. In some embodiments, the wall thickness is decreased by about 10%, about 15%, about 20%, about 25%, about 30%, about 20%-40% or more than 45% from baseline. In some embodiments, the bowel wall contrast (RCE) is decreased about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 20%-40% or more than 40% from baseline. In some embodiments, the mural edema is decreased by about 25%, about 40%, about 50%, about 60%, about 70%, more than 75%, or about 70%-90% from baseline. In some embodiments, the perienteric vascularity is decreased by about 25%, about 30%, about 40%, about 50%, about 60%, about 50%-70%, about 75% or more than 75% from baseline.
  • In an embodiment, a Crohn's disease patient who is responsive to vedolizumab has an SES-CD of ≦4 at week 6, 10, 12, 14, 22 or 26 after initiating treatment. In an embodiment, a Crohn's disease patient who is responsive to vedolizumab has an endoscopic response or ≧50% reduction of SES-CD at week 6, 10, 12, 14, 22 or 26 after initiating treatment. In an embodiment, a Crohn's disease patient who is responsive to vedolizumab has a clinical remission or ≧70 or ≧100 point reduction of CDAI score at week 6, 10, 12, or 14 after initiating treatment. In an embodiment, a Crohn's disease patient who is responsive to vedolizumab has a MaRIA score of <15, <12, <10 or <7 at week 6, 10, 12, 14, respectively, globally or on a per segment basis, after initiating treatment. In an embodiment, a Crohn's disease patient who is responsive to vedolizumab has no ulceration at week 14, 22 or 26 after initiating treatment. In an embodiment, a Crohn's disease patient who is responsive to vedolizumab has no fistulae at week 14, 22, 26 or 30 after initiating treatment. A patient who is responsive to vedolizumab may continue to be treated, e.g., may continue a maintenance regimen, with vedolizumab. In one embodiment, a maintenance regimen comprises a dose of vedolizumab once every 2 weeks. In one embodiment, a maintenance regimen comprises a dose of vedolizumab once every 4 weeks. In one embodiment, a maintenance regimen comprises a dose of vedolizumab once every 6 weeks. In one embodiment, a maintenance regimen comprises a dose of vedolizumab once every 8 weeks.
  • In another aspect, the present invention relates to a method for maintaining remission of inflammatory bowel disease in a patient. The patient may have received at least one dose of vedolizumab in the previous two months, the previous three months or the previous four months. The method may comprise the steps of obtaining a serum sample from the patient, measuring the concentration of vedolizumab in the sample, and administering vedolizumab thereafter every eight weeks if the concentration is at least about 8, about 10, about 12, about 14, about 17, about 20, about 25, about 30, about 35, or about 40 μg per ml. The patient's serum concentration may be between about 12-25, about 15-17, about 17-25, about 12-40, or about 17-40 μg per ml. The patient's serum concentration, e.g., a trough serum concentration, may be greater than 17 μg/ml, greater than 25 μg/ml, or greater than 35 μg/mi. In some embodiments, the patient received the prior dose of vedolizumab about two weeks, about three weeks, about four weeks, about five weeks or about six weeks prior to the sampling for serum vedolizumab measurement.
  • In yet another aspect of the present invention, a method for continuing to treat an IBD patient with vedolizumab comprises the steps of providing two doses of vedolizumab, wherein the second dose is about two weeks after the first; waiting about four weeks; and continuing to treat the patient with vedolizumab if the patient is in deep remission. Deep remission may be determined by measuring an endoscopic subscore for the patient and is defined as having an endoscopic subscore of 0 to 1. The method may further comprise the step of determining a patient-reported outcome score (e.g., a subscore of 0 to 1). The patient-reported outcome may comprise a rectal bleeding subscore (e.g., 0) and/or a stool frequency subscore (e.g., decrease or no change; 0 or 1).
  • The invention also relates to a method for identifying a patient for continued treatment with vedolizumab comprising the steps of measuring the clearance of vedolizumab in a biological sample obtained from a patient suffering from IBD and who was administered at least two doses of vedolizumab within the previous four months (e.g., within the previous three months, within the previous two months), and identifying the patient for continued treatment with vedolizumab if the clearance in the patient is less than 0.25 L/day, less than 0.20 L/day, or between 0.1 to 0.2 L/day. The biological sample may be any biological sample, for example, serum, plasma, saliva, urine, or feces. Optionally, the method may further comprise measuring anti-α4β7 antibody antibodies. A low or absent immune response to the anti-α4β7 antibody, e.g., anti-vedolizumab antibody, e.g., a titer of ≦50, ≦125 or ≦575 may further identify the patient for continued treatment with vedolizumab.
  • Methods to determine whether a patient will respond to treatment with an anti-α4β7 antibody or whether to continue treating a patient with an anti-α4β7 antibody may further comprise measuring albumin concentration. In therapeutic antibody therapy, this can be a reflection of clearance activity, such as ability to bind the neonate FcR. In cases of low serum albumin levels, the anti-α4β7 antibody can have a high clearance. Consequently, a patient with high serum albumin levels may not respond or may take longer to respond to treatment with anti-α4β7 antibody. An albumin concentration greater than about 3.0 g/dL, about 3.2 g/dL, about 4.0 g/dL, about 4.7 g/dL, or about 5.0 g/dL, or in the range of 3.3 to 5.0 g/dL, in the range of 3.5 to 5.0 g/dL, in the range of 3.8 to 5.0 g/dL or in the range of 4.0 to 5.0 g/dL may further identify the patient for continued treatment with the anti-α4β7 antibody, e.g., vedolizumab. The albumin concentration measurement may be accompanied by measurement of patient weight. A high weight patient, e.g., greater than 90 kg, greater than 100 kg, greater than 110 kg, or greater than 120 kg, with low albumin levels, e.g., less than 4.2 g/dL, less than 4.0 g/dL, less than 3.5 g/dL or less than 3.2 g/dL, may have high anti-α4β7 antibody clearance and thus may not respond to therapy with the anti-α4β7 antibody or may need a higher or more frequent dose of the anti-α4β7 antibody for continued treatment.
  • Clearance, e.g., linear clearance, e.g., the volume of blood which is cleared of drug per unit time, may be calculated/estimated/derived by any appropriate means known to those skilled in the art. For example, clearance may be estimated by population approach or Bayesian methods, e.g., the full Bayesian Markov Chain Monte Carlo (MCMC) method. One method of developing the pharmacokinetic model of linear clearance is described in the Examples. The anti-α4β7 antibody exposure metric, such as trough serum concentration, e.g., serum concentration of anti-α4β7 antibody prior, e.g., 1 day, 2 days, 3 days, 4 days or up to a week prior, to administering a new dose, peak serum concentration, average serum concentration measured at more than one sampling or area under the concentration time curve, is inputted into the model to determine clearance.
  • Alternatively, the method for identifying a patient for continued treatment with vedolizumab comprising the steps of measuring the clearance of vedolizumab in a biological sample obtained from a patient suffering from IBD can be performed on a patient who was administered at least one dose of vedolizumab within the previous one or two months, and identifying the patient for continued treatment with vedolizumab if the clearance in the patient is less than 0.25 L/day, less than 0.20 L/day, between 0.1 to 0.2 L/day, less than 0.15 L/day or less than 0.1. L/day. The biological sample may be any biological sample, for example, serum, plasma, saliva, urine, or feces.
  • The method may further comprise measuring an endoscopic subscore. Anti-α4β7 antibody, e.g., vedolizumab treatment may be continued with an endoscopic subscore of less than about 3, less than about 2.5, less than about 2, between about 0-2, or less than or equal to 1.
  • Fecal levels of calprotectin, a neutrophil cytosolic protein, correlate with endoscopic activity in ulcerative colitis. Typically, a non-diseased subject will have a fecal calprotectin level of less than 50 μg/g. A fecal calprotectin level greater than 50 but less than 150 μg/g may be a sign of possible mucosal inflammation, whereas fecal calprotectin levels greater than 150 μg/g is usually a sign of active inflammation. The methods described herein may further comprise measuring the fecal calprotectin concentration. Higher levels of fecal calprotectin are associated with a greater risk of relapse. Vedolizumab treatment may be continued with a fecal calprotectin concentration of less than 1500 μg/g, less 1250 μg/g, less than 1000 μg/g, less than 750 μg/g, less than 500 μg/g, less than 400 μg/g, less than 300 μg/g, less than 250 μg/g, between 200-1200 μg/g, between 350 to 800 μg/g, between 300-1000 μg/g, <50 μg/g, <100 μg/g, <150 μg/g, <200 μg/g, ≦250-499 μg/g, or between 500 to 900 μg/g. Alternatively, fecal calprotectin may be reduced to less than about 50%, 45%, 40%, 35%, 30%, 25%, 20%, between 10-55%, between 10-30%, between 15-35%, between 15-45% or between 20-40% of the baseline or concentration before treatment. Fecal calprotectin in a stool sample can be measured using the PHICAL test kit (Calpro, Lysaker Norway).
  • Pharmacokinetic and Pharmacodynamic Assays
  • The anti-α4β7 antibody, e.g., vedolizumab, serum concentration may be measured by any appropriate means known by those skilled in the art. In one aspect, the vedolizumab serum concentration is measured by a sandwich enzyme-linked immunosorbent assay (ELISA) assay. In another aspect, use of a pharmacodynamic assay, inhibition of MAdCAM-1-Fc binding to α4β7-expressing peripheral blood cells by the anti-α4β7 antibody, e.g., vedolizumab in the blood is used as a measure of the extent of α4β7 saturation by the anti-α4β7 antibody, e.g., vedolizumab.
  • In an embodiment, the anti-α4β7 antibody amount in serum can be measured in a pharmacokinetic assay. An immobilized phase, such as a microtiter plate, vessel or bead is coated with a reagent which specifically binds to the anti-α4β7 antibody. The immobilized reagent is contacted with a patient sample, e.g., serum, which may or may not comprise the anti-α4β7 antibody. After incubation and washing, the anti-α4β7 antibody complexed to the coating reagent is contacted with a reagent which binds to the captured antibody and may be detected, e.g., using a label such as horseradish peroxidase (HRP). The binding reagent may be an anti-human antibody, e.g., polyclonal or monoclonal, which binds to the Fc portion of the anti-α4β7 antibody. Addition of an HRP substrate, such as 3,3′,5,5′-tetramethylbenzidine (TMB), can allow signal accumulation, such as color development, that can be measured, e.g., spectrophotographically.
  • In some embodiments, the coating reagent is an anti-idiotypic antibody which specifically binds to the anti-α4β7 antibody, e.g., its variable region or a portion thereof comprising one or more CDRs, such as heavy chain CDR3, SEQ ID NO:6. The anti-idiotypic anti-α4β7 antibody for use in the assay can be specific for, and thus bind, the α4β7 integrin-binding portion of the anti-α4β7 antibody but is not specific for the Fc portion of the anti-α4β7 antibody and thus does not bind the Fc portion of the anti-α4β7 antibody. The anti-idiotypic anti-α4β7 antibody for use in the assay can be specific for, and thus bind, a variable region of the heavy and/or light chain of anti-α4β7 antibody, e.g., selected from the group consisting of amino acids 20 to 140 of SEQ ID NO:1, amino acids 20 to 131 of SEQ ID NO:2 and amino acids 21 to 132 of SEQ ID NO:3. The anti-idiotypic anti-α4β7 antibody for use in the assay can be specific for, and thus bind, an antigen-binding fragment of the anti-α4β7 antibody. The anti-idiotypic antibody can be isolated from an immunization process using the anti-α4β7 antibody or an α4β7 integrin-binding portion thereof, such as an antibody fragment comprising one or more CDRs, and used as isolated or produced by a recombinant method. In some embodiments, the anti-idiotypic anti-α4β7 antibody is raised against an immunogen comprising heavy chain CDR3, SEQ ID NO:6. In other embodiments, the anti-idiotypic anti-α4β7 antibody is raised against an immunogen comprising a variable region of the heavy and/or light chain of anti-α4β7 antibody, e.g., selected from the group consisting of amino acids 20 to 140 of SEQ ID NO:1, amino acids 20 to 131 of SEQ ID NO:2 and amino acids 21 to 132 of SEQ ID NO:3. In some embodiments, the anti-idiotypic antibody is a monoclonal antibody. In some embodiments, an scFv fragment of the anti-idiotypic antibody is used in the assay. In other embodiments, the intact anti-idiotypic antibody is used in the assay.
  • Generation of an anti-idiotypic anti-α4β7 antibody can proceed in the following general methods. Immunization of a suitable animal (e.g., mouse, rat, rabbit or sheep) with protein, e.g., anti-α4β7 antibody or an α4β7 integrin binding portion thereof, or fusion protein comprising the portion, can be performed with the immunogen prepared for injection in a manner to induce a response, e.g., with adjuvant, e.g., complete Freund's adjuvant. Other suitable adjuvants include TITERMAX GOLD® adjuvant (CYTRX Corporation, Los Angeles, Calif.) and alum. Small peptide immunogens, such as a fragment comprising a CDR, such as CDR3 of the heavy chain can be linked to a larger molecule, such as keyhole limpet hemocyanin. Mice can be injected in a number of manners, e.g., subcutaneous, intravenous or intramuscular at a number of sites, e.g., in the peritoneum (i.p.), base of the tail, or foot pad, or a combination of sites, e.g., i.p. and base of tail. Booster injections can include the same or a different immunogen and can additionally include adjuvant, e.g., incomplete Freund's adjuvant. Generally, where a monoclonal antibody is desired, a hybridoma is produced by fusing a suitable cell from an immortal cell line (e.g., a myeloma cell line such as SP2/0, P3X63Ag8.653 or a heteromyeloma) with antibody-producing cells. Antibody-producing cells can be obtained from the peripheral blood or, preferably the spleen or lymph nodes, of animals immunized with the antigen of interest. Cells that produce antibodies can be produced using suitable methods, for example, fusion of a human antibody-producing cell and a heteromyeloma or trioma, or immortalization of an activated human B cell via infection with Epstein Barr virus. (See, e.g., U.S. Pat. No. 6,197,582 (Trakht); Niedbala et al., Hybridoma, 17:299-304 (1998); Zanella et al., J Immunol Methods, 156:205-215 (1992); Gustafsson et al., Hum Antibodies Hybridomas, 2:26-32 (1991).) The fused or immortalized antibody-producing cells (hybridomas) can be isolated using selective culture conditions, and cloned by limiting dilution. Cells which produce antibodies with the desired specificity can be identified using a suitable assay (e.g., ELISA (e.g., with immunogen immobilized on the microtiter well).
  • The anti-α4β7 antibody or the anti-idiotypic anti-α4β7 antibody may be produced by expression of nucleic acid sequences encoding each chain in living cells, e.g., cells in culture. A variety of host-expression vector systems may be utilized to express the antibody molecules of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an anti-α4β7 antibody in situ. These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3, NSO cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
  • In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of an antibody molecule, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited to, the E. coli expression vector pUR278 (Ruther et al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Heeke & Schuster, J. Biol. Chem. 24:5503-5509 (1989)); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety. In an insect system, Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda cells. The antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • In other embodiments, the coating reagent is a ligand of the antibody, such as MAdCAM or an α4β7 integrin-binding fragment thereof or fusion protein comprising an α4β7-integrin binding fragment of MAdCAM fused with a non-MAdCAM protein, such as an immunoglobulin G constant domain. Examples of MAdCAM reagents and fusion proteins are described in PCT publication WO9624673 and U.S. Pat. No. 7,803,904, the entire teachings of which are incorporated herein by reference.
  • HAHA Assay
  • The human anti-anti-α4β7 antibody activity (HAHA) can be determined by detecting and/or measuring anti-drug antibodies (ADAs) or antibodies specific to the anti-α4β7 antibody (anti-vedolizumab antibodies). There are a number of options, for example, using a screening and titration assay, a confirmation assay, and a neutralizing assay. Serum samples can be measured first in the screening sample at dilutions, for example, 1:5 and 1:50. Positive samples can be confirmed for specificity, titered, and examined for the ability to neutralize anti-α4β7 antibody, e.g., vedolizumab activity.
  • A screening assay can use a bridging ELISA in which the plate is coated with the anti-α4β7 antibody. The immobilized anti-α4β7 antibody captures the ADA in the test sample which is bound by an anti-α4β7 antibody conjugated to biotin, which is tagged by horseradish peroxidase (HRP)-labeled streptavidin, then detected with an enzymatic substrate, such as TMB. A positive color development, e.g., as measured in a microplate reader, such as Spectramax, with analytical software, such as SOFTMAX Pro3.1.2, indicates the presence of ADAs in the sample. The assay cut point, e.g., in biotin-avidin-HRP based bridging assay, can be determined by using normal human serum samples as negative controls. The mean absorbance values of the 10 negative control serums can be added to 1.65 times the standard deviation of the negative controls to determine the cut point. Thus, the cut point can allow for approximately a 5% false positive rate. In the presence of 1 μg/mL vedolizumab, low titer responses are interfered with such that they may become undetectable, although high levels of immunogenicity are detectable at vedolizumab concentrations greater than 1 μg/mL. For example, while the standard assay sensitivity can be 0.44 ng/ml, in the presence of 0.5 μg/ml vedolizumab, the sensitivity of the assay can be 180 ng/ml. For these reasons, serum samples can be taken greater than 4 weeks, greater than 8 weeks, greater than 12 weeks or greater than 16 weeks after the final dose of anti-α4β7 antibody. With a longer time period between the prior dose and the sampling, serum drug levels typically can be below the interference level.
  • Another assay method uses streptavidin coated plates, biotin-labeled anti-α4β7 antibody anchored to streptavidin coated vessels, beads or microtiter plates for the immobilized side of the bridge and heavy metal, such as ruthenium, osmium or rhenium-labeled (e.g., via a sulfo tag) anti-α4β7 antibody for the other side of the bridge. The bridged complex can be built on the plate by stepwise additions and washes between or in solution, with both sides of the bridge contacting diluted serum sample, then transferred to the plate. An example of an assay using this method has a sensitivity of 3.90 ng/ml anti-anti-α4β7 antibody. Detection of the heavy metal labeled bridge complex, e.g., a ruthenium-labeled complex, by electrochemiluminescence (ECL), e.g., in a Meso Scale Discovery Sector Imager 6000 (Rockville, Md.), may be more sensitive than an HRP method and/or have higher tolerance to the amount of anti-α4β7 antibody in the serum. Thus there would not be a need to wait for a delayed sample after the serum drug level lowers. In some embodiments, pretreatment of the serum sample with acid, e.g., acetic acid or low pH glycine, to release the anti-α4β7 antibody from the patient-derived anti-anti-α4β7 antibodies prior to contacting with the bridging anti-α4β7 antibodies can reduce the interference from the drug in the serum. For example, while the standard assay sensitivity can be 3.90 ng/ml, in the presence of 5 μg/ml vedolizumab in serum, the sensitivity of the assay can be 10 ng/ml.
  • In an embodiment, an assay to detect anti-vedolizumab antibodies in a sample of serum from a patient comprises diluting serum by a standard dilution factor, such as 1:5, 1:25, 1:50, and/or 1:125; treating with acetic acid; combining the acid treated diluted sample with an assay composition comprising a high pH reagent, such as high concentration TRIS buffer for neutralizing the acid, a biotin-labeled vedolizumab and a ruthenium-labeled vedolizumab for a time sufficient to form a bridge with serum-derived anti-vedolizumab antibodies between the two tagged versions of vedolizumab; transferring the complexes to a streptavidin-coated plate; washing the plate so only ruthenium complexed by the antibody bridge is present. Detection of the bound ruthenium-labeled complex and measuring the sample by electrochemiluminescence in the microplate reader can be achieved by adding a read solution such as tripropylamine and applying voltage to stimulate the ruthenium label complexed to the plate via the antibody bridge.
  • After the initial screening assay, samples can be further tested in a confirmatory assay that uses excess unlabeled anti-α4β7 antibody to demonstrate specificity. Confirmed positive samples can be further assessed for the ability of the HAHA to neutralize the binding of the anti-α4β7 antibody, e.g., vedolizumab to cells. A competitive flow cytometry-based assay was designed to determine the ability of the immune serum to inhibit the binding of labeled vedolizumab to an α4β7 integrin-expressing cell line, RPMI8866, and detection by flow cytometry.
  • The results can indicate categories of immunogenicity status: Negative: no positive HAHA sample; Positive: at least 1 positive HAHA sample; Transiently positive: at least 1 positive HAHA sample and no consecutive positive HAHA samples; and Persistently positive: at least 2 or more consecutive positive HAHA samples. Negative patients are likely to respond to anti-α4β7 antibody and can continue being treated with the antibody. Persistently positive patients are likely to have high clearance of anti-α4β7 antibody and may not respond to anti-α4β7 antibody treatment. Positive patients may have high clearance of anti-α4β7 antibody and may not respond to anti-α4β7 antibody. Positive patients can have an additional serum sample 2, 3, 4, 5 or 6 weeks after another dose of anti-α4β7 antibody to determine if they are persistently positive or transiently positive. Transiently positive patients are likely to respond to anti-α4β7 antibody treatment and treatment of these patients can be continued.
  • Titers of immunogenicity levels also may be determined. Titer categories include ≧5 (low), ≧50, ≧125, ≧625 and ≧3125 (high). A patient with a high titer in a positive sample may have high clearance of anti-α4β7 antibody and may not respond to anti-α4β7 antibody treatment. A patient with a low titer in a positive sample may respond to anti-α4β7 antibody treatment.
  • The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention. All literature and patent citations are incorporated herein by reference.
  • EXEMPLIFICATION Example 1. Pharmacokinetic and Pharmacodynamic Modeling
  • Here, we report a comprehensive population pharmacokinetic and pharmacodynamic analysis of vedolizumab therapy in patients with UC and CD. Our objectives were to (1) characterize the pharmacokinetics of vedolizumab in patients who received repeated IV infusions of vedolizumab 300 mg for up to 52 weeks; (2) identify clinically relevant determinants of vedolizumab clearance in patients; and (3) describe the pharmacokinetic-pharmacodynamic relationship of vedolizumab in patients using MAdCAM-1 as the pharmacodynamic endpoint.
  • Materials and Methods Study Design and Sample Collection
  • Analyses were conducted using vedolizumab serum concentrations obtained from 5 randomized, placebo-controlled clinical studies: a phase 1 study in healthy volunteers, a phase 2 study in patients with active UC (NCT01177228), a phase 3 study in patients with moderately to severely active UC (GEMINI 1 [NCT00783718]), and 2 phase 3 studies in patients with moderately to severely active CD (GEMINI 2 [NCT00783692] and GEMINI 3 [NCT01224171]) (Table S1). The study designs and clinical data for the phase 2 and 3 studies have been previously reported. All study protocols and consent forms were approved by institutional review boards or ethics committees at the study sites, and studies were conducted in accordance with the principles of good clinical practice and the Declaration of Helinski. All patients provided written informed consent before study participation.
  • Details of the blood sampling times for pharmacokinetic, pharmacodynamic (MAdCAM-1), and anti-drug antibody (ADA) analyses are provided in Table S1. Extensive pharmacokinetic and pharmacodynamic sampling was used in the phase 1 and 2 studies, whereas sparse sampling was used in the phase 3 studies. Pharmacodynamic samples were not collected in GEMINI 3.
  • TABLE S1
    Clinical studies included in vedolizumab population pharmacokinetic/pharmacodynamic analyses
    Partipicants Blood Sampling
    Study Study Design/Study Dosing Enrolled/With Times for PK, PD,
    Identifier Population Regimen PK and ADA Analyses
    C13009 Phase
    1, single-dose Single dose Total = 87 PK: Day 1 (predose; 35
    study evaluating PK, VDZ VDZ = 62 min and 1, 2, and 12 h
    PD, safety, and 300 mg IV Placebo = 25 postdose).
    tolerability of VDZ (n = 13) PK and PD: Day 2 (24
    Part 1, open-label; Part VDZ h postdose), Day 8
    2, double-blind. 600 mg IV PK = 55 (168 h postdose), Day
    Healthy subjects (aged (n = 23) 29 (696 h postdose),
    18-45 years). VDZ Day 57 (1344 h
    600 mg IV postdose), Day 85
    (n = 26) (2016 h postdose), Day
    Placebo IV 113 (2688 postdose),
    (n = 25) Day 141 (3360
    postdose), Day 169
    (4032 postdose), Day
    197 (4704 h postdose).
    C13002 Phase 2, randomized, 4 doses Total = 47 PK and PD:# Day 1
    (NCT01177228) double-blind, placebo- ( days 1, 15, VDZ = 38* (predose; 2 and 12 h
    controlled, PK/PD, 29, and 85) Placebo = 9 postdose), Day 2 (24 h
    multiple-dose, VDZ postdose), Day 3 (48 h
    multicenter study. 2.0 mg/kg PK = 35 postdose), Day 4 (72 h
    Patients with mildly to IV (n = 13) postdose), Day 8 (at
    moderately active UC VDZ any time), Days 15 and
    (aged 18-70 years). 6.0 mg/kg 29 (predose; 2 h
    IV (n = 14) postdose), Days 43, 57,
    VDZ 71 (at any time), Day
    10.0 mg/kg 85 (predose; 2 and 12 h
    IV (n = 11) postdose), Day 86 (24
    Placebo IV h postdose), Day 87
    (n = 9) (48 h postdose), Day
    89 (96 h postdose), and
    at any time on Days
    92, 99, 113, 127,
    141, 155, 169, 183,
    197, 211, 225, 239, and
    253.
    ADA: Day 1, 15, 29,
    and 85 (predose) and
    Days 57, 113, 141,
    169, 197, 225, and 253
    GEMINI 1 Phase 3, 52-week, Multiple ITT: PK: Weeks 0, 2, 6, 22,
    (NCT00783718) randomized, placebo- dose VDZ = 225 and 46 (predose
    controlled, double- Induction Placebo = 149 [within 30 min before
    blind, multicenter study (Weeks 0 Non-ITT: start of infusion] and
    of efficacy and safety and 2): VDZ = 521 postdose [within 2 h
    of VDZ induction and VDZ after start of infusion]);
    maintenance therapy. 300 mg IV ITT: Weeks 14 and 38
    Patients with Placebo VDZ Q4W = (predose); Weeks 4 and
    moderately to severely 125 52 (anytime during
    active UC and Maintenance VDZ Q8W = visit).
    inadequate response to, (for 122 PD: Weeks 0 and 6
    loss of response to, or 44 weeks): Placebo = 126 (predose); Week 52.
    intolerance of 1 of VDZ Non-ITT: ADA: Weeks 0, 6, 14,
    more of the following 300 mg IV VDZ Q4W = 26, 38, 52 (or Early
    agents: corticosteroids Q4W 373 Termination visit), 66
    (ex-US only), VDZ Placebo = 149 (or Final Safety visit)
    immunomodulators, or 300 mg IV and at the time of
    TNF-α antagonists Q8W disease exacerbation.
    (aged 18-80 years). Placebo PK = 654 Samples were collected
    predose, if applicable.
    GEMINI 2 Phase 3, 52-week, Multiple ITT: PK: Weeks 0, 2, 6, 22,
    (NCT00783692) randomized, placebo- dose VDZ = 220 and 46 (predose
    controlled, double- Placebo = 148 [within 30 min before
    blind, multicenter study Induction Non-ITT: start of infusion] and
    of efficacy and safety (Weeks 0-2): VDZ = 747 postdose [within 2 h
    of VDZ induction and VDZ after start of infusion]);
    maintenance therapy. 300 mg IV ITT: Weeks 14 and 38
    Placebo VDZ Q4W = (predose); Weeks 4 and
    Patients with 154 52 (anytime during
    moderately to severely VDZ Q8W = visits).
    active CD and Maintenance 154 PD: Weeks 0 and 6
    demonstrated (for Placebo = 153 (predose); Week 52.
    inadequate response to, 44 weeks): Non-ITT: ADA: Weeks 0, 6, 14,
    loss of response to, or VDZ VDZ Q4W = 26, 38, 52 (or Early
    intolerance of 1 of 300 mg IV 506 Termination visit), 66
    more of the following Q4W (or Final Safety visit)
    agents: corticosteroids VDZ Placebo = 148 and at the time of
    (ex-US only), 300 mg IV disease exacerbation.
    immunomodulators, or Q8W Samples were collected
    TNF-α antagonists Placebo PK = 827 predose, if applicable.
    (aged 18-80 years).
    GEMINI 3 Phase 3, 10-week, Multiple ITT: PK: Week 0
    (NCT01224171) randomized, placebo- dose VDZ = 209 (postdose); Week 6
    controlled, double- Induction Placebo = 207 (predose and postdose
    blind, multicenter study (Weeks 0, 2 [within 2 h after start
    of efficacy and safety and 6) of infusion]); Week 10
    of VDZ induction VDZ TNF-α (anytime during visit)
    therapy. 300 mg IV antagonist or Early Termination.
    failure: ADA: Weeks 0, 6, 10
    Patients with VDZ = 158 (or Early Terminatio
    moderately to severely Placebo = 157 visit), and 22 (or Final
    active CD and Safety visit). Samples
    demonstrated were collected predose,
    inadequate response to, PK = 352 if applicable.
    loss of response to, or
    intolerance of 1 of
    more of the following
    agents: corticosteroids
    (ex-US only),
    immunomodulators, or
    TNF-α antagonists
    (aged 18-80 years).
  • Assays
  • Vedolizumab serum concentrations were determined using a sandwich enzyme-linked immunosorbent assay (ELISA), with a lower limit of detection of 0.00125 μg/mL at a 1:100 dilution. The accuracy of the assay was 10.1% coefficient of variation (CV), intra-sample precision ranged from 1.8% to 3.0% CV, and inter-sample precision ranged from 4.0% to 16.0% CV.
  • To quantitate α4β7 integrin saturation by vedolizumab in peripheral serum, a MAdCAM-1-Fc binding interference flow cytometry assay was developed. In this pharmacodynamic assay, inhibition of MAdCAM-1-Fc binding to α4β7-expressing peripheral blood cells by vedolizumab in the blood is used as a measure of the extent of α4β7 saturation by vedolizumab [1]. The assay, which was developed by Millennium Pharmaceuticals, Inc. (d/b/a Takeda Pharmaceuticals International, Co.), demonstrated an overall intra-sample variability of 6% CV and an intra-subject variability of 20% CV.
  • The presence of anti-drug antibodies (ADAs) or antibodies specific to the anti-α4β7 antibody (anti-vedolizumab antibodies) was determined using a validated, biotinylated, bridging ELISA and 2 dilutions of serum (1:5 and 1:50). The plate is coated with the anti-α4β7 antibody. The immobilized anti-α4β7 antibody captures the ADA in the test sample. After washing, the bound complex captures an anti-α4β7 antibody conjugated to biotin, which after another wash, captures HRP-labeled streptavidin. When incubated with TMB substrate, a positive color development indicates the presence of ADAs in the sample. All samples that screened positive were further diluted to determine the final ADA titer using standard techniques. If both screening dilutions were negative, the sample was considered negative. Patients were classified as positive for ADAs if antibodies were detected at any visit; otherwise they were classified as negative.
  • Data Assembly
  • The dosing, covariate, and pharmacokinetic-pharmacodynamic data were merged and formatted for the population analysis using R, Version 2.10.1 or higher (www.r-project.org). Vedolizumab serum concentration measurements that were missing, or any values with unknown or missing associated observation times, dose times, dose amounts, or dosing intervals, were excluded from the analysis. MAdCAM-1 measurements were treated similarly. All vedolizumab samples reported as below the limit of quantification (BLQ) (n=3189) were not evaluated during the population pharmacokinetic model development. More than half of the BLQ observations (n=1722) were samples obtained prior to the first vedolizumab dose.
  • Covariates present in the population pharmacokinetic dataset were serum C-reactive protein (CRP), serum albumin, fecal calprotectin, body weight, disease activity (Crohn's Disease Activity Index [CDAI], complete Mayo score, partial Mayo score), Mayo endoscopic subscore, age, sex, ADA status (positive or negative), prior TNF-α antagonist therapy status (naïve or failed), body mass index (BMI), serum globulin, diagnosis (CD or UC), lymphocyte count, and concomitant therapy use (methotrexate, azathioprine, mercaptopurine, or aminosalicylates). The start date and end date of concomitant therapy was populated in the dataset to evaluate the time-dependent effects of concomitant treatments. Covariates with missing data were imputed using different imputation methods, based on the remaining available data (e.g. median of the remaining values). No covariates present in the population pharmacokinetic dataset were missing more than 10% of values.
  • Population Pharmacokinetic Model Development
  • The population pharmacokinetic analysis for repeated measures was conducted using a nonlinear mixed effects modeling approach (NONMEM 7, Version 7.2; ICON Development Solutions, Ellicott City, Md., USA) [13]. The base population pharmacokinetic model was developed using the first-order conditional estimation with the η-ε interaction (FOCEI) method and extensively sampled phase 1 and 2 data. Results from the base model were subsequently used as prior information to selectively inform a subset of population pharmacokinetic model parameters in the full covariate model, which was fit to sparse phase 3 data from GEMINI 1, 2, and 3. The full covariate model was fit to the phase 3 data using the full Bayesian Markov Chain Monte Carlo (MCMC) method. All parameter estimates were reported with Bayesian 95% credible intervals (CDIs) as a measure of estimation uncertainty. Population parameters, including fixed effects parameters (covariate coefficients and structural model parameters) and random effects parameters, were estimated. Interindividual random effect distributions were modeled using exponential variance models for linear clearance (CLL), central compartment volume of distribution (Vc), and maximum elimination rate (Vmax), with a full block covariance matrix, whereas residual random effects were described with a proportional model.
  • A covariate modeling approach emphasizing parameter estimation rather than stepwise hypothesis testing was implemented for the population pharmacokinetic analysis [14]. First, predefined covariate parameter relationships were identified based on exploratory graphics, scientific interest, and mechanistic plausibility. Then a full covariate model was constructed with care to avoid correlation or collinearity in predictors; covariates with correlation coefficients greater than −0.35 were not simultaneously included as potential predictors. Construction of the full model was also guided by evaluating the adequacy of the study design and covariate data to support quantification of the covariate effects of interest.
  • Inferences about the clinical relevance of parameters were based on the resulting parameter estimates and measures of estimation precision (Bayesian 95% CDIs) from the full model. In the absence of an exposure-response relationship for efficacy-related clinical endpoints to provide a context for interpretation of pharmacokinetic variability, covariate effect sizes greater than ±25% of the typical population value were proposed as clinically meaningful changes.
  • Modeling assumptions included log transformation of pharmacokinetic/pharmacodynamic (where appropriate) parameters. In those cases where no physiologic relationship was known a priori, the effects of continuous covariates were modeled using an additive model in the log domain, while the effects of categorical covariates were similarly described:
  • TPV = exp ( θ n + l m log ( cov m i ref m ) · θ ( m + n ) + l p θ ( p + m + n ) cov pi )
  • where the typical value of a model parameter (TPV) is described as a function of m individual continuous covariates (covmi) and p individual categorical covariates categories; θn is an estimated parameter describing the typical log-transformed pharmacokinetic parameter value for an individual with covariates equal to the reference covariate values (covmi=refm); and θ(m+n) and θ(p+m+n) are estimated parameters describing the magnitude of the covariate-parameter relationship.
  • The final population pharmacokinetic model and parameter estimates were investigated with a predictive check method. This method is similar to the posterior predictive check, but assumes that parameter uncertainty is negligible relative to interindividual and residual variance. Five hundred Monte Carlo simulation replicates of the original dataset were compared with the distribution of the same exposure metric in the observed vedolizumab dataset, using quantile-quantile plots.
  • Population Pharmacokinetic-Pharmacodynamic Model Development
  • The population pharmacokinetic-pharmacodynamic analysis for repeated measures was conducted using the nonlinear mixed effects modeling approach (NONMEM, Version 7.2). The pharmacokinetic-pharmacodynamic data were modeled using a sequential approach, where individual predicted vedolizumab serum concentrations from the population pharmacokinetic model were used to drive the pharmacodynamic response. The pharmacodynamic evaluations were based on percentage of MAdCAM-1 binding by lymphocytes expressing high levels of α4β7 integrin (CD4+ CD45ROhigh). A direct effect sigmoid Emax model was selected to characterize the exposure-response relationship for the effect of vedolizumab on MAdCAM-1 binding to α4β7. No formal covariate modeling or model evaluation was conducted.
  • Results Pharmacokinetic Analysis Population
  • The study population consisted of 2554 individuals who contributed 18427 evaluable vedolizumab serum samples, including 87 healthy volunteers from the phase 1 study, 46 patients from the phase 2 study (UC), and 891, 1115, and 415 patients from the phase 3 GEMINI 1 (UC), GEMINI 2 (CD), and GEMINI 3 (CD) studies, respectively.
  • Demographics and other characteristics of the pharmacokinetic analysis population are summarized in Table 1. The analysis population consisted of 1290 men and 1264 women with ages ranging from 18 to 78 years and baseline body weights ranging from 28 to 170 kg. A total of 1530 individuals had CD and 937 had UC; 87 were healthy volunteers.
  • TABLE 1
    Summary of demographics and other characteristics of
    the pharmacokinetic analysis population (N = 2554)
    Categorical Covariate n (%)
    Sex
    Women 1264 (50)
    Men 1290 (50)
    Disease diagnosis
    Crohn's disease 1530 (60)
    Ulcerative colitis 937 (37)
    Healthy volunteers 87 (3)
    Mayo Endoscopic Subscore
    1 1 (0.039)
    2 408 (16)
    3 482 (19)
    Missing* 1663 (65)
    Prior TNF-α antagonist therapy status
    Failed 1321 (52)
    Naïve 1100 (43)
    Missing# 133 (5)
    ADA status
    Positive (≧1 positive titer) 124 (5)
    Negative (no positive titers) 2430 (95)
    Continuous Covariate n Median (Range)
    Age, years 2554 36 (18, 78)
    Body weight, kg 2554 68 (28, 170)
    Albumin, g/L 2467 37 (11, 53)
    C-reactive protein, mg/L 1576 11 (0.2, 200)
    Fecal calprotectin, mg/kg 2421 720 (23.75, 20000)
    CDAI score 1530 320 (93, 580)
    Mayo Score 891 9 (3, 12)
    Partial Mayo Score 937 6 (1, 9)
    ADA, anti-drug antibody; CDAI, Crohn's Disease Activity Index; TNF-α, tumor necrosis factor-α.
    *Data not collected in phase 1 and 2 studies and in GEMINI 2 and 3 studies.
    #Data not collected in phase 1 and 2 studies.
  • The median (interquartile range) vedolizumab serum trough concentration-time profiles for patients with UC from GEMINI 1 and patients with CD from GEMINI 2 are shown in FIG. 1.
  • Population Pharmacokinetic Modeling Results Base Pharmacokinetic Model
  • Vedolizumab pharmacokinetics was described by a 2-compartment model with parallel linear and nonlinear elimination. A 2-compartment model resulted in a significant improvement in goodness-of-fit criteria over a 1-compartment model, as did a parallel linear and nonlinear elimination model over a linear model. The population pharmacokinetic model of vedolizumab is represented diagrammatically in FIG. 2.
  • Covariate Pharmacokinetic Model
  • Strong correlations were identified between the following covariates:body weight˜BMI, sex˜body weight, CRP˜albumin, CRP˜fecal calprotectin, CRP˜globulin, albuming globulin, complete Mayo score˜partial Mayo score, Mayo endoscopic subscore˜complete Mayo score, and Mayo endoscopic subscore˜partial Mayo score. Accordingly, sex, CRP, complete Mayo score, Mayo endoscopic subscore, globulin, and BMI were excluded from the full covariate model. The effects of sex, CRP, and endoscopic subscore on the pharmacokinetics of vedolizumab were independently evaluated in an exploratory post hoc fashion once the population pharmacokinetic model was finalized, as described below.
  • Body weight was chosen to represent changes in vedolizumab pharmacokinetics as a function of body size and was described using an allometric model with a reference weight of 70 kg. The other continuous covariates of albumin, fecal calprotectin, partial Mayo score, age, and CDAI score entered the model as power functions normalized by a reference value (typically near the observed median value of the data). The categorical covariates of prior TNF-α antagonist therapy status, ADA status, concomitant therapy use, and IBD diagnosis entered the model as power functions, with a separate dichotomous (0, 1) covariate serving as an on-off switch for each effect. Time-dependent covariates were body weight, albumin, fecal calprotectin, and concomitant therapy use. The effect of IBD diagnosis on linear clearance (CLL) was investigated by modeling separate CLL parameters for patients with UC and those with CD, while the effect of IBD diagnosis on central compartment volume of distribution (Vc) was evaluated by including IBD as a predictor of Vc in the covariate model.
  • Final Pharmacokinetic Model Results
  • The typical pharmacokinetic parameter and variance parameter estimates and 95% CDIs from the final population pharmacokinetic model are shown in Table 2 for the reference covariate values (listed in Table 2 footnotes).
  • TABLE 2
    Parameter estimates from the final population
    pharmacokinetic model for vedolizumab
    Bayesian 95%
    Parameter Estimate* CDI
    Ulcerative colitis: CLL 0.159 L/day (0.153, 0.165)
    Crohn's Disease: CLL 0.155 L/day (0.149, 0.161)
    Central compartment volume 3.19 L (3.14, 3.25)
    of distribution (Vc)
    Peripheral compartment volume 1.65 L (1.59, 1.71)
    of distribution (Vp)
    Intercompartmental clearance (Q) 0.12 L/day (0.112, 0.129)
    Maximum elimination rate (Vmax) 0.265 mg/day (0.219, 0.318)
    Concentration at half-maximum 0.964 μg/mL (0.706, 1.27) 
    elimination rate (Km)
    Proportional residual 0.0554 (0.0539, 0.0568)
    error variance (σ2 prop) (% CV = 23.5)
    CDI, credible interval; CLL, clearance of linear elimination pathway; CV, coefficient of variation.
    *Parameter estimate and 95% credible interval were derived from the median, 2.5th and 97.5th quantiles of the Bayesian posterior probability distributions from 4 MCMC chains. Separate typical values of CLL were modeled for patients with UC and those with CD with a shared inter-individual variance term and shared covariate effects except for partial Mayo score and CDAI score. The reference individual weighs 70 kg, is 40 years old, has an albumin level of 4 g/dL, fecal calprotectin level of 700 mg/kg, CDAI score of 300 (for patient with CD), partial Mayo score of 6 (for patient with UC), ADA negative, a diagnosis of UC (for Vc parameter), and no concomitant therapy use, and is TNF-α antagonist therapy naïve.
  • The pharmacokinetic parameter estimates and overlapping 95% CDIs indicated that CLL were the same in patients with UC and those with CD. The half-life of vedolizumab for the linear elimination phase was 25.5 days for the typical reference patient and individual estimates ranged from 14.6 to 36.0 days (5th and 95th percentiles). The variance parameter estimates were indicative of moderate to large unexplained interindividual variability. The typical values of CLL were 0.159 L/day for patients with UC and 0.155 L/day for patients with CD. The individual estimates of CLL for patients with UC and CD were distributed over a wide range as represented in FIG. 3. The proportional residual variance estimate (unexplained random residual variability in the model) was relatively small. The estimates of interindividual variances and correlations are presented in Table S2, and the standard deviations and shrinkage estimates of interindividual random effects are presented in Table S3.
  • TABLE S2
    Estimates of interindividual variances (% CV) and correlations
    from the vedolizumab final population pharmacokinetic model
    Bayesian
    95%
    Credible
    Parameter Estimate* Interval*
    ω2 CLL (% CV) 34.6 (33.3, 36.1)
    corrCLL-Vc 0.566 (0.515, 0.615)
    ω2 Vc (% CV) 19.1 (18.2, 20.1)
    corrCLL-Vmax −0.192  (−0.285, −0.0945)
    corrVc-Vmax −0.267 (−0.378, −0.151)
    ω2 Vmax (% CV) 105 (94.5, 117) 
    ω2 Vp (% CV) 0 Fixed
    ω2 Q (% CV) 0 Fixed
    ω2 Km (% CV) 0 Fixed
    CLL, clearance of linear elimination pathway; corr, correlation coefficient; CV, coefficient of variation; Km, concentration at half-maximum elimination rate; Q, intercompartmental clearance; Vc, central compartment volume of distribution; Vmax, maximum elimination rate; Vp, peripheral compartment volume of distribution; ω2, interindividual variance.
    *Parameter estimate and 95% credible interval were derived from the median, 2.5th and 97.5th quantiles of the Bayesian posterior probability distributions from 4 MCMC chains. Separate typical values of CLL were modeled for patients with UC and those with CD with a shared interindividual variance term and shared covariate effects except for partial Mayo score and CDAI score.
  • TABLE S3
    Standard deviations and shrinkage estimates of
    interindividual random effects from the vedolizumab
    final population pharmacokinetic model
    Parameter SD({tilde over (η)})* ω# ηshrink (%)
    ETACLL 0.329 0.346 4.85
    ETAVc 0.156 0.191 18.5
    ETAVmax 0.542 1.05 48.5
    CLL, clearance of linear elimination pathway; ETA, interindividual random effect; SD({tilde over (η)}), standard deviation of individual estimates; Vc, central compartment volume of distribution; Vmax, maximum elimination rate; ω, standard deviation of variance estimate; ηshrink (%) = 100 · (1 - SD(η)/ω).
    *Individual estimate used for the calculation SD(η) was derived as the median of the individual's random effect estimates from 4 MCMC chains.
    #ω estimate was derived from the median of the Bayesian posterior probability distributions from 4 MCMC chains.
  • Goodness-of-fit plots from the final population pharmacokinetic model are presented in FIGS. 4 and 9. Diagnostic plots indicated that the full covariate pharmacokinetic model was consistent with the observed data and no systematic bias was evident. The relative changes in CLL for a reference individual with various covariate values are illustrated in FIG. 5. The point estimates and 95% CDIs for the effects of covariates on CLL are presented in Table S4.
  • TABLE S4
    Covariate parameter estimates from the vedolizumab
    final population pharmacokinetic model
    Bayesian 95%
    Credible
    Parameter Estimate* Interval*
    Continuous covariates effects (NULL effect = 0)
    Weight on CLL 0.362 (0.299, 0.427)
    Albumin on CLL −1.18 (−1.24, −1.13)
    Fecal calprotectin on CLL 0.0310 (0.0252, 0.0358)
    CDAI score on CLL −0.0515  (−0.14, 0.0358)
    Partial Mayo score on CLL 0.0408 (−0.0336, 0.115) 
    Age on CLL −0.0346 (−0.0788, 0.01)   
    Weight on Vc 0.467 (0.423, 0.511)
    Weight on Vp   1 Fixed (1, 1)
    Weight on Vmax 0.75 Fixed (0.75, 0.75)
    Weight on Q 0.75 Fixed (0.75, 0.72)
    Categorical covariates effects (NULL effect = 1)
    Prior TNF-α antagonist therapy 1.04 (1.01, 1.07)
    status on CLL
    ADA status on CLL 1.12 (1.05, 1.2) 
    AZA on CLL 0.998 (0.964, 1.03) 
    MP on CLL 1.04 (0.943, 1.15) 
    MTX on CLL 0.983 (0.899, 1.07) 
    AMINO on CLL 1.02 (0.988, 1.05) 
    IBD diagnosis on Vc 1.01 (0.989, 1.03) 
    AMINO, aminosalicylate concomitant therapy; AZA, azathioprine concomitant therapy; CD, Crohn's Disease; CDAI, Crohn's disease activity index; CLL, clearance of linear elimination pathway; ADA, anti-drug antibody; MP, 6-mercaptopurine concomitant therapy; MTX, methotrexate concomitant therapy; Q, intercompartmental clearance; TNF-α, tumor necrosis factor-α; UC, ulcerative colitis; Vc, central compartment volume of distribution; Vmax, maximum elimination rate; Vp, peripheral compartment volume of distribution.
    *Parameter estimate and 95% credible interval were derived from the median, 2.5th and 97.5th quantiles of the Bayesian posterior probability distributions from 4 MCMC chains. Separate typical values of CLL were modeled for patients with UC and those with CD with a shared interindividual variance term and shared covariate effects except for partial Mayo score and CDAI score. The reference individual weighs 70 kg, is 40 years old, has an albumin level of 4 g/dL, fecal calprotectin level of 700 mg/kg, CDAI score of 300 (for patient with CD), partial Mayo score of 6 (for patient with UC), ADA negative, a diagnosis of UC (for Vc parameter), and no concomitant therapy use, and is TNF-α antagonist therapy naïve
  • In general, the 95% CDIs were narrow indicating that the effect of each covariate on vedolizumab CLL was well defined. Only the effects of albumin and body weight had potential to be clinically meaningful (effect sizes greater than 25%) at extremes values of a representative range. The typical CLL values for patients with albumin levels of 4.7 and 3.2 g/dL were approximately 0.8 and 1.3 times, respectively, that of the reference patient (albumin, 4 g/dL) (FIG. 5). The typical CLL value for a patient of 40 kg was approximately 0.8 times that of the reference patient (weight, 70 kg). At the other end of the body weight range, a patient of 120 kg had 1.2 times higher CLL than the reference patient (FIG. 5). A patient of 120 kg with a serum albumin concentration of 4.0 g/dL had a 19% probability of having clearance greater than the pre-specified criterion for clinical significance. When evaluated across a representative range of covariate values and categories, the effects of fecal calprotectin, CDAI score, partial Mayo score, age, prior TNF-α antagonist therapy status, ADA status, and concomitant therapy use on vedolizumab CLL were not considered clinically relevant since the covariate effect sizes were less than ±25% from the typical reference values (Table S4). In addition, the 95% CDIs for these covariate effects were generally narrow and contained the null effect value.
  • The final population pharmacokinetic model was rerun with all covariates and pharmacokinetic parameters fixed to estimates from the final model (interindividual variances were re-estimated), and any remaining effects of sex on CLL and Vc were quantified. The results of this analysis suggest that, after adjusting for other predictors of vedolizumab pharmacokinetics, the typical CLL value and Vc are approximately 10% lower and 6% lower, respectively, for a female patient compared with a male patient. However, these effects were not considered clinically relevant since the covariate effect sizes were less than ±25% from the typical reference values (male patient). Addition of the sex effect explained approximately 4.2% and 6.0% of the unexplained interindividual variability in CLL and Vc, respectively.
  • The final population pharmacokinetic model also was re-run to estimate any remaining effect of CRP on CLL. The results suggest that, after adjusting for other predictors of vedolizumab pharmacokinetics, the effect of CRP on CLL was not clinically relevant since the covariate effect size was less than ±25% from the typical reference value (CRP, 11 mg/dL) when evaluated over a range of baseline CRP values of 0.46 to 93 mg/dL (observed 2.5th and 97.5th percentiles). The addition of the CRP effect explained <1% of the unexplained interindividual variability in CLL.
  • For patients with UC, the effect of Mayo endoscopic subscore was evaluated graphically by plotting individual estimates of CLL from the final population pharmacokinetic model by endoscopic subscores at week 6 (FIG. 6). From this analysis, at week 6 (end of induction treatment), patients with an endoscopic subscore of 3 have on average 25% higher CLL than patients with an endoscopic subscore of 0.
  • Pharmacokinetic Model Evaluation
  • The final population pharmacokinetic model and parameter estimates were evaluated with a predictive check method and Bayesian 95% CDIs derived from the posterior probability distributions. The basic premise of a predictive check is that a model and parameters derived from an observed dataset should produce simulated data that are similar to the original observed data. The predictive check plots demonstrated overall good agreement between the observed and simulated data (FIGS. 10, 11, and 12). The precision of the parameters estimates was assessed by evaluating the Bayesian 95% CDIs (Tables 2, S2, and S4). Overall, the structural pharmacokinetic model parameters, covariates effects, and variance parameters were estimated with good precision.
  • Pharmacokinetic-Pharmacodynamic Analysis Population
  • The vedolizumab population pharmacokinetic-pharmacodynamic dataset was composed of 593 individuals contributing a total of 2442 evaluable MAdCAM-1 observations. The analysis population consisted of 297 patients with UC and 296 patients with CD (from the phase 2 study and GEMINI 1 and 2).
  • During the analysis, the log-transformed values of MAdCAM-1 (free α4β7 receptors not blocked by vedolizumab) were modeled. A plot of observed MAdCAM-1 measurements (percentage of free α4β7 receptors) versus observed vedolizumab serum concentrations for patients with UC and CD from GEMINI 1 and 2 is presented in FIG. 7. Plots of observed MAdCAM-1 measurements versus time by treatment regimen for patients with UC and CD in GEMINI 1 and 2 are presented in FIG. 8. The percentage of free α4β7 receptors declined rapidly after the first dose and was maintained during repeated IV infusions of vedolizumab 300 mg.
  • Pharmacokinetic-Pharmacodynamic Modeling Results
  • A direct effect sigmoid Emax model was chosen as the structural model to describe the pharmacokinetic-pharmacodynamic relationship of vedolizumab as follows:
  • MAdCAM - 1 = E 0 * ( 1 - E max * Conc γ EC 50 + Conc γ )
  • where Eo is the baseline MAdCAM-1 percent binding, Emax is the maximum effect, Conc is the vedolizumab serum concentration, EC50 is the vedolizumab serum concentration at half-maximum effect, and γ is the Hill-coefficient or slope factor.
  • The model was parameterized in terms of baseline MAdCAM-1 inhibition (E0), maximum effect (Emax), vedolizumab serum concentration at half-maximum effect (EC50), and Hill-coefficient or slope factor (γ). The structural parameter estimates are presented in Table 3 and were estimated with adequate precision. An attempt was made to model a placebo effect, but the estimated effect was negligible. The lack of an apparent effect was consistent with the observed MAdCAM-1 data in placebo-treated patients.
  • TABLE 3
    Parameter estimates from base MAdCAM-1 population pharmacokinetic-
    pharmacodynamic model for vedolizumab
    Percent Relative
    Parameter Estimate Standard Error
    Baseline MAdCAM-1 12.1% 3.49
    inhibition (E0)
    Concentration at half 0.093 μg/mL 25.8
    maximum effect (EC50)
    Maximum effect (Emax) 0.959 0.503
    Hill-coefficient or 0.801 11.1
    slope factor (γ)
    Exponential residual 0.613 (% CV = 78.3) 10.4
    error variance (σ2 exp)
    CV, coefficient of variation.
  • The base model provided a reasonable description of the data as judged by visual inspection of diagnostic plots (FIG. 13) but some deficiencies in the model were noted. Variance parameter estimates were indicative of moderate to large unexplained interindividual and residual variability, with estimates of 41.8% CV for E0, 0.551 (SD logistic distribution) for Emax, and 78.3% CV for the exponential residual error variance (σ2 exp) (Table 3).
  • Discussion
  • We developed a population pharmacokinetic model for vedolizumab following IV administration to healthy volunteers and patients with UC and CD using data collected in clinical trials with identical designs and sampling schedules. These design features allowed the direct comparison of the disposition and the pharmacokinetic variability of vedolizumab in patients with UC and those in patients with CD. The estimated half-life of vedolizumab was not different between the 2 diseases and was 25.5 days (range 14.6-36.0 days [5th and 95th percentiles]), which is typical of human IgG1 (25 days) and typical of monoclonal antibodies of the IgG1 type A2. This half-life is substantially longer than values for current biologic treatments for IBD. Clinicians should be aware of the relatively long half-life of vedolizumab in circumstances when it is desirable to minimize exposure to the drug. Relevant examples include patients who develop an enteric infection during treatment and women who become pregnant and receive drug within the third trimester.
  • A 2-compartment pharmacokinetic model consisting of parallel clearance via the nonlinear pathway (CLNL) and the linear pathway (CLL) from the central compartment was selected as the base model. The nonlinear elimination was best described by Michaelis-Menten elimination. Physiologically, the nonlinear pathway is thought to be due to clearance by saturable, target-mediated mechanisms such as receptor-mediated endocytosis. In contrast, the linear pathway represents components that are non-saturable at therapeutic concentrations, such as Fc-mediated elimination. Parallel elimination is typical of monoclonal antibodies with disposition that is affected by binding to the target, in this case the α4β7 integrin on circulating T lymphocytes. Similar effects have been reported for efalizumab, tocilizumab, and cetuximab. In contrast, the elimination of TNF-α antagonists is best described by a linear elimination process. After IV administration, the elimination of infliximab in patients with UC or ankylosing spondylitis and of golimumab in patients with rheumatoid arthritis follows a 2-compartment model with linear elimination. TNF-α exists in both soluble and membrane-bound forms and is present in abnormally high concentrations in serum and gut mucosa in patients with IBD. The localization of TNF-α in inflammatory tissues may make it difficult to rapidly achieve target saturation because of slow redistribution of the drug from plasma to the target sites. Accordingly, it may be difficult to achieve target saturation at therapeutic concentrations. Furthermore, variability in TNF-α concentrations in different compartments results in drug redistribution, with possible effects on pharmacokinetic and pharmacokinetic-efficacy/safety relationships of TNF-α antagonists. The clinical implications of these differences in elimination pathways are not currently understood.
  • The estimated vedolizumab CLL values are consistent with those of other monoclonal antibodies that are administered intravenously. Although some authors have reported that the clearance of antibodies could be affected by IBD type, no apparent differences were observed in the pharmacokinetics of vedolizumab in patients with UC and those with CD. Results from the final pharmacokinetic model showed that vedolizumab CLL for a 40-year-old, 70-kg patient with a serum albumin concentration of 4 g/dL was 0.159 L/day for a patient with UC and 0.155 L/day for a patient with CD.
  • We evaluated the potential effects of intrinsic and extrinsic covariates (body weight, age, albumin, fecal calprotectin, CDAI score, partial Mayo score, concomitant therapy use, AVA status, and prior TNF-α antagonist therapy status) on vedolizumab CLL. Extreme values of albumin and weight had important effects on drug clearance. Specifically, clearance increased as albumin concentrations decreased and weight increased. Albumin concentrations below 3.2 g/dL, for a patient of 70 kg, were associated with increased clearance of vedolizumab that was greater than the pre-specified criterion for clinical significance. Similar association of low albumin concentrations with the clearance of other monoclonal antibodies have been reported in population pharmacokinetics analyses. Although the mechanism of this interaction is incompletely understood, the Fc receptor (FcRn), which is expressed in a wide variety of cells and tissues throughout the body, including the vascular endothelium, monocytes, macrophages, dendritic cells, hepatocytes, and epithelial cells of the intestine, renal proximal convoluted tubules, and upper airways, facilitates IgG and albumin homeostasis by salvaging these molecules from proteolysis and recycling them into the central circulatory system. It has been postulated that decreased expression and/or activity of FcRn results in low albumin concentrations as a result of less efficient salvage of IgGs, including therapeutic antibodies, and albumin; the net result is increased antibody clearance and in this case lower serum vedolizumab concentrations. However, this hypothesis has not been proven. Another explanation might be that inflammation of the digestive tract in patient with IBD can result in an unconventional route of elimination. Specifically, in the setting of severe colitis, patients may develop a protein-losing colopathy in which large amounts of protein are lost from the luminal surface. Albumin may therefore serve as a surrogate marker for loss of endogenous IgG and monoclonal antibodies via this pathway. Support for this hypothesis comes from observations in patients with severe colitis treated with infliximab in which high concentrations of the monoclonal antibody were identified in the stool. Conversely, serum drug concentrations were low in these patients. Successful treatment was associated with resolution of this phenomenon. Vedolizumab concentrations in feces were not measured during the GEMINI trials precluding us from evaluating this hypothesis.
  • The second important covariate identified was body weight, which was positively correlated with the clearance of vedolizumab. A patient of 120 kg with a serum albumin concentration of 4.0 g/dL had a 19% probability of having clearance greater than the pre-specified criterion for clinical significance. Measures of body size are the most commonly identified covariates influencing the pharmacokinetics of therapeutic monoclonal antibodies. The impact of body weight on vedolizumab pharmacokinetics is consistent with that reported in population analyses of other therapeutic monoclonal antibodies.
  • Other potential covariates had lesser effects on vedolizumab clearance than weight and albumin. Intensity of inflammation has been identified as a predictor of high drug clearance for other monoclonal antibodies. In our analyses, patients with UC with lower Mayo endoscopic subscores had on average lower clearance than patients with higher endoscopic subscores. However, these results should be interpreted with caution since higher TNF-α antagonist trough levels have been reported in patients with UC and CD with mucosal healing, which raises the possibility that this relationship is not causal and merely reflects the association between drug-losing enteropathy, mucosal healing, and albumin concentrations.
  • Intensity of inflammation has been reported as a positive predictor of clearance for other monoclonal antibodies. A post hoc exploratory analysis estimated the effect of CRP as a predictor of unexplained random variability in vedolizumab clearance after accounting for effects of other covariates present in the existing final population pharmacokinetic model (such as albumin). These results suggest that the effect of CRP on CLL is not clinically relevant and explained less than 1% of the unexplained interindividual variability in vedolizumab CLL. In contrast, high CRP concentrations are strongly associated with increased clearance of anti-TNF-α monoclonal antibodies. Therefore, as albumin and CRP were strongly correlated, any potential effect of CRP on vedolizumab CLL was already accounted for in the model by incorporating albumin.
  • The development of ADAs has been reported to increase infliximab clearance. In the current analysis, the presence of ADAs was estimated to increase vedolizumab clearance by only 12%. The small impact may be a result of the low incidence of ADAs observed in the GEMINI trials. However, in the few patients who were persistently positive for ADAs in these studies, vedolizumab trough concentrations were below the limit of quantification. We believe that sensitization of patients to monoclonal antibodies is an important cause of treatment failure and that vedolizumab is not unique in this regard.
  • The current population pharmacokinetic analysis showed no clinically meaningful impact of the following covariates on the pharmacokinetics of vedolizumab: disease activity, prior TNF-α antagonist therapy status, age (from 18 to 78 years old), and concomitant medication use. The lack of association between prior TNF-α antagonist therapy use was associated with a higher probability of clinical remission or response in vedolizumab-treated patients with UC and those with CD. Taken together, these observations suggest that the impact of prior TNF-α antagonist therapy use on vedolizumab efficacy is not related to any effect on vedolizumab CLL.
  • The lack of an effect of thiopurines and methotrexate on the pharmacokinetics of vedolizumab differs from TNF-α antagonists in both IBD and rheumatoid arthritis patients where co-administration of these agents is associated with higher trough concentrations and lower drug clearance. The mechanism by which antimetabolites increase concentrations of biologic drugs is not well understood, however, modulation of Fcγ expression is one possible explanation. For example, methotrexate is known to down-regulate Fcγ receptors on monocytes and other Fc-receptor subtypes. Furthermore, prevention of the ADA development increases drug exposure by reducing immune-mediated drug clearance. The reason behind the lack of effect of concomitant medications on vedolizumab CLL is not currently understood. A sensitivity analysis was performed to determine whether the rate of concomitant medication use in the vedolizumab population pharmacokinetic data set was sufficient to achieve at least 80% power to detect no drug interaction, as recommended by the Population Pharmacokinetic Therapeutic Protein-Drug Interaction (PK TPDI) Working Group. This analysis revealed that the data set met the sample size requirements to ensure at least 80% power and confirmed that vedolizumab CLL was not impacted by concomitant use of azathioprine, mercaptopurine, methotrexate or aminosalicylates.
  • Interestingly, α4β7 receptor saturation was maintained at vedolizumab concentrations considered subtherapeutic, raising the question as to whether receptor saturation is necessary but not sufficient for clinical efficacy. The EC50 estimate from population pharmacokinetic-pharmacodynamic model was 0.093 μg/mL, suggesting that full saturation is reached at vedolizumab serum concentration of approximately 1 μg/mL. The exposure-efficacy results indicated that vedolizumab concentrations below 17 and 15 mg/mL at induction were associated with efficacy similar to placebo in patients with UC and CD, respectively. This discrepancy might be explained by the fact that MAdCAM-1 assay measures α4β7 saturation in circulating T-cells or may be due to a slow onset of action of the drug. The MAdCAM-1 assay is insensitive to dose and should not be used for dose selection. Further studies to evaluate the relative pharmacodynamic contributions of α4β7 receptor blockade in peripheral blood in distinction to effects on cell-cell interactions in the tissue compartment are a research priority.
  • In conclusion, a population model characterizing the pharmacokinetic-pharmacodynamic properties of vedolizumab was successfully developed for patients with UC and CD. The modeling results suggested that the typical pharmacokinetics of vedolizumab was similar in patients with UC and those with CD. Albumin and body weight were identified as important predictors of vedolizumab clearance, but the effects of these covariates were only considered clinically meaningful at extreme values. Surprisingly, co-administration of immunosuppressants had no clinically relevant impact on the pharmacokinetics of vedolizumab. However, this finding contrasts with the well-established relationship between prevention of immunogenicity and reduced drug concentrations for TNF-α antagonists. This analysis supports use of fixed dosing with vedolizumab in patients with UC and those with CD.
  • Example 2. Fecal Calprotectin Assessment
  • Fecal levels of calprotectin, a neutrophil cytostolic protein, correlate with endoscopic severity of inflammation in inflammatory bowel disease: <50 μg/g: Typical of non-diseased population; >50 to <150 μg/g: Possible mucosal inflammation; >150 μg/g: Active inflammation; Higher levels associated with greater risk of relapse. The aim is to determine if induction treatment with VDZ reduces fecal calprotectin levels in patients with moderately to severely active UC.
  • Patients from the blinded induction portion of the GEMINI 1 phase 3 clinical trial were tested for fCal in the stool and examined for response or remission relative to calprotectin levels. Stool samples were taken at weeks 0 and 6. fCal levels were measured using the PhiCal calprotectin kit (assay sensitivity of 9 ng/mL of diluted sample).
  • The Percent of Baseline Calprotectin at Week 6 by Treatment or Response Status is shown in FIG. 14. The P-value is from Wilcoxon rank sum test (2-sided) based on log-transformed data. Only patients with both baseline and week 6 calprotectin values are included in this analysis. A 69% and 32% decrease in geometric mean of fCal was observed for VDZ and PBO-treated patients, respectively (P=0.0011).
  • TABLE 4
    Clinical Response and Remission at Week
    6 by Baseline Starting fCal Category
    Baseline
    Categorical Response, n (%) Remission, n (%)
    Division PBO VDZ PBO VDZ
    ≦250 μg/g  8 (29.6) 20 (54.1) 2 (7.4) 7 (18.9)
     >250 μg/g* 28 (25)   82 (46.6) 6 (5.4) 29 (16.5) 
    ≦500 μg/g** 13 (27.7) 27 (47.4) 2 (4.3) 9 (15.8)
     >500 μg/g 23 (25.0) 75 (48.1) 6 (6.5) 27 (17.3) 
    *Includes >500 μg/g population.
    **Includes ≦250 μg/g population.
  • At baseline, mean fCAL levels were 2370 and 2552 μg/g in PBO and VDZ-treated patients, respectively.
  • FCAL concentrations declined in patients who responded to induction therapy regardless of treatment. VDZ induction therapy for UC reduced fCal levels at week 6, a marker of mucosal inflammation, significantly more than PBO. Response and remission rates were higher with VDZ regardless of baseline fCal concentration.
  • Example 3. Deep Remission at Week 6 as Predictor of Clinical Outcome in Ulcerative Colitis
  • Deep remission, a combination of endoscopic and patient-reported outcomes, is an emerging treatment goal for patients with ulcerative colitis (UC). However, there is currently no standardised definition for assessment of deep remission in UC patients. Statistically significant and clinically meaningful improvement in deep remission endpoints at weeks 6 and 52 were previously reported in the GEMINI 1 population using varying combinations of Mayo subscores of endoscopic and patient-reported outcomes. Here we investigate whether deep remission with VDZ is a predictor of clinical outcomes and health-related quality of life (HRQoL) in patients with UC using post hoc analyses of data from GEMINI 1.
  • Clinical and HRQoL endpoints were evaluated at week 52 in the maintenance ITT population who were in deep remission at weeks 6 or at 52:
      • Clinical remission: complete Mayo score of ≦2 points and no individual subscore >1 point
      • Mucosal healing: Mayo endoscopic subscore of ≦1
      • Corticosteroid (CS)-free remission: patients using oral CS at baseline who have discontinued CS and are in clinical remission at week 52
      • European Quality of Life-5 Dimension (EQ-5D) visual analogue scale (VAS): increase of ≧7 points represents clinically meaningful improvement.
      • Inflammatory Bowel Disease Questionnaire (IBDQ) total score: a score >170 is characteristic of the HRQoL of patients in remission. Clinical endpoints were analysed using observed cases at week 52; HRQoL endpoints used the last observation carried forward method to account for missing patients.
  • TABLE 5
    Definitions of Deep Remission based on Mayo
    endoscopic and patient-reported subscores
    Investigator Patient reported
    Reported Rectal Stool
    Endoscopic Bleeding Frequency
    Definition Subscore Subscore Subscore
    1a Endoscopic 0 0 Decrease or
    remission and no change
    symptomatic
    improvement
    2 Endoscopic 0 or 1 0 0 or 1
    and
    symptomatic
    improvement
    aDefinition 1 is considered more stringent than definition 2.
  • All patients in the maintenance ITT population received 2 doses of VDZ during induction, and rates of deep remission were comparable across treatment groups at week 6 (maintenance baseline) using both definitions. At week 52, almost 3 times as many VDZ-treated patients were in deep remission as were PBO-treated patients, according to either definition.
  • A greater percentage of patients in deep remission (definition 1 or 2) at week 6 met all clinical endpoints at week 52 compared with those not in deep remission, with VDZ Q4W demonstrating the greatest difference. More patients who were in deep remission at week 6 had improved clinical outcomes with VDZ maintenance treatment than with PBO (FIGS. 15A-15F and 16A-16F). Under definition 1, clinical remission after maintenance treatment with PBO was similar between those in and not in deep remission at week 6. In general, more patients in deep remission (definition 1 or 2) at week 6 reported clinically meaningful improvement in HRQoL at week 52 compared with those not in deep remission. Compared with PBO-treated patients in deep remission, more VDZ-treated patients in deep remission had improved IBDQ and EQ-5D VAS. Even among patients not in deep remission at week 6, maintenance treatment with VDZ resulted in more patients with improved clinical and HRQoL outcomes than those re-randomised to PBO.
  • Conclusions: A greater percentage of VDZ-treated patients were in deep remission at week 52 using either definition compared with PBO-treated patients; use of the less stringent definition 2 resulted in more patients classified as having achieved deep remission. Using either definition, a greater percentage of patients in deep remission at week 6 met most clinical and HRQoL endpoints at week 52 compared to those not in deep remission. More VDZ-treated patients had improved outcomes than PBO-treated patients. Achievement of deep remission at week 52 aligned with improved clinical and HRQoL outcomes at week 52 regardless of treatment group. These results suggest that monitoring deep remission in future studies could be used to predict patient outcomes.

Claims (30)

1. A method of treating a patient suffering from Inflammatory Bowel Disease (IBD), the method comprising the steps of:
a) measuring the concentration of vedolizumab in a patient wherein the patient was administered an induction phase dosing regimen of vedolizumab at least two weeks prior,
b) selecting the patient whose measurement of vedolizumab in step a) indicates low clearance; and
c) administering vedolizumab to the selected patient, thereby treating IBD.
2. The method of claim 1, wherein the concentration of vedolizumab is a serum concentration.
3. The method of claim 2, wherein the serum concentration is a trough serum concentration.
4. The method of claim 3, wherein the patient is selected if the serum concentration is at least 8 μg per ml.
5. The method of claim 1, wherein the concentration measurement is used to calculate the rate of clearance.
6. The method of claim 5, wherein the patient is selected if the rate of clearance is less than 0.25 L/day.
7. A method of treating a patient suffering from Inflammatory Bowel Disease (IBD), the method comprising the steps of:
a) administering two doses of vedolizumab to the patient suffering from IBD, wherein the second dose is administered about two weeks after the first dose is administered to the patient;
b) selecting the patient wherein the patient has a serum concentration of at least 8 μg per ml as measured at least two weeks after the second dose; and
c) administering vedolizumab to the selected patient, thereby treating the patient having IBD.
8. The method of claim 7, wherein the serum concentration is measured
about four weeks after the second dose.
9. The method of claim 3, wherein the patient's serum concentration is selected from the group consisting of 12-25 μg per ml, 15-17 μg per ml, 17-25 μg per ml, 12-40 μg per ml, and 17-40 μg per ml.
10. The method of claim 2, wherein the vedolizumab serum concentration is measured by a sandwich ELISA assay.
11. The method of claim 1, wherein the patient has mucosal healing.
12. The method of claim 1, further comprising measuring an endoscopic subscore, wherein vedolizumab is continued with an endoscopic subscore of less than 3.
13. (canceled)
14. The method of claim 12, wherein vedolizumab is continued with an endoscopic subscore of less than 2.
15. The method of claim 1, further comprising measuring SES-CO, wherein vedolizumab is continued with a SES-CD score ≦6.
16. (canceled)
17. The method of claim 1, further comprising measuring MaRIA, wherein vedolizumab is continued with a MaRIA score <12.
18. (canceled)
19. The method of claim 1, further comprising measuring the fecal calprotectin concentration.
20. The method of claim 19, wherein vedolizumab is continued with a fecal calprotectin concentration of <1500 μgig.
21. The method of claim 19, wherein vedolizumab is continued if the fecal calprotectin is reduced to <50% of the concentration before treatment.
22-35. (canceled)
36. A method for maintaining remission of inflammatory bowel disease in a patient, wherein the patient has received at least one dose of vedolizumab in the previous two months, the method comprising:
a) obtaining a serum sample from the patient;
b) measuring the concentration of vedolizumab in the sample;
c) administering vedolizumab thereafter every eight weeks if the concentration is at least 8 μg per ml.
37-45. (canceled)
46. The method of claim 6, further comprising measuring anti-vedolizumab antibodies.
47. The method of claim 46, wherein an anti-vedolizumab antibody titer of ≦125 further identifies the patient for continued treatment with vedolizumab.
48. The method of claim 1, further comprising measuring albumin concentration.
49. The method of claim 48, wherein an albumin concentration >3.2 g/dL further identifies the patient for continued treatment with vedolizumab.
50-59. (canceled)
60. A method for achieving corticosteroid-free remission of a patient having inflammatory bowel disease (IBD), wherein the patient was dependent on corticosteroids, the method comprising the steps of:
a) selecting a human patient having IBD and having a low clearance of vedolizumab at a time point that is four weeks after a second dose of vedolizumab, wherein a first dose of vedolizumab was administered to the subject two weeks prior to the second dose of vedolizumab,
b) administering vedolizumab to the human patient having IBD, and
c) decreasing the amount of corticosteroid by a tapering regimen, until no corticosteroid is being administered; thereby achieving corticosteroid-free remission in the patient having IBD.
US15/538,869 2014-12-24 2015-12-23 PREDICTING OUTCOME OF TREATMENT WITH AN ANTI-alpha4beta7 INTEGRIN ANTIBODY Abandoned US20170360926A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/538,869 US20170360926A1 (en) 2014-12-24 2015-12-23 PREDICTING OUTCOME OF TREATMENT WITH AN ANTI-alpha4beta7 INTEGRIN ANTIBODY

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201462096636P 2014-12-24 2014-12-24
US201562117750P 2015-02-18 2015-02-18
US201562132917P 2015-03-13 2015-03-13
US15/538,869 US20170360926A1 (en) 2014-12-24 2015-12-23 PREDICTING OUTCOME OF TREATMENT WITH AN ANTI-alpha4beta7 INTEGRIN ANTIBODY
PCT/US2015/000476 WO2016105572A1 (en) 2014-12-24 2015-12-23 PREDICTING OUTCOME OF TREATMENT WITH AN ANTI-α4β7 INTEGRIN ANTIBODY

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/000476 A-371-Of-International WO2016105572A1 (en) 2014-12-24 2015-12-23 PREDICTING OUTCOME OF TREATMENT WITH AN ANTI-α4β7 INTEGRIN ANTIBODY

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/455,103 Continuation US11596688B2 (en) 2014-12-24 2019-06-27 Predicting outcome of treatment with an anti-α4β7 integrin antibody

Publications (1)

Publication Number Publication Date
US20170360926A1 true US20170360926A1 (en) 2017-12-21

Family

ID=56151288

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/538,869 Abandoned US20170360926A1 (en) 2014-12-24 2015-12-23 PREDICTING OUTCOME OF TREATMENT WITH AN ANTI-alpha4beta7 INTEGRIN ANTIBODY
US16/455,103 Active US11596688B2 (en) 2014-12-24 2019-06-27 Predicting outcome of treatment with an anti-α4β7 integrin antibody

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/455,103 Active US11596688B2 (en) 2014-12-24 2019-06-27 Predicting outcome of treatment with an anti-α4β7 integrin antibody

Country Status (4)

Country Link
US (2) US20170360926A1 (en)
EP (1) EP3237004A4 (en)
JP (2) JP6904905B2 (en)
WO (1) WO2016105572A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020117795A1 (en) * 2018-12-04 2020-06-11 Prometheus Biosciences, Inc. Assessment and monitoring of mucosal healing in children and adults with crohn's disease
US11160863B2 (en) 2011-05-10 2021-11-02 Prometheus Laboratories Inc. Methods of disease activity profiling for personalized therapy management
US11162943B2 (en) 2017-05-31 2021-11-02 Prometheus Biosciences Inc. Methods for assessing mucosal healing in Crohn's disease patients
CN114974595A (en) * 2022-05-13 2022-08-30 江苏省人民医院(南京医科大学第一附属医院) Crohn's disease patient mucosa healing prediction model and method

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170327584A1 (en) 2014-11-26 2017-11-16 Millennium Pharmaceuticals, Inc. Vedolizumab for the Treatment of Fistulizing Crohn's Disease
US9862760B2 (en) 2015-09-16 2018-01-09 Novartis Ag Polyomavirus neutralizing antibodies
US11760803B2 (en) 2016-03-24 2023-09-19 Takeda Pharmaceutical Company Limited Methods of treating gastrointestinal immune-related adverse events in immune oncology treatments
EP3452070A1 (en) 2016-05-04 2019-03-13 Millennium Pharmaceuticals, Inc. Triple combination therapy for treating inflammatory bowel disease
WO2017218434A1 (en) 2016-06-12 2017-12-21 Millennium Pharmaceuticals, Inc. Method of treating inflammatory bowel disease
WO2018104893A1 (en) * 2016-12-06 2018-06-14 Glaxosmithkline Intellectual Property Development Limited Alpha4-beta7 antibodies with incrased fcrn binding and/or half-life
EP3630184A4 (en) * 2017-05-26 2021-03-17 Millennium Pharmaceuticals, Inc. Methods for the treatment of chronic pouchitis
SG11202003216QA (en) * 2017-10-10 2020-05-28 Prometheus Biosciences Inc Methods for monitoring vedolizumab treatment
US20210031012A1 (en) 2018-01-26 2021-02-04 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a pde4 inhibitor
US20210363233A1 (en) 2018-06-20 2021-11-25 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an il-12/il-23 inhibitor
EP3810085A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an integrin inhibitor
EP3810095A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a tnf inhibitor
WO2019246312A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an immunomodulator
US20230009902A1 (en) 2018-06-20 2023-01-12 Progenity, Inc. Treatment of a disease or condition in a tissue orginating from the endoderm
WO2019246273A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a jak or other kinase inhibitor
EP4165078A1 (en) * 2020-06-12 2023-04-19 Vera Therapeutics, Inc. Dosing of polyomavirus neutralizing antibodies

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
MX9706049A (en) 1995-02-10 1998-02-28 Leukosite Inc Mucosal vascular addressins and uses thereof.
US7803904B2 (en) 1995-09-01 2010-09-28 Millennium Pharmaceuticals, Inc. Mucosal vascular addressing and uses thereof
US7147851B1 (en) 1996-08-15 2006-12-12 Millennium Pharmaceuticals, Inc. Humanized immunoglobulin reactive with α4β7 integrin
US6197582B1 (en) 1998-03-18 2001-03-06 The Trustees Of Columbia University In The City Of New York Development of human monoclonal antibodies and uses thereof
US20010046496A1 (en) 2000-04-14 2001-11-29 Brettman Lee R. Method of administering an antibody
JP2007527407A (en) * 2003-05-22 2007-09-27 メタプロテオミクス, エルエルシー Anti-inflammatory pharmaceutical composition for reducing inflammation and for treating or preventing gastrointestinal toxicity
JP2009515552A (en) 2005-11-17 2009-04-16 ミレニアム・ファーマシューティカルズ・インコーポレイテッド α4β7 integrin-reactive humanized immunoglobulin
CA2741566A1 (en) * 2008-11-03 2010-06-03 Schering Corporation Inflammatory bowel disease biomarkers and related methods of treatment
CA2831732C (en) 2011-03-31 2019-12-31 Genentech, Inc. Use of a monoclonal anti-beta7 antibody for treating gastrointestinal inflammatory disorders
UA116189C2 (en) * 2011-05-02 2018-02-26 Мілленніум Фармасьютікалз, Інк. FORMULATION FOR ANTI-α4β7 ANTIBODY
MX367097B (en) 2011-05-02 2019-08-05 Millennium Pharm Inc FORMULATION FOR ANTI-a4ß7 ANTIBODY.
US20170327584A1 (en) 2014-11-26 2017-11-16 Millennium Pharmaceuticals, Inc. Vedolizumab for the Treatment of Fistulizing Crohn's Disease
CA2916283A1 (en) * 2015-01-09 2016-07-09 Pfizer Inc. Dosage regimen for madcam antagonists

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11160863B2 (en) 2011-05-10 2021-11-02 Prometheus Laboratories Inc. Methods of disease activity profiling for personalized therapy management
US11162943B2 (en) 2017-05-31 2021-11-02 Prometheus Biosciences Inc. Methods for assessing mucosal healing in Crohn's disease patients
US11796541B2 (en) 2017-05-31 2023-10-24 Prometheus Laboratories Inc. Methods for assessing mucosal healing in ulcerative colitis disease patients
WO2020117795A1 (en) * 2018-12-04 2020-06-11 Prometheus Biosciences, Inc. Assessment and monitoring of mucosal healing in children and adults with crohn's disease
CN114974595A (en) * 2022-05-13 2022-08-30 江苏省人民医院(南京医科大学第一附属医院) Crohn's disease patient mucosa healing prediction model and method

Also Published As

Publication number Publication date
JP2018503618A (en) 2018-02-08
WO2016105572A1 (en) 2016-06-30
JP2021102612A (en) 2021-07-15
EP3237004A1 (en) 2017-11-01
US11596688B2 (en) 2023-03-07
JP6904905B2 (en) 2021-07-21
US20200155673A1 (en) 2020-05-21
EP3237004A4 (en) 2018-06-13

Similar Documents

Publication Publication Date Title
US11596688B2 (en) Predicting outcome of treatment with an anti-α4β7 integrin antibody
JP6044904B2 (en) Methods and products for assessing immune responses to therapeutic proteins
RU2642295C2 (en) Analysis of identification of antibodies specific to therapeutic antibodies against ige, and their application in anaphylaxis
CN102124344B (en) Use of biomarkers for assessing treatment of gastrointestinal inflammatory disorders with beta7 integrin antagonists
Meroni et al. New strategies to address the pharmacodynamics and pharmacokinetics of tumor necrosis factor (TNF) inhibitors: A systematic analysis
KR102148063B1 (en) Methods of administering beta7 integrin antagonists
US20210252141A1 (en) Triple combination therapy for treating inflammatory bowel disease
JP6942053B2 (en) How to treat primary sclerosing cholangitis
Falkenburg et al. Evolution of autoantibody responses in individuals at risk of rheumatoid arthritis
Dreesen et al. Pharmacokinetic modeling and simulation of biologicals in inflammatory bowel disease: the dawning of a new era for personalized treatment
US11389533B2 (en) Method of treating inflammatory bowel disease
JP2023113655A (en) Method of treating pediatric disorders
Kernstock et al. Clinical immunogenicity risk assessment strategy for a low risk monoclonal antibody
Gehin Therapeutic drug monitoring of tumour necrosis factor alpha inhibitors in inflammatory joint diseases
Detrez et al. Characterization and application of a unique panel of monoclonal antibodies generated against etanercept
RU2778567C2 (en) Method for treatment of pediatric disorders/diseases
Rispens et al. Cross-Reactivity of Antibodies to Rituximab with Other Therapeutic Anti-CD20 Antibodies
Jani Immunogenicity in response to biologic agents
JPWO2011115231A1 (en) Osteopontin-specific monoclonal antibody
EA045594B1 (en) ANTIBODIES TO B7-H4 AND METHODS OF THEIR APPLICATION
JPWO2009125831A1 (en) Monoclonal antibody against NK4

Legal Events

Date Code Title Description
AS Assignment

Owner name: TAKEDA PHARMACEUTICAL COMPANY, LTD., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TAKEDA DEVELOPMENT CENTRE EUROPE LTD.;REEL/FRAME:043172/0005

Effective date: 20151211

Owner name: MILLENNIUM PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ROSARIO, MARIA;WYANT, TIMOTHY L.;SIGNING DATES FROM 20151207 TO 20151209;REEL/FRAME:043170/0238

Owner name: MILLENNIUM PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TAKEDA PHARMACEUTICAL COMPANY, LTD.;REEL/FRAME:043172/0093

Effective date: 20151218

Owner name: TAKEDA DEVELOPMENT CENTRE EUROPE LTD., UNITED KING

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABHYANKAR, BRIHAD;REEL/FRAME:043171/0689

Effective date: 20151214

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION