WO2017019673A2 - Méthodes permettant de diagnostiquer et de traiter une maladie inflammatoire de l'intestin - Google Patents

Méthodes permettant de diagnostiquer et de traiter une maladie inflammatoire de l'intestin Download PDF

Info

Publication number
WO2017019673A2
WO2017019673A2 PCT/US2016/044022 US2016044022W WO2017019673A2 WO 2017019673 A2 WO2017019673 A2 WO 2017019673A2 US 2016044022 W US2016044022 W US 2016044022W WO 2017019673 A2 WO2017019673 A2 WO 2017019673A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
level
patient
seq
antibody
Prior art date
Application number
PCT/US2016/044022
Other languages
English (en)
Other versions
WO2017019673A3 (fr
Inventor
Teresa Ramirez MONTAGUT
Akiko CHAI
Franklin FUH
Original Assignee
Genentech, Inc.
F. Hoffmann-La Roche Ag
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 Genentech, Inc., F. Hoffmann-La Roche Ag filed Critical Genentech, Inc.
Priority to JP2018503499A priority Critical patent/JP2018521661A/ja
Priority to CN201680055933.8A priority patent/CN108139385A/zh
Priority to EP16750314.3A priority patent/EP3329276A2/fr
Publication of WO2017019673A2 publication Critical patent/WO2017019673A2/fr
Publication of WO2017019673A3 publication Critical patent/WO2017019673A3/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/06Gastro-intestinal diseases
    • G01N2800/065Bowel diseases, e.g. Crohn, ulcerative colitis, IBS
    • 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

  • Biomarkers predictive of responsiveness to integrin beta7 antagonists including anti-beta7 integrin subunit antibodies, and methods of using such biomarkers are provided.
  • methods of treating gastrointestinal inflammatory disorders such as inflammatory bowel diseases including ulcerative colitis and Crohn's disease are provided.
  • methods of using such predictive biomarkers for the treatment of inflammatory bowel diseases including ulcerative colitis and Crohn's disease.
  • IBD Inflammatory bowel disease
  • GI gastrointestinal
  • CD ulcerative colitis
  • CD Crohn's disease
  • UC ulcerative colitis
  • CD Crohn's disease
  • Both conditions are characterized clinically by frequent bowel motions, malnutrition, and dehydration, with disruption in the activities of daily living.
  • CD is frequently complicated by the development of malabsorption, strictures, and fistulae and may require repeated surgery.
  • UC less frequently, may be complicated by severe bloody diarrhea and toxic megacolon, also requiring surgery.
  • Both IBD conditions are associated with an increased risk for malignancy of the GI tract.
  • the etiology of IBD is complex, and many aspects of the pathogenesis remain unclear.
  • TNF-a tumor necrosis factor alpha
  • infliximab a chimeric antibody
  • adalimumab a fully human antibody
  • AEs adverse events associated with anti TNFs
  • Other adverse events (AEs) associated with anti TNFs include elevated rates of bacterial infection, including tuberculosis, and, more rarely, lymphoma and demyelination (Chang et al, Nat Clin Pract Gastroenterol Hepatology 3:220 (2006); Hoentjen et al, World J. Gastroenterol. 15(17):2067 (2009)).
  • No currently available therapy achieves sustained remission in more than 20%-30% of IBD patients with chronic disease (Hanauer et al, Lancet 359: 1541-49 (2002); Sandborn et al., N Engl J Med 353: 1912-25 (2005)).
  • the integrins are alpha/beta heterodimeric cell surface glycoprotein receptors that play a role in numerous cellular processes including leukocyte adhesion, signaling, proliferation, and migration, as well as in gene regulation (Hynes, R. O., Cell, 1992, 69: 11-25; and Hemler, M. E., Annu. Rev. Immunol., 1990, 8:365-368). They are composed of two heterodimeric, non-covalently interacting a and ⁇ transmembrane subunits that bind specifically to distinct cell adhesion molecules (CAMs) on endothelia, epithelia, and extracellular matrix proteins.
  • CAMs cell adhesion molecules
  • integrins can function as tissue-specific cell adhesion receptors aiding in the recruitment of leukocytes from blood into nearly all tissue sites in a highly regulated manner, playing a role in the homing of leukocytes to normal tissue and to sites of inflammation (von Andrian et al, N Engl J Med 343: 1020-34 (2000)).
  • integrins are involved in leukocyte trafficking, adhesion and infiltration during inflammatory processes (Nakajima, H. et al, J. Exp. Med., 1994, 179: 1145-1154). Differential expression of integrins regulates the adhesive properties of cells and different integrins are involved in different inflammatory responses. (Butcher, E. C.
  • the leukocyte then arrests and begins the process of extravasation through the vascular endothelium to underlying tissue. This extravasation process is believed to occur in both the normal immune cell recirculation state and in inflammatory conditions (von Andrian et al., supra).
  • the numbers of ⁇ 4 ⁇ 7 + cells in infiltrates and the expression of the ligand MAdCAM-1 are higher at sites of chronic inflammation such as in the intestinal tract of patients with UC or CD (Briskin et al, Am J Pathol 151 :97-110 (1997); Souza et al, Gut 45:856-63 (1999)).
  • ⁇ 4 ⁇ 7 binds preferentially to high endothelial venules expressing MAdCAM-1 and vascular cell adhesion molecule
  • VCAM extracellular matrix molecule fibronectin fragment CS-1
  • MAdCAM-1 extracellular matrix molecule fibronectin fragment CS-1
  • the ⁇ 4 ⁇ 7 integrin plays a selective role in leukocyte gut tropism but does not seem to contribute to homing of leukocytes to the peripheral tissue or the CNS. Instead, peripheral lymphoid trafficking has been associated with ⁇ 4 ⁇ 1 interaction with VCAM-1 (Yednock et al., Nature 356:63-6 (1992); Rice et al, Neurology 64: 1336-42 (2005)).
  • ⁇ 7 integrin Another member of the ⁇ 7 integrin family, expressed exclusively on T lymphocytes and associated with mucosal tissues, is the ⁇ 7 integrin, otherwise known as CD103.
  • the ⁇ 7 integrin binds selectively to E-cadherin on epithelial cells and has been proposed to play a role in the retention of T cells in the mucosal tissue in the intraepithelial lymphocyte compartment (Cepek et al., J Immunol 150:3459-70 (1993); Karecla et al. Eur J Immunol 25:852-6 (1995)).
  • ⁇ 7 + cells in the lamina propria have been reported to exhibit cytotoxicity against stressed or infected epithelial cells (Hadley et al, J Immunol 159:3748-56 (1997); Buri et al, J Pathol 206: 178-85 (2005)).
  • ⁇ 7 The expression of ⁇ 7 is increased in CD (Elewaut et al, Acta Gastroenterol Belg 61 :288-94 (1998); Oshitani et al, Int J Mol Med 12:715-9 (2003)), and anti-a ⁇ 7 antibody treatment has been reported to attenuate experimental colitis in mice, implicating a role for ⁇ 7 + lymphocytes in experimental models of IBD (Ludviksson et al, J Immunol 162:4975-82 (1999)).
  • An anti-a4 antibody (natalizumab) reportedly has efficacy in treatment of patients with CD (Sandborn et al, N Engl J Med 2005;353: 1912-25) and an anti-a4p7 antibody (MLN-02, MLN0002, vedolizumab) reportedly is effective in patients with UC (Feagan et al, N Engl J Med 2005;352:2499-507).
  • a second anti-alpha4/beta7 antibody (AMG 181) is also in development and clinical trials have recently begun (clinicaltrials(dot)gov identifier,
  • rhuMAb Beta7 is derived from the rat anti-mouse/human monoclonal antibody FIB504 (Andrew et al. 1994). It was engineered to include human IgGl -heavy chain and Kl-light chain frameworks. Intn'l Patent Pub. No. WO2006/026759. Administration of etrolizumab to human patients according to certain dosing regimens has been described previously. See, e.g., Intn'l Patent Pub. No. WO/2012/135589.
  • RhuMAb Beta7 (etrolizumab) binds ⁇ 4 ⁇ 7 (Holzmann et al, Cell 56:37-46 (1989); Hu et al, Proc Natl Acad Sci USA 89:8254-8 (1992)) and ⁇ 7 (Cepek et al., J Immunol 150:3459-70 (1993)), which regulate trafficking and retention of lymphocyte subsets, respectively, in the intestinal mucosa.
  • rhuMAb Beta7 binds specifically to the ⁇ 7 subunit of ⁇ 4 ⁇ 7 and ⁇ 7 and does not bind to a4 or ⁇ integrin individual subunits. This was demonstrated by the inability of the antibody to inhibit adhesion of ⁇ 4 ⁇ 1+ ⁇ 4 ⁇ 7- Ramos cells to vascular cell adhesion molecule 1 (VCAM 1) at concentrations as high as 100 nM. Importantly, this characteristic of rhuMAb Beta7 indicates selectivity: T cell subsets expressing ⁇ 4 ⁇ 1 but not ⁇ 7 should not be directly affected by rhuMAb Beta7.
  • rhuMAb Beta7 Support for the gut-specific effects of rhuMAb Beta7 on leukocyte homing comes from several in vivo nonclinical studies. In severe combined immunodeficient (SCID) mice reconstituted with CD45RB hi 8 h CD4+ T cells (an animal model of colitis), rhuMAb Beta7 blocked radiolabeled lymphocyte homing to the inflamed colon but did not block homing to the spleen, a peripheral lymphoid organ. See, e.g., Intn'l Patent Pub. No. WO2006/026759.
  • rhuMAb Beta7 induced a moderate increase in peripheral blood lymphocyte numbers that was largely due to a marked (approximately three- to sixfold) increase in CD45RA ⁇ 7 1 ⁇ 1 ⁇ 11 peripheral blood T cells, a subset that is phenotypically similar to gut-homing memory/effector T cells in humans. See, e.g., Intn'l Patent Pub. No. WO2009/140684; Stefanich et al, Br. J. Pharmacol. 162: 1855-1870 (2011).
  • rhuMAb Beta7 had minimal to no effect on the number of CD45RA+ ⁇ 7intermediate peripheral blood T cells, a subset that is phenotypically similar to naive T cells in humans, and no effect on the number of CD45RA ⁇ 7 1 ⁇ peripheral blood T cells, a subset that is phenotypically similar to peripheral homing memory /effector T cells in humans, confirming the specificity of rhuMAb Beta7 for the gut homing lymphocyte subpopulation.
  • natalizumab treatment has been associated with confirmed cases of progressive multifocal leukoencephalopathy (PML) in patients with Crohn's disease (and separately, multiple sclerosis) who received concomitant treatment with natalizumab and immunosupressives.
  • PML is a potentially fatal neurological condition linked to reactivation of a polyomavirus (JC virus) and active viral replication in the brain. No known interventions can reliably prevent PML or adequately treat PML, if it occurs.
  • vedolizumab treatment is that it is administered intravenously which can be inconvenient for the patient and can also be associated with undesirable or adverse events, e.g., infusion site reactions. Accordingly, there is a need for improved therapeutic approaches to the treatment of gastrointestinal inflammatory disorders such as IBD, e.g., ulcerative colitis and Crohn's disease, as well as more desirable dosing regimens.
  • IBD gastrointestinal inflammatory disorders
  • Crohn's disease e.g., ulcerative colitis and Crohn's disease
  • the methods of the invention are based, at least in part, on the discovery that levels of certain cells, such as T cells, Treg cells, and/or Thl7 cells in biological samples obtained from patients, e.g., blood, are predictive of responsiveness of patients suffering from a gastrointestinal inflammatory disorder to treatment with integrin beta7 antagonists.
  • methods of predicting the response of a patient suffering from a gastrointestinal inflammatory disorder to a therapy comprising an integrin beta7 antagonist are provided.
  • the method comprises:
  • methods of predicting responsiveness of a gastrointestinal inflammatory disorder patient to an integrin beta7 antagonist treatment comprises determining the level of Treg cells in a biological sample from the patient, wherein an elevated level of Treg cells compared to a reference level of Treg cells identifies the patient as one who is likely to respond to the integrin beta7 antagonist treatment.
  • an elevated level of Treg cells is a level that is greater than a particular percentage or proportion of Treg cells in the sample.
  • an elevated level of Treg cells is a level that is equal to or greater than about 1.2% or about 1.25% or about 1.28% or about 1.3% or about 1.32% of cells in the sample.
  • the sample is a peripheral blood sample.
  • methods of identifying a patient suffering from a gastrointestinal inflammatory disorder as likely to respond to a therapy comprising an integrin beta7 antagonist are provided.
  • the method comprises:
  • methods of treating a patient having a gastrointestinal inflammatory disorder are provided.
  • the method comprises:
  • the patient is TNF-nai ' ve.
  • the reference level is the level of Treg cells in a population of patients suffering from a gastrointestinal inflammatory disorder. In some embodiments, the reference level is the level of Treg cells in a TNF-nai ' ve population of patients suffering from a gastrointestinal inflammatory disorder. In some embodiments, the level of Treg cells is determined as a proportion of Treg cells in the biological sample. In some embodiments, the proportion of Treg cells is determined by dividing the number of copies of demethylated FOXP3 gene in the biological sample by the number of copies of a control gene in the biological sample. In some embodiments, the method comprises determining the number of copies of demethylated FOXP3 Treg-specific demethylated region (TSDR). In some embodiments, the control gene is selected from GAPDH, ACTB, and COQ3.
  • methods of monitoring the response of a patient suffering from a gastrointestinal inflammatory disorder to a therapy comprising an integrin beta7 antagonist are provided.
  • the method comprises:
  • a method of monitoring responsiveness of a gastrointestinal inflammatory disorder patient to an integrin beta7 antagonist treatment comprises determining a first level of cells selected from CD3+ T cells, Treg cells, and Thl7 in a first biological sample from the patient taken at a first time point and determining a second level of the respective cells in a second biological sample from the patient taken at a second time point, wherein a higher level of the cells in the second biological sample compared to the first biological sample identifies the patient as one who is likely to experience clinical remission.
  • methods of identifying a patient suffering from a gastrointestinal inflammatory disorder and receiving a therapy comprising an integrin beta7 antagonist as likely to experience clinical remission comprise: a) measuring a first level of cells selected from CD3+ T cells, Treg cells, and Thl7 in a first biological sample from the patient taken at a first time point;
  • a method of treating a patient having a gastrointestinal inflammatory disorder comrprises:
  • the cells are CD3+ T cells. In some embodiments, the cells are Treg cells. In some embodiments, the cells are Thl7 cells.
  • the second time point is after the first time point.
  • the patient began treatment with an integrin beta7 antagonist after the first time point but prior to the second time point.
  • the patient began treatment with an integrin beta7 antagonist prior to the first time point, and continues treatment until at least the second time point.
  • the second time point is between 2 and 10 weeks after beginning treatment, or between 2 and 9 weeks, or between 2 and 8 weeks, or between 2 and 7 weeks, or between 2 and 6 weeks, or between 2 and 5 weeks, or between 2 and 4 weeks after beginning treatment.
  • the second time point is between 10 and 30 weeks after beginning treatment, or between 10 and 26 weeks after beginning treatment, or between 10 and 22 weeks after beginning treatment, or between 10 and 18 weeks after beginning treatment, or between 10 and 14 weeks after beginning treatment.
  • the first time point is before treatment is begun. In some embodiments, the first time point is within 4 weeks before treatment is begun, or within 3 weeks, or within 2 weeks, or within 1 week, or within 5 days, or within 4 days, or within 3 days, or within 2 days, or within 1 day before treatment is begun.
  • the proportion of Thl7 cells is determined by dividing the number of copies of demethylated IL17A gene in the biological sample by the number of copies of a control gene in the biological sample.
  • the control gene is selected from GAPDH, ACTB, and COQ3.
  • the inflammatory bowel disease is ulcerative colitis or Crohn's disease. In some embodiments, the inflammatory bowel disease is ulcerative colitis and the response is selected from clinical response, mucosal healing and remission. In some embodiments, remission in the patient is determined to be induced when the absolute Mayo Clinic Score ⁇ 2 and no individual subscore >1, which is also referred to as clinical remission. In certain embodiments, mucosal healing is determined to have occurred when the patient is determined to have an endoscopy subscore of 0 or 1 as assessed by flexible sigmoidoscopy. In certain such embodiments, patients who experience mucosal healing are determined to have an endoscopy subscore of 0. In certain embodiments, clinical response is determined to have occurred when the patient experiences a 3-point decrease and 30% reduction from baseline in MCS and > 1 -point decrease in rectal bleeding subscore or absolute rectal bleeding score of 0 or 1.
  • patients who experience mucosal healing are determined to have an endoscopy subscore of 0.
  • clinical response is determined to have occurred when the patient experiences a 3-point decrease and 30% reduction from baseline in MCS and > 1 -point decrease in rectal bleeding subscore or absolute rectal bleeding score of 0 or 1.
  • the kit comprises instructions for (i) determining the level of Treg cells in the biological sample; (ii) comparing the level to a reference level; and (iii) stratifying the patient into the category of integrin beta7 antagonist responder or integrin beta7 antagonist non- responder based on the comparison.
  • the integrin beta7 antagonist is for use in treating the patient wherein 105 mg is administered subcutaneously once every four weeks.
  • administering results in one or more of the following: (1) a 3-point decrease and 30% reduction from baseline in MCS and > 1-point decrease in rectal bleeding subscore or absolute rectal bleeding score of 0 or 1, (2) an endoscopic subscore of 0 or 1, (3) MCS ⁇ 2 with no individual subscore > 1.
  • 105 mg of the integrin beta7 antagonist is administered subcutaneously once every four weeks.
  • an initial dose of 210 mg of the integrin beta7 antagonist is administered subcutaneously followed by subsequent doses, each of 210 mg of the integrin beta7 antagonist administered subcutaneously, administered at each of weeks 2, 4, 8 and 12 after the initial dose.
  • the gastrointestinal inflammatory disorder is an inflammatory bowel disease, and in certain such embodiments, the inflammatory bowel disease is ulcerative colitis (UC) or Crohn's disease (CD), and in certain such embodiments, the integrin beta7 antagonist is a monoclonal anti-beta7 antibody.
  • UC ulcerative colitis
  • CD Crohn's disease
  • the integrin beta7 antagonist is a monoclonal anti-beta7 antibody.
  • HVR-H2 comprises amino acid sequence E1-E17 wherein E1-E17 is GYISYSGSTSYNPSLKS (SEQ ID NO:5), or a variant of SEQ ID NO:5 (SEQ ID NO:29) wherein amino acid E2 is selected from the group consisting of Y, F, V, and D, and/or amino acid E6 is selected from the group consisting of S and G, and/or amino acid E10 is selected from the group consisting of S and Y, and/or amino acid El 2 is selected from the group consisting of N, T, A, and D, and/or amino acid 13 is selected from the group consisting of P, H, D, and A, and/or amino acid E15 is selected from the group consisting of L and V, and/or amino acid El 7 is selected from the group consisting of S and G; and (vi) HVR-H3 comprises amino acid sequence F2-F11 wherein F2 -Fl l is MTGSSGYFDF (SEQ ID NO:6) or RT
  • HVR-L1 comprises SEQ ID NO:7, SEQ ID NO:8 or SEQ ID NO:9;
  • HVR-L2 comprises SEQ ID NO:2;
  • HVR-H3 comprises SEQ ID NO: 19.
  • the anti-beta7 antibody comprises a variable light chain comprising the amino acid sequence of SEQ ID NO:31 and a variable heavy chain comprising the amino acid sequence of SEQ ID NO:32.
  • FIG. 1A SEQ ID NO: 14
  • humanized hu504K graft variable heavy chain FIG. IB
  • variants hu504-5, hu504-16, and hu504-32 amino acid variations from humanized hu504K graft are indicated in FIG. 1 A
  • Figure 6A-C show changes from baseline levels of T cells (Fig. 6A), Treg cells (Fig. 6B), and Thl7 cells (Fig. 6C) over time in UC patients administered rhuMAb Beta7 (etrolizumab) that experienced clinical remission, UC patients administered rhuMAb Beta7 (etrolizumab) that did not experience remission, and UC patients that received placebo.
  • Treatment refers to clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • Effective treatment regimen refers to a treatment regimen that will offer beneficial response to a patient receiving the treatment.
  • Modifying a treatment refers to changing the treatment regimen including, changing dosage, frequency of administration, or duration of treatment, and/or addition of a second medication.
  • a "beneficial response" of a patient to treatment with an integrin beta7 antagonist and similar wording refers to the clinical or therapeutic benefit imparted to a patient at risk for or suffering from a gastrointestinal inflammatory disorder from or as a result of the treatment with the antagonist, such as an anti-beta7 integrin antibody.
  • Such benefit includes cellular or biological responses, a complete response, a partial response, a stable disease (without progression or relapse), or a response with a later relapse of the patient from or as a result of the treatment with the antagonist.
  • sample refers to a composition that is obtained or derived from a subject of interest that contains a cellular and/or other molecular entity that is to be characterized and/or identified, for example based on physical, biochemical, chemical and/or physiological characteristics.
  • disease sample and variations thereof refers to any sample obtained from a subject of interest that would be expected or is known to contain the cellular and/or molecular entity that is to be characterized.
  • the sample can be obtained from a tissue for the subject of interest or from peripheral blood of the subject.
  • the sample may be obtained from blood and other liquid samples of biological origin and tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom.
  • the source of the tissue sample may be solid tissue as from a fresh, frozen and/or preserved organ or tissue sample or biopsy or aspirate; blood or any blood constituents; bodily fluids; and cells from any time in gestation or development of the subject or plasma.
  • sample or “test sample” includes biological samples that have been manipulated in any way after their procurement, such as by treatment with reagents, solubilization, or enrichment for certain components, such as proteins or polynucleotides, or embedding in a semi-solid or solid matrix for sectioning purposes.
  • a "section" of a tissue sample is meant a single part or piece of a tissue sample, e.g. a thin slice of tissue or cells cut from a tissue sample. Samples include, but are not limited to, whole blood, blood- derived cells, serum, plasma, lypmph fluid, synovial fluid, cellular extracts, and combinations thereof.
  • a "reference sample,” as used herein, refers to any sample, standard, or level that is used for comparison purposes.
  • a reference sample is obtained from a healthy and/or non-diseased part of the body (e.g., tissue or cells) of the same subject or patient.
  • a reference sample is obtained from an untreated tissue and/or cell of the body of the same subject or patient.
  • a reference sample is obtained from a healthy and/or non-diseased part of the body (e.g., tissues or cells) of an individual who is not the subject or patient.
  • a reference sample is obtained from an untreated tissue and/or cell part of the body of an individual who is not the subject or patient.
  • a beta7 integrin antagonist or “beta7 antagonist” refers to any molecule that inhibits one or more biological activities or blocking binding of beta7 integrin with one or more of its associated molecules.
  • Antagonists of the invention can be used to modulate one or more aspects of beta7 associated effects, including but not limited to association with alpha4 integrin subunit, association with alphaE integrin subunit, binding of alpha4beta7 integrin to MAdCAM, VCAM-1 or fibronectin and binding of alphaEbeta7 integrin to E-cadherin.
  • beta7 antagonist is an anti-beta7 integrin antibody (or anti-beta7 antibody).
  • the anti-beta7 integrin antibody is a humanized anti-beta7 integrin antibody and more particularly a recombinant humanized monoclonal anti-beta7 antibody, for example, rhuMAb beta7, also referred to as etrolizumab.
  • the anti-beta7 antibodies of the present invention are anti-integrin beta7 antagonistic antibodies that inhibit or block the binding of beta7 subunit with alpha4 integrin subunit, association with alphaE integrin subunit, binding of alpha4beta7 integrin to MAdCAM, VCAM-1 or fibronectin and binding of alphaEbeta7 integrin to E-cadherin.
  • beta7 subunit or " ⁇ 7 subunit” is meant the human ⁇ 7 integrin subunit (Erie et al, (1991) J. Biol. Chem. 266: 11009-11016).
  • the beta7 subunit associates with alpha4 integrin subunit, such as the human .alpha.4 subunit (Kilger and Holzmann (1995) J. Mol. Biol. 73:347-354).
  • alpha4beta7 integrin is reportedly expressed on a majority of mature lymphocytes, as well as a small population of thymocytes, bone marrow cells and mast cells. (Kilshaw and Murant (1991) Eur. J. Immunol.
  • the beta7 subunit also associates with the alphaE subunit, such as the human alphaE integrin subunit (Cepek, K. L, et al. (1993) J. Immunol. 150:3459).
  • the alphaEbeta7 integrin is expressed on intra- intestinal epithelial lymphocytes (ilELs) (Cepek, K. L. (1993) supra).
  • alphaE subunit or "alphaE integrin subunit” or “ocE subunit” or “ocE integrin subunit” or “CD 103” is meant an integrin subunit found to be associated with beta7 integrin on intra-epithelial lymphocytes, which alphaEbeta7 integrin mediates binding of the iELs to intestinal epithelium expressing E-cadherin (Cepek, K. L. et al. (1993) J. Immunol. 150:3459; Shaw, S. K. and Brenner, M. B. (1995) Semin. Immunol. 7:335).
  • MAdCAM or “MAdCAM- 1” are used interchangeably in the context of the present invention and refer to the protein mucosal addressin cell adhesion molecule-1, which is a single chain polypeptide comprising a short cytoplasmic tail, a transmembrane region and an extracellular sequence composed of three immunoglobulin-like domains.
  • the cDNAs for murine, human and macaque MAdCAM-1 have been cloned (Briskin, et al, (1993) Nature, 363:461-464; Shyjan ⁇ a/., (1996) J. Immunol. 156:2851-2857).
  • VCAM-1 or "vascular cell adhesion molecule-1”
  • CD 106 refers to a ligand of alpha4beta7 and alpha4betal, expressed on activated endothelium and important in
  • CD45 refers to a protein of the protein tyrosine phosphatase (PTP) family.
  • PTPs are known to be signaling molecules that regulate a variety of cellular processes including cell growth, differentiation, mitotic cycle, and oncogenic transformation.
  • This PTP contains an extracellular domain, a single transmembrane segment and two tandem intracytoplasmic catalytic domains, and thus belongs to receptor type PTP. This gene is specifically expressed in hematopoietic cells. This PTP has been shown to be an essential regulator of T- and B-cell antigen receptor signaling.
  • CD45RA CD45RA, CD45RB, CD45RC, CD45RAB, CD45RAC, CD45RBC, CD45RO, CD45R (ABC).
  • CD45 is also highly glycosylated.
  • CD45R is the longest protein and migrates at 200 kDa when isolated from T cells.
  • B cells also express CD45R with heavier glycosylation, bringing the molecular weight to 220 kDa, hence the name B220;
  • B220 expression is not restricted to B cells and can also be expressed on activated T cells, on a subset of dendritic cells and other antigen presenting cells.
  • Stanton T, Boxall S Bennett A, et al. (2004).
  • CD45 variant alleles possibly increased frequency of a novel exon 4 CD45 polymorphism in HIV seropositive Kenyans.”
  • "Gut-homing lymphocytes” refer to a subgroup of lymphocytes having the characteristic of selectively homing to intestinal lymph nodes and tissues but not homing to peripheral lymph nodes and tissues. This subgroup of lymphocytes are characterized by an unique expression partem of a combination of multiples cell surface molecules, including, but not limited to, the combination of CD4, CD45RA and Beta7. Typically, at least two subsets of peripheral blood CD4 + lymphocytes can be subdivided based on the markers of CD45RA and Beta7, CD45RA ⁇ 7 hi ⁇ h , and CD45RA ⁇ 7 low CD4 + cells.
  • CD45RA ⁇ 7 hi ⁇ h CD4 + cells home preferentially to intestinal lymph nodes and tissues
  • CD45RA ⁇ 7 low CD4 + cells home preferentially to peripheral lymph nodes and tissues
  • Gut- homing lymphocytes are therefore a distinctive subgroup of lymphocytes identified as CD45RA ⁇ 7 hl h CD4 + in a flow cytometry assay. The methods of identifying this group of lymphocytes are well-known in the art.
  • CD4 + lymphocytes are a group of lymphocytes having CD4 expressed on their cell surfaces.
  • CD45RA lymphocytes are a group of lymphocytes having no CD45RA expressed on their cell surfaces.
  • An “amount” or “level” of biomarker can be determined using methods known in the art and disclosed herein, such as flow cytometry analysis or qPCR.
  • a "change in the amount or level of a biomarker" is as compared to a
  • a reference or comparator amount of the biomarker can be the amount of a biomarker before treatment and more particularly, can be the baseline or pre-dose amount.
  • Gastrointestinal inflammatory disorders are a group of chronic disorders that cause inflammation and/or ulceration in the mucous membrane. These disorders include, for example, inflammatory bowel disease (e.g., Crohn's disease, ulcerative colitis, indeterminate colitis and infectious colitis), mucositis (e.g., oral mucositis, gastrointestinal mucositis, nasal mucositis and proctitis), necrotizing enterocolitis and esophagitis.
  • inflammatory bowel disease e.g., Crohn's disease, ulcerative colitis, indeterminate colitis and infectious colitis
  • mucositis e.g., oral mucositis, gastrointestinal mucositis, nasal mucositis and proctitis
  • necrotizing enterocolitis and esophagitis necrotizing enterocolitis and esophagitis.
  • IBD Inflammatory Bowel Disease
  • IBD ulcerative colitis
  • Crohn's disease (CD) and “ulcerative colitis (UC)” are chronic inflammatory bowel diseases of unknown etiology. Crohn's disease, unlike ulcerative colitis, can affect any part of the bowel. The most prominent feature Crohn's disease is the granular, reddish-purple edematous thickening of the bowel wall. With the development of inflammation, these granulomas often lose their circumscribed borders and integrate with the surrounding tissue. Diarrhea and obstruction of the bowel are the predominant clinical features.
  • Crohn's disease As with ulcerative colitis, the course of Crohn's disease may be continuous or relapsing, mild or severe, but unlike ulcerative colitis, Crohn's disease is not curable by resection of the involved segment of bowel. Most patients with Crohn's disease require surgery at some point, but subsequent relapse is common and continuous medical treatment is usual.
  • Crohn's disease may involve any part of the alimentary tract from the mouth to the anus, although typically it appears in the ileocolic, small-intestinal or colonic-anorectal regions. Histopathologically, the disease manifests by discontinuous granulomatomas, crypt abscesses, fissures and aphthous ulcers.
  • the inflammatory infiltrate is mixed, consisting of lymphocytes (both T and B cells), plasma cells, macrophages, and neutrophils. There is a disproportionate increase in IgM- and IgG-secreting plasma cells, macrophages and neutrophils.
  • Anti-inflammatory drugs sulfasalazine and 5-aminosalisylic acid (5-ASA) are used for treating mildly active colonic Crohn's disease and are commonly prescribed in an attempt to maintain remission of the disease.
  • Metroidazole and ciprofloxacin are similar in efficacy to sulfasalazine and are particularly prescribed for treating perianal disease.
  • corticosteroids are prescribed to treat active exacerbations and can sometimes maintain remission.
  • Azathioprine and 6-mercaptopurine have also been used in patients who require chronic administration of corticosteroids. It has been suggested that these drugs may play a role in the long-term prophylaxis.
  • Antidiarrheal drugs can also provide symptomatic relief in some patients.
  • Nutritional therapy or elemental diet can improve the nutritional status of patients and induce symptomatic improvement of acute disease, but it does not induce sustained clinical remissions.
  • Antibiotics are used in treating secondary small bowel bacterial overgrowth and in treatment of pyogenic complications.
  • Ulcerative colitis afflicts the large intestine.
  • the course of the disease may be continuous or relapsing, mild or severe.
  • the earliest lesion is an inflammatory infiltration with abscess formation at the base of the crypts of Lieberkuhn. Coalescence of these distended and ruptured crypts tends to separate the overlying mucosa from its blood supply, leading to ulceration.
  • Symptoms of the disease include cramping, lower abdominal pain, rectal bleeding, and frequent, loose discharges consisting mainly of blood, pus and mucus with scanty fecal particles.
  • a total colectomy may be required for acute, severe or chronic, unremitting ulcerative colitis.
  • UC ulcerative colitis
  • onset may be insidious or abrupt, and may include diarrhea, tenesmus and relapsing rectal bleeding.
  • fulminant involvement of the entire colon toxic megacolon, a life-threatening emergency, may occur.
  • Extraintestinal manifestations include arthritis, pyoderma gangrenoum, uveitis, and erythema nodosum.
  • Treatment for UC includes sulfasalazine and related salicylate-containing drugs for mild cases and corticosteroid drugs in severe cases.
  • Topical administration of either salicylates or corticosteroids is sometimes effective, particularly when the disease is limited to the distal bowel, and is associated with decreased side effects compared with systemic use.
  • Supportive measures such as administration of iron and antidiarrheal agents are sometimes indicated.
  • Azathioprine, 6-mercaptopurine and methotrexate are sometimes also prescribed for use in refractory corticosteroid-dependent cases.
  • an “effective dosage” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • a "subject" herein is typically a human.
  • a subject is a non-human mammal.
  • exemplary non-human mammals include laboratory, domestic, pet, sport, and stock animals, e.g., mice, cats, dogs, horses, and cows.
  • the subject is eligible for treatment, e.g., treatment of a gastrointestinal inflammatory disorder.
  • antibody and “immunoglobulin” are used interchangeably in the broadest sense and include monoclonal antibodies (for example, full length or intact monoclonal antibodies), polyclonal antibodies, multivalent antibodies, multispecific antibodies (e.g., bispecific antibodies so long as they exhibit the desired biological activity) and may also include certain antibody fragments (as described in greater detail herein).
  • An antibody can be human, humanized and/or affinity matured.
  • Antibody fragments comprise only a portion of an intact antibody, wherein the portion preferably retains at least one, and typically most or all, of the functions normally associated with that portion when present in an intact antibody.
  • an antibody fragment comprises an antigen binding site of the intact antibody and thus retains the ability to bind antigen.
  • an antibody fragment for example one that comprises the Fc region, retains at least one of the biological functions normally associated with the Fc region when present in an intact antibody, such as FcRn binding, antibody half life modulation, ADCC function and complement binding.
  • an antibody fragment is a monovalent antibody that has an in vivo half life substantially similar to an intact antibody.
  • such an antibody fragment may comprise on antigen binding arm linked to an Fc sequence capable of conferring in vivo stability to the fragment.
  • 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 except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigen. Furthermore, 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 monoclonal antibodies herein specifically include “chimeric” antibodies in which a portion of the heavy and/or light chain is identical with or homologous to
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin lo sequence.
  • the humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • a "human antibody” is one which comprises an amino acid sequence corresponding to that of an antibody produced by a human and/or has been made using any of the techniques for making human antibodies as disclosed herein. Such techniques include screening human- derived combinatorial libraries, such as phage display libraries (see, e.g., Marks et al. , J. Mol. Biol, 222: 581-597 (1991) and Hoogenboom et al. , Nucl. Acids Res. , 19: 4133-4137 (1991)); using human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies (see, e.g., Kozbor J.
  • human- derived combinatorial libraries such as phage display libraries (see, e.g., Marks et al. , J. Mol. Biol, 222: 581-597 (1991) and Hoogenboom et al. , Nucl. Acids Res. , 19: 4
  • An "isolated" antibody is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or
  • the antibody will be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method, and often 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.
  • hypervariable region when used herein refers to the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops.
  • antibodies comprise six hypervariable regions; three in the VH (HI, H2, H3), and three in the VL (LI, L2, L3).
  • a number of hypervariable region delineations are in use and are encompassed herein.
  • the Kabat Complementarity Determining Regions are based on sequence variability and are the most commonly used (Kabat et al, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)).
  • Chothia refers instead to the location of the structural loops (Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)).
  • the AbM hypervariable regions represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular's AbM antibody modeling software.
  • the "contact" hypervariable regions are based on an analysis of the available complex crystal structures. The residues from each of these HVRs are noted below.
  • Hypervariable regions may comprise "extended hypervariable regions” as follows: 24-36 or 24-34 (LI), 46-56 or 49-56 or 50-56 or 52-56 (L2) and 89-97 (L3) in the VL and 26- 35 (HI), 50-65 or 49-65 (H2) and 93-102, 94-102 or 95-102 (H3) in the VH.
  • the variable domain residues are numbered according to Kabat et al, supra for each of these definitions.
  • affinity matured antibody is one with one or more alterations in one or more CDRs thereof which result in an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s).
  • affinity matured antibodies will have nanomolar or even picomolar affinities for the target antigen.
  • Affinity matured antibodies are produced by procedures known in the art. Marks et al. Bio/Technology 10:779-783 (1992) describes affinity maturation by VH and VL domain shuffling. Random mutagenesis of CDR and/or framework residues is described by: Barbas et al. Proc Nat. Acad.
  • Low-affinity antibodies generally bind antigen slowly and tend to dissociate readily, whereas high-affinity antibodies generally bind antigen faster and tend to remain bound longer.
  • a variety of methods of measuring binding affinity are known in the art, any of which can be used for purposes of the present invention.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual "Fc” fragment, whose name reflects its ability to crystallize readily.
  • Pepsin treatment yields an F(ab')2 fragment that has two antigen-binding sites and is still capable of cross-linking antigen.
  • Fv is the minimum antibody fragment which contains a complete antigen- recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three hypervariable regions of each variable domain interact to define an antigen- binding site on the surface of the VH-VL dimer. Collectively, the six hypervariable regions confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • a "naked antibody” for the purposes herein is an antibody that is not conjugated to a cytotoxic moiety or radiolabel.
  • Fc region herein is used to define a C-terminal region of an immunoglobulin heavy chain, including native sequence Fc regions and variant Fc regions.
  • the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof.
  • the C-terminal lysine (residue 447 according to the EU numbering system) of the Fc region may be removed, for example, during production or purification of the antibody, or by recombinantly engineering the nucleic acid encoding a heavy chain of the antibody. Accordingly, a composition of intact antibodies may comprise antibody populations with all K447 residues removed, antibody populations with no K447 residues removed, and antibody populations having a mixture of antibodies with and without the K447 residue.
  • Antibody-dependent cell-mediated cytotoxicity and “ADCC” refer to a cell- mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (FcRs) (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcRs Fc receptors
  • FcR expression on hematopoietic cells in summarized is Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991).
  • FcR FcR
  • 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 regulates homeostasis of immunoglobulins.
  • Antibodies with improved binding to the neonatal Fc receptor (FcRn), and increased half-lives, are described in WO00/42072 (Presta, L.) and US2005/0014934A1 (Hinton et al).
  • Single-chain Fv or “scFv” antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding.
  • HER2 antibody scFv fragments are described in W093/16185; U.S. Patent No. 5,571,894; and U.S. Patent No. 5,587,458.
  • amino acid sequence variant antibody herein is an antibody with an amino acid sequence which differs from a main species antibody.
  • amino acid sequence variants will possess at least about 70% homology with the main species antibody, or they will be at least about 80%, or at least about 90% homologous with the main species antibody.
  • the amino acid sequence variants possess substitutions, deletions, and/or additions at certain positions within or adjacent to the amino acid sequence of the main species antibody.
  • amino acid sequence variants herein include an acidic variant (e.g., deamidated antibody variant), a basic variant, an antibody with an amino-terminal leader extension (e.g.
  • cytokine is a generic term for proteins released by one cell population which act on another cell as intercellular mediators.
  • cytokines are lymphokines, monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor-a and - ⁇ ; mullerian- inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF- ⁇ ; platelet
  • ILs interleukins
  • IL-1 interleukins
  • IL-la interleukins
  • IL-2 interleukins
  • IL-3 interleukins
  • IL-4 interleukins
  • IL-5 IL-6
  • IL-7 tumor necrosis factor
  • IL-8 IL-9
  • IL-10 IL- 11, IL-12
  • KL kit ligand
  • cytokine includes proteins from natural sources or from recombinant cell culture and biologically active equivalents of the native sequence cytokines.
  • non-steroidal antiinflammatory drugs NSAIDs
  • ganciclovir tacrolimus
  • glucocorticoids such as Cortisol or aldosterone
  • anti-inflammatory agents such as a cyclooxygenase inhibitor; a 5-lipoxygenase inhibitor; or a leukotriene receptor antagonist
  • purine antagonists such as azathioprine or mycophenolate mofetil (MMF)
  • alkylating agents such as cyclophosphamide; bromocryptine; danazol; dapsone; glutaraldehyde (which masks the MHC antigens, as described in U.S. Patent No. 4,120,649)
  • anti-idiotypic antibodies for MHC antigens and MHC fragments NSAIDs
  • tacrolimus such as Cortisol or aldosterone
  • anti-inflammatory agents such as a cyclooxygenase inhibitor; a 5-lipoxygenase inhibitor; or a leukot
  • methylprednisolone sodium succinate, and dexamethasone dihydrofolate reductase inhibitors such as methotrexate (oral or subcutaneous); anti-malarial agents such as chloroquine and hydroxychloroquine; sulfasalazine; leflunomide; cytokine or cytokine receptor antibodies or antagonists including anti-interferon-alpha, -beta, or -gamma antibodies, antitumor necrosis factor(TNF)-alpha antibodies (infliximab (REMICADE.RTM.) or
  • adalimumab anti-TNF-alpha immunoadhesin (etanercept), anti-TNF-beta antibodies, anti- interleukin-2 (IL-2) antibodies and anti-IL-2 receptor antibodies, and anti-interleukin-6 (IL-6) receptor antibodies and antagonists; anti-LFA-1 antibodies, including anti-CDl la and anti- CD 18 antibodies; anti-L3T4 antibodies; heterologous anti -lymphocyte globulin; pan-T antibodies, anti-CD3 or anti-CD4/CD4a antibodies; soluble peptide containing a LFA-3 binding domain (WO 90/08187 published Jul. 26, 1990); streptokinase; transforming growth factor-beta (TGF-beta); streptodomase; RNA or DNA from the host; FK506; RS-61443;
  • T-cell receptor Cohen et al, U.S. Patent No. 5,114,721
  • T-cell receptor fragments Offner et al, Science, 251 : 430-432 (1991); WO 90/11294; Ianeway, Nature, 341 : 482 (1989); and WO 91/01133
  • BAFF antagonists such as BAFF or BR3 antibodies or immunoadhesins and zTNF4 antagonists (for review, see Mackay and Mackay, Trends Immunol, 23: 113-5 (2002) and see also definition below
  • biologic agents that interfere with T cell helper signals such as anti-CD40 receptor or anti-CD40 ligand (CD154), including blocking antibodies to CD40-CD40 ligand.(e.g., Durie et al., Science, 261 : 1328-30 (1993); Mohan et al, J. Immunol, 154: 1470-80 (1995)) and CTLA
  • Antagonist refers to a molecule capable of neutralizing, blocking, inhibiting, abrogating, reducing or interfering with the activities of a particular or specified protein, including its binding to one or more receptors in the case of a ligand or binding to one or more ligands in case of a receptor.
  • Antagonists include antibodies and antigen-binding fragments thereof, proteins, peptides, glycoproteins, glycopeptides, glycolipids,
  • Oligonucleotide refers to short, single stranded
  • Oligonucleotides that are at least about seven nucleotides in length and less than about 250 nucleotides in length. Oligonucleotides may be synthetic. The terms “oligonucleotide” and “polynucleotide” are not mutually exclusive. The description above for polynucleotides is equally and fully applicable to oligonucleotides.
  • Amplification refers to the process of producing one or more copies of a reference nucleic acid sequence or its complement. Amplification may be linear or exponential (e.g., PCR). A “copy” does not necessarily mean perfect sequence
  • copies can include nucleotide analogs such as deoxyinosine, intentional sequence alterations (such as sequence alterations introduced through a primer comprising a sequence that is hybridizable, but not fully complementary, to the template), and/or sequence errors that occur during amplification.
  • nucleotide analogs such as deoxyinosine
  • intentional sequence alterations such as sequence alterations introduced through a primer comprising a sequence that is hybridizable, but not fully complementary, to the template
  • sequence errors that occur during amplification.
  • multiplex-PCR refers to a single PCR reaction carried out on nucleic acid obtained from a single source (e.g., a patient) using more than one primer set for the purpose of amplifying two or more DNA sequences in a single reaction.
  • diagnosis is used herein to refer to the identification or classification of a molecular or pathological state, disease or condition.
  • diagnosis may refer to identification of a particular type of gastrointestinal inflammatory disorder, or the classification of a particular sub-type of gastrointestinal inflammatory disorder, by tissue/organ involvement (e.g., inflammatory bowel disease), or by other features (e.g., a patient subpopulation characterized by responsiveness to a treatment, such as to a treatment with an integrin beta7 antagonist), or by molecular features (e.g., a subtype characterized by expression of one or a combination of particular genes or proteins encoded by said genes).
  • tissue/organ involvement e.g., inflammatory bowel disease
  • other features e.g., a patient subpopulation characterized by responsiveness to a treatment, such as to a treatment with an integrin beta7 antagonist
  • molecular features e.g., a subtype characterized by expression of one or a combination of particular genes or proteins encoded by said genes.
  • control subject refers to a healthy subject who has not been diagnosed as having a particular disease, e.g., IBD, and who does not suffer from any sign or symptom associated with that disease.
  • a first analysis or protocol may be used in carrying out a second protocols and/or one may use the results of a first analysis or protocol to determine whether a second analysis or protocol should be performed.
  • biomarker level analysis or protocol one may use the results of the biomarker level analysis or protocol to determine whether a specific therapeutic regimen should be performed.
  • comparing refers to comparing the level of the biomarker in the sample from the individual or patient with the reference level of the biomarker specified elsewhere in this description. It is to be understood that comparing as used herein usually refers to a comparison of corresponding parameters or values, e.g., an absolute amount is compared to an absolute reference amount while a concentration is compared to a reference concentration or an intensity signal obtained from the biomarker in a sample is compared to the same type of intensity signal obtained from a reference sample.
  • the comparison may be carried out manually or computer assisted. Thus, the comparison may be carried out by a computing device (e.g., of a system disclosed herein).
  • a "kit” is any manufacture (e.g. a package or container) comprising at least one reagent, e.g. , a medicament for treatment of an IBD, e.g., UC or Crohn's disease, or a probe for specifically detecting a biomarker gene or protein of the invention.
  • the manufacture is promoted, distributed, or sold as a unit for performing the methods of the present invention.
  • a "target audience” is a group of people or an institution to whom or to which a particular medicament is being promoted or intended to be promoted, as by marketing or advertising, especially for particular uses, treatments, or indications, such as individual patients, patient populations, readers of newspapers, medical literature, and magazines, television or internet viewers, radio or internet listeners, physicians, drug companies, etc.
  • Such responsiveness may be assessed by a clinician skilled in treating the disorder in question.
  • a subject who experiences "a clinically unacceptably high level of toxicity" from previous or current treatment with one or more medicaments experiences one or more negative side-effects or adverse events associated therewith that are considered by an experienced clinician to be significant, such as, for example, serious infections, congestive heart failure, demyelination (leading to multiple sclerosis), significant hypersensitivity, neuropathological events, high degrees of
  • a cancer such as endometrial cancer, non-Hodgkin's lymphoma, breast cancer, prostate cancer, lung cancer, ovarian cancer, or melanoma, tuberculosis (TB), and the like.
  • the "amount” or "level” of a biomarker associated with an increased clinical benefit to a patient suffering from a certain disease or disorder, or predictive of response to a particular therapeutic agent or treatment regimen, is a detectable level in a biological sample. These can be measured by methods known to one skilled in the art and also disclosed herein. The level of a biomarker can be used to determine the response or the predicted response to a treatment or therapeutic agent.
  • beta7 integrin antagonists include an oligonucleotide that binds to the fusions of immunoglobulin with beta7 integrin, and, in particular, antibodies including, without limitation, poly- and monoclonal antibodies and antibody fragments, single-chain antibodies, anti-idiotypic antibodies, and chimeric or humanized versions of such antibodies or fragments, as well as human antibodies and antibody fragments.
  • a potential antagonist may be a closely related protein, for example, a mutated form of the beta7 integrin that recognizes the ligand but imparts no effect, thereby competitively inhibiting the action of the beta7 integrin.
  • beta7 integrin antagonist is an antisense RNA or DNA construct prepared using antisense technology, where, e.g., an antisense RNA or DNA molecule acts to block directly the translation of mRNA by hybridizing to targeted mRNA and preventing protein translation.
  • Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA.
  • the 5' coding portion of the polynucleotide sequence, which encodes the beta7 integrin herein is used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length.
  • the antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into beta7 integrin protein (antisense—Okano, Neurochem, 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression (CRC Press: Boca Raton, Fla., 1988).
  • the oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of the PRO polypeptide.
  • antisense DNA is used, oligodeoxyribonucleotides derived from the translation-initiation site, e.g., between about -10 and +10 positions of the target gene nucleotide sequence, are typical.
  • Other potential antagonists include small molecules that bind to the active site, the ligand or binding molecule binding site, thereby blocking the normal biological activity of the beta7 integrin.
  • small molecules include, but are not limited to, small peptides or peptide-like molecules, typically soluble peptides, and synthetic non-peptidyl organic or inorganic compounds.
  • Nucleic acid molecules in triple-helix formation used to inhibit transcription should be single-stranded and composed of deoxynucleotides.
  • the base composition of these oligonucleotides is designed such that it promotes triple-helix formation via Hoogsteen base- pairing rules, which generally require sizeable stretches of purines or pyrimidines on one strand of a duplex.
  • PCT Publication No. WO 97/33551 See, e.g., PCT Publication No. WO 97/33551.
  • These small molecules can be identified by any one or more of the screening assays discussed hereinabove and/or by any other screening techniques well known for those skilled in the art.
  • Screening assays for antagonists are designed to identify compounds that bind or complex with the beta7 integrin encoded by the genes identified herein, or otherwise interfere with the interaction of the encoded polypeptides with other cellular proteins.
  • Such screening assays will include assays amenable to high-throughput screening of chemical libraries, making them particularly suitable for identifying small molecule drug candidates.
  • the assays can be performed in a variety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays, and cell-based assays, which are well characterized in the art.
  • the beta7 integrin antagonists are anti-beta7 antibodies.
  • Exemplary antibodies include polyclonal, monoclonal, humanized, human, bispecific, and heteroconjugate antibodies, etc., as described below.
  • SC subcutaneous
  • IP intraperitoneal
  • a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyanin, serum albumin
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 ⁇ g or 5 ⁇ g of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites.
  • the animals are boosted with 1/5 to 1/10 the original amount of peptide or conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites.
  • Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus.
  • the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent.
  • Conjugates also can be made in recombinant cell culture as protein fusions. Also, aggregating agents such as alum are suitably used to enhance the immune response.
  • Monoclonal antibodies may be made using 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. Patent No. 4,816,567).
  • a mouse or other appropriate host animal such as a hamster, is immunized as described above to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. After immunization, lymphocytes are isolated and then fused with a myeloma cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • a suitable fusing agent such as polyethylene glycol
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium which medium may contain one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells (also referred to as fusion partner).
  • a suitable culture medium which medium may contain one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells (also referred to as fusion partner).
  • the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT)
  • HGPRT or HPRT the selective culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • fusion partner myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody- producing cells, and are sensitive to a selective medium that selects against the unfused parental cells.
  • myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 and derivatives e.g., X63-Ag8-653 cells available from the American Type Culture Collection, Manassas, Va., USA.
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunosorbent assay
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis described in Munson et al, Anal. Biochem, 107:220 (1980). Once hybridoma cells that produce antibodies of the desired specificity, affinity, and/or activity are identified, the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59- 103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D- MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal e.g., by i.p. injection of the cells into mice.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional antibody purification procedures such as, for example, affinity chromatography (e.g., using protein A or protein G-Sepharose) or ion-exchange
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells serve as a source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or myeloma cells that do not otherwise produce antibody protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or myeloma cells that do not otherwise produce antibody protein.
  • Review articles on recombinant expression in bacteria of DNA encoding the antibody include Skerra et al, Curr. Opinion in Immunol, 5:256-262 (1993) and
  • monoclonal antibodies or antibody fragments can be isolated from antibody phage libraries generated using e.g., the techniques described in McCafferty et al, Nature, 348:552-554 (1990). Clackson et al, Nature, 352:624-628 (1991) and Marks et al, J. Mol. Biol, 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • the DNA that encodes the antibody may be modified to produce chimeric or fusion antibody polypeptides, for example, by substituting human heavy chain and light chain constant domain (CH and CL) sequences for the homologous murine sequences (U.S. Patent No. 4,816,567; and Morrison, et al, Proc. Natl. Acad. Sci. USA, 81 :6851 (1984)), or by fusing the immunoglobulin coding sequence with all or part of the coding sequence for a non- immunoglobulin polypeptide (heterologous polypeptide).
  • CH and CL constant domain
  • non-immunoglobulin polypeptide sequences can substitute for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • Exemplary anti-beta7 antibodies are Fib504, Fib 21, 22, 27, 30 (Tidswell, M. J
  • the anti-beta7 integrin antibodies of the invention may further comprise humanized antibodies or human antibodies.
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non- human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • humanized antibodies are replaced by corresponding non-human residues.
  • Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin [Jones et al, Nature, 321 :522-525 (1986); Riechmann et al, Nature 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol, 2:593-596 (1992)].
  • Fc immunoglobulin constant region
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers [Jones et al, Nature, 321 :522-525 (1986); Riechmann et al, Nature, 332:323-327 (1988); Verhoeyen et al, Science, 239: 1534-1536 (1988)], by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized” antibodies are chimeric antibodies (U.S.
  • humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • the choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity and HAMA response (human anti-mouse antibody) when the antibody is intended for human therapeutic use.
  • the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable domain sequences.
  • the human V domain sequence which is closest to that of the rodent is identified and the human framework region (FR) within it accepted for the humanized antibody (Sims et al, J. Immunol. 151 :2296 (1993); C o a. et al, J. Mol. Biol, 196:901 (1987)).
  • Another method uses a particular framework region derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (Carter et al, Proc. Natl. Acad. Sci.
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three- dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three- dimensional conformational structures of selected candidate immunoglobulin sequences.
  • the humanized antibody may be an antibody fragment, such as a Fab, which is optionally conjugated with one or more cytotoxic agent(s) in order to generate an
  • humanized anti-beta7 antibodies include, but are not limited to rhuMAb Beta7, which is a humanized monoclonal antibody against the integrin subunit ⁇ 7 and was derived from the rat anti-mouse/human monoclonal antibody FIB504 (Andrew et al, 1994 J Immunol 1994;153:3847-61). It has been engineered to include human immunoglobulin IgGl heavy chain and ⁇ light chain frameworks and is produced by Chinese hamster ovary cells. This antibody binds to two integrins, ⁇ 4 ⁇ 7 (Holzmann et al.
  • IBD inflammatory bowel diseases
  • UC ulcerative colitis
  • CD Crohn's disease
  • rhuMAb Beta7 is a potent in vitro blocker of the cellular interaction between ⁇ 4 ⁇ 7 and its ligands (mucosal addressin cell adhesion molecule-1 [MAdCAM]-l, vascular cell adhesion molecule [VCAM]-1, and fibronectin) as well as the interaction between ⁇ 7 and its ligand (E-cadherin).
  • rhuMAb Beta7 binds reversibly, with similar high affinity, to ⁇ 7 on lymphocytes from rabbits, cynomolgus monkeys, and humans. It also binds to mouse ⁇ 7 with high affinity.
  • the amino acid sequence as well as the making and using of rhuMAb Beta7 and its variants are disclosed in detail in e.g., U.S. Patent Application Publication No. 20060093601 (issued as U.S. Patent No.
  • FIGS. 1 A and IB depict alignment of sequences of the variable light and heavy chains for the following: light chain human subgroup kappa I consensus sequence (FIG. 1 A, SEQ ID NO: 12), heavy chain human subgroup III consensus sequence (FIG. IB, SEQ ID NO: 13), rat anti-mouse beta7 antibody (Fib504) variable light chain (FIG. 1A, SEQ ID NO: 10), rat anti-mouse beta7 antibody (Fib504) variable heavy chain (FIG. IB, SEQ ID NO: 11), and humanized antibody variants: Humanized hu504Kgraft variable light chain (FIG. 1A, SEQ ID NO: 14), humanized hu504K graft variable heavy chain (FIG.
  • human antibodies can be generated.
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • JH antibody heavy-chain joining region
  • transfer of the human germ-line immunoglobulin gene array into such germ-line mutant mice will result in the production of human antibodies upon antigen challenge. See, e.g., Jakobovits et al, Proc. Natl. Acad. Sci.
  • V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as Ml 3 or fd, and displayed as functional antibody fragments on the surface of the phage particle. Because the filamentous particle contains a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties. Thus, the phage mimics some of the properties of the B-cell.
  • a filamentous bacteriophage such as Ml 3 or fd
  • Phage display can be performed in a variety of formats, reviewed in, e.g., Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3 :564-571 (1993).
  • V-gene segments can be used for phage display.
  • Clackson et al, Nature, 352:624- 628(1991) isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice.
  • a repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by Marks et al, J. Mol. Biol. 222:581-597 (1991), or Griffith et al, EMBO J.
  • human antibodies may also be generated by in vitro activated B cells (see U. S. Patent Nos. 5,567,610 and 5,229,275).
  • F(ab')2 fragments can be isolated directly from recombinant host cell culture.
  • the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; US Patent No. 5,571,894; and US Patent No. 5,587,458.
  • the antibody fragment may also be a "linear antibody," e.g., as described in US Patent No. 5,641,870 for example. Such linear antibody fragments may be monospecific or bispecific.
  • Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes.
  • Exemplary bispecific antibodies may bind to two different epitopes of beta7 integrin as described herein.
  • Other such antibodies may combine a TAT binding site with a binding site for another protein.
  • an anti-Beta7 integrin arm may be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g., CD3), or Fc receptors for IgG (Fc.y.R), such as FcyRI (CD64),
  • Bispecific antibodies may also be used to localize cytotoxic agents to cells which express TAT. These antibodies possess a TAT-binding arm and an arm which binds the cytotoxic agent (e.g., saporin, anti-interferon-. alpha., vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten). Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g., F(ab')2 bispecific antibodies).
  • immunoglobulin heavy chain-light chain pairs where the two chains have different specificities (Millstein et al, Nature 305 :537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion is with an Ig heavy chain constant domain, comprising at least part of the hinge, Cm, and CH3 regions.
  • the first heavy-chain constant region (CHI) containing the site necessary for light chain bonding present in at least one of the fusions.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details of generating bispecific antibodies see, for example, Suresh et al, Methods in Enzymology 121 :210 (1986).
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture.
  • the interface comprises at least a part of the Cro domain.
  • one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g., tyrosine or tryptophan).
  • Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g., alanine or threonine).
  • Bispecific antibodies include cross-linked or "heteroconjugate" antibodies.
  • one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin.
  • Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089).
  • Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Patent No. 4,676,980, along with a number of cross-linking techniques.
  • bispecific antibodies can be prepared using chemical linkage.
  • Brennan et al, Science 229: 81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab').sub.2 fragments. These fragments are reduced in the presence of the dithiol complexing agent, sodium arsenite, to stabilize vicinal dithiols and prevent intermolecular disulfide formation.
  • the Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives:
  • TAB thionitrobenzoate
  • One of the Fab'-TNB derivatives is then reconverted to the Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody.
  • the bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
  • the leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion.
  • the antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers.
  • the "diabody” technology described by Hollinger et al, Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments.
  • the fragments comprise a V.sub.H connected to a V.sub.L by a linker which is too short to allow pairing between the two domains on the same chain.
  • V.sub.H and V.sub.L domains of one fragment are forced to pair with the complementary V.sub.L and V.sub.H domains of another fragment, thereby forming two antigen-binding sites.
  • Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See Gruber et al, J. Immunol, 152:5368 (1994).
  • Antibodies with more than two valencies are contemplated.
  • trispecific antibodies can be prepared. Tutt et al, J. Immunol. 147:60 (1991).
  • Heteroconjugate antibodies are also within the scope of the present invention.
  • Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells [U.S. Patent No. 4,676,980], and for treatment of HIV infection [WO 91/00360; WO 92/200373; EP 03089]. It is contemplated that the antibodies may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No.
  • a multivalent antibody may be internalized (and/or catabolized) faster than a bivalent antibody by a cell expressing an antigen to which the antibodies bind.
  • the antibodies of the present invention can be multivalent antibodies (which are other than of the IgM class) with three or more antigen binding sites (e.g., tetravalent antibodies), which can be readily produced by recombinant expression of nucleic acid encoding the polypeptide chains of the antibody.
  • the multivalent antibody can comprise a dimerization domain and three or more antigen binding sites.
  • the dimerization domain comprises (or consists of) an Fc region or a hinge region. In this scenario, the antibody will comprise an Fc region and three or more antigen binding sites amino-terminal to the Fc region.
  • the multivalent antibody herein comprises (or consists of) three to about eight, but typically four, antigen binding sites.
  • the multivalent antibody comprises at least one polypeptide chain (and typically two polypeptide chains), wherein the polypeptide chain(s) comprise two or more variable domains.
  • the polypeptide chain(s) may comprise VDl-(Xl).sub.n-VD2-(X2).sub.n-Fc, wherein VDl is a first variable domain, VD2 is a second variable domain, Fc is one polypeptide chain of an Fc region, XI and X2 represent an amino acid or polypeptide, and n is 0 or 1.
  • the polypeptide chain(s) may comprise: VH- CHl-flexible linker-VH-CHl-Fc region chain; or VH-CHl -VH-CHl-Fc region chain.
  • the multivalent antibody herein may further comprise at least two (and typically four) light chain variable domain polypeptides.
  • the multivalent antibody herein may, for instance, comprise from about two to about eight light chain variable domain polypeptides.
  • the light chain variable domain polypeptides contemplated here comprise a light chain variable domain and, optionally, further comprise a CL domain.
  • ADCC antigen-dependent cell-mediated cytotoxicity
  • CDC complement dependent cytotoxicity
  • cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region.
  • the homodimeric antibody thus generated may have improved internalization capability and/or increased complement- mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al, J. Exp Med. 176: 1191 -1195 (1992) and Shopes, B. J. Immunol. 148:2918-2922 (1992).
  • Conjugation will ordinarily be achieved through a covalent linkage, the precise nature of which will be determined by the targeting molecule and the linking site on the integrin beta7 antagonist or antibody polypeptide.
  • a non-peptidic agent is modified by the addition of a linker that allows conjugation to anti-beta7 integrin antibody through its amino acid side chains, carbohydrate chains, or reactive groups introduced on antibody by chemical modification.
  • a drug may be attached through the .epsilon.
  • nucleic acids encoding the anti-beta7 antibodies or polypeptide agents described herein, vectors and host cells comprising the nucleic acids and recombinant techniques for the production of the antibodies.
  • the nucleic acid encoding it may be isolated and inserted into a replicable vector for further cloning (amplification of the DNA) or for expression.
  • the antibody may be produced by homologous recombination, e.g., as described in U.S. Patent No. 5,204,244, specifically incorporated herein by reference.
  • DNA encoding the monoclonal antibody is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody). Many vectors are available.
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above.
  • Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B.
  • Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus
  • Salmonella e.g., Salmonella typhimurium
  • Serratia e.g.,
  • Suitable host cells for the expression of glycosylated anti-Beta7 antibody are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified.
  • the host cells used to produce the anti-beta7 integrin antibody of this invention may be cultured in a variety of media.
  • Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma) are suitable for culturing the host cells.
  • the antibody When using recombinant techniques, the antibody can be produced
  • gamma.1 , .gamma.2, or .gamma.4 heavy chains (Lindmark et al, J. Immunol. Meth. 62: 1 -13 (1983)). Protein G is recommended for all mouse isotypes and for human .gamma.3 (Guss et al, EMBO J. 5 : 15671575 (1986)).
  • the matrix to which the affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices such as controlled pore glass or poly(styrenedivinyl)benzene allow for faster flow rates and shorter processing times than can be achieved with agarose.
  • the mixture comprising the antibody of interest and contaminants may be subjected to low pH hydrophobic interaction chromatography using an elution buffer at a pH between about 2.5-4.5, typically performed at low salt concentrations (e.g., from about 0-0.25 M salt).
  • the active ingredients may also be entrapped in microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin
  • microspheres microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the immunoglobulin of the invention, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Patent No. 3,773,919), copolymers of L-glutamic acid and .gamma.
  • sustained-release preparations include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Patent No. 3,773,919), copolymers of L-glutamic acid and .gamma.
  • the physician administering treatment will be able to determine the appropriate dose for the individual subject for weight-based dosing or, for flat dosing, will follow the instructions on the label.
  • Preparation and dosing schedules for commercially available second therapeutic and other compounds administered in combination with the integrin beta7 antagonists may be used according to manufacturers' instructions or determined empirically by the skilled practitioner.
  • the appropriate dosage of the integrin beta7 antagonist and any second therapeutic or other compound administered in combination with the non-depleting antibody will depend on the type of gastrointestinal inflammatory disorder to be treated, e.g., IBD, UC, CD, the severity and course of the disease, whether the integrin beta7 antagonist or combination is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the integrin beta7 antagonist or combination, and the discretion of the attending physician.
  • the integrin beta7 antagonist or combination is suitably administered to the patient at one time or more typically over a series of treatments.
  • the integrin beta7 antagonist is administered once every week, or once every two weeks, or once every four weeks, or once every six weeks, or once every eight weeks for a period of one month (4 weeks), or two months, three months, or six months, or 12 months, or 18 months, or 24 months, or chronically for the lifetime of the patient.
  • the treatment is self-administered by the patient.
  • anti-beta7 antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • the treatment is sustained until a desired suppression of disease symptoms occurs.
  • other dosage regimens may be useful.
  • an initial flat loading dose of anti-beta7 antibody is followed by one or more flat maintenance doses of anti-beta7 antibody.
  • the loading dose is a larger quantity of anti-beta7 antibody than the maintenance dose.
  • the loading dose is between about 400 mg and 450 mg and the maintenance dose is between about 50 mg and 350 mg.
  • the loading dose is 400 mg, or 420 mg, or 430 mg, or 450 mg.
  • the maintenance dose is 50 mg, or 100 mg, or 105 mg, or 150 mg, or 200 mg, or 210 mg, or 300 mg, or 315 mg or 350 mg.
  • the clinician will administer an antibody (alone or in combination with a second compound) of the invention until a dosage(s) is reached that provides the required biological effect.
  • an antibody alone or in combination with a second compound of the invention.
  • the progress of the therapy of the invention is easily monitored by conventional techniques and assays.
  • each exposure may be provided using the same or a different administration means.
  • each exposure to anti-beta7 antibody is by subcutaneous administration.
  • the first exposure to anti-beta7 antibody, e.g., the loading dose is by intravenous administration and each subsequent exposure is by subcutaneous administration.
  • an anti-beta7 antibody is administered using, for example, a self-inject device, autoinjector device, or other device designed for self- administration.
  • a self-inject device for example, a self-inject device, autoinjector device, or other device designed for self- administration.
  • autoinjector device for example, a self-inject device, autoinjector device, or other device designed for self- administration.
  • Various self-inject devices, including autoinjector devices are known in the art and are commercially available.
  • Exemplary devices include, but are not limited to, prefilled syringes (such as BD HYPAK SCF®, READYFILLTM, and STERIFILL SCFTM from Becton Dickinson; CLEARSHOTTM copolymer prefilled syringes from Baxter; and Daikyo Seiko CRYSTAL ZENITH® prefilled syringes available from West Pharmaceutical Services); disposable pen injection devices such as BD Pen from Becton Dickinson; ultra-sharp and microneedle devices (such as INJECT-EASETM and microinfuser devices from Becton Dickinson; and H-PATCHTM available from Valeritas) as well as needle-free injection devices (such as BIOJECTOR® and IJECT® available from Bioject; and SOF-SERTER® and patch devices available from Medtronic).
  • prefilled syringes such as BD HYPAK SCF®, READYFILLTM, and STERIFILL SCFTM
  • the integrin beta7 antagonist can be administered alone or in combination with at least a second therapeutic compound.
  • second therapeutic compounds are generally used in the same dosages and with administration routes as used heretofore, or about from 1 to 99% of the heretofore-employed dosages. If such second compounds are used, they are used in certain embodiments in lower amounts than if the integrin beta7 antagonist were not present, so as to eliminate or reduce side effects caused thereby.
  • a variety of suitable second therapeutic compounds for the treatment of IBD e.g., ulcerative colitis and Crohn's disease are known in the art, and dosages and administration methods for such second therapeutic compounds have likewise been described.
  • Administration of the integrin beta7 antagonist and any second therapeutic compound can be done simultaneously, e.g., as a single composition or as two or more distinct compositions using the same or different administration routes. Alternatively, or additionally, the administration can be done sequentially, in any order. In certain embodiments, intervals ranging from minutes to days, to weeks to months, can be present between the administrations of the two or more compositions.
  • the integrin beta7 antagonist may be administered first, followed by the second therapeutic compound.
  • simultaneous administration or administration of the second therapeutic compound prior to the integrin beta7 antagonist is also contemplated.
  • the treatment of the present invention for inflammatory bowel disease (IBD) in a human subject with IBD comprises administering to the subject an effective amount of an therapeutic agent, such as an anti-beta7 integrin antibody, and further comprising administering to the subject an effective amount of a second medicament, that is an immunosuppressant, a pain-control agent, an antidiarrheal agent, an antibiotic, or a
  • an therapeutic agent such as an anti-beta7 integrin antibody
  • a second medicament that is an immunosuppressant, a pain-control agent, an antidiarrheal agent, an antibiotic, or a
  • said secondary medicine is selected from the group consisting of an aminosalicylate, an oral corticosteroid, 6-mercaptopurine (6-MP) and azathioprine.
  • said secondary medicine is another integrin beta7 antagonist, such as another anti-beta7 integrin antibody or an antibody against a cytokine.
  • second medicaments may be used in combination with each other or by themselves with the first medicament, so that the expression "second medicament” as used herein does not mean it is the only medicament besides the first medicament, respectively.
  • the second medicament need not be one medicament, but may constitute or comprise more than one such drug.
  • Combined administration herein includes co-administration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein generally there is a time period while both (or all) active agents simultaneously exert their biological activities.
  • the combined administration of a second medicament includes coadministration (concurrent administration), using separate formulations or a single
  • PK Pharmacokinetics
  • PD Pharmacodynamics
  • PK/PD modeling establishes a mathematical and theoretical link between these two processes and helps better predict drug action.
  • Integrated PK/PD modeling and computer-assisted trial design via simulation are being incorporated into many drug development programs and are having a growing impact (Lakshmi Kamath, Drug Discovery and Development; Modeling Success in PK/PD Testing Drug Discovery &
  • PK/PD testing is typically performed at every stage of the drug development process. Because development is becoming increasingly complex, time consuming, and cost intensive, companies are looking to make better use of PK/PD data to eliminate flawed candidates at the beginning and identify those with the best chance of clinical success.
  • PK/PD modeling approaches are proving useful in determining relationships between biomarker response, drug levels, and dosing regimens.
  • the PK/PD profile of a drug candidate and the ability to predict a patient's response to it are critical to the success of clinical trials.
  • Recent advances in molecular biology techniques and a better understanding of targets for various diseases have validated biomarkers as a good clinical indicator of a drug's therapeutic efficacy.
  • Biomarker assays (including those described herein) and use of such biomarker assays help identify a biological response to a drug candidate. Once a biomarker is clinically validated, trial simulations can be effectively modeled. Biomarkers have the potential to achieve surrogate status that may someday substitute for clinical outcomes in drug development. (Lakshmi Kamath, supra).
  • biomarkers in blood such as those described herein can be used in identifying the biological response to a treatment with integrin beta7 antagonists and can therefore function as a good clinical indicator for the therapeutic efficacy of a candidate treatment.
  • PK/PD modeling in drug development defines parameters such as drug dose concentration, drug exposure effects, drug half-life, drug concentrations against time, and drug effects against time.
  • quantitative techniques such as drug modeling, disease modeling, trial modeling, and market modeling can support the entire development process, which results in better decisions through explicit consideration of risk and better utilization of knowledge.
  • PKS Knowledgebase Server
  • Primers, oligonucleotides and polynucleotides employed in the present invention can be generated using standard techniques known in the art.
  • Biomarkers associated with predicting responsiveness of IBD patients including patient suffering from UC or Crohn's disease to certain therapeutic agents are provided herein. Accordingly, the invention disclosed herein is useful in a variety of settings, e.g., in methods and compositions related to diagnosis and therapy of inflammatory bowel diseases.
  • the biomarkers provided herein including T cells, Treg cells, and Thl7 cells
  • T cells may be detected using methods in the art.
  • the T cells, Treg cells, and/or Thl7 cells are detecting using epigenetic analysis, which detects the methylation state of genomic DNA.
  • T cells are identified as cells in blood that have a demethylated region in the CD3D/G gene.
  • Treg cells are identified as cells in blood that have a demethylated region in the FOXP3 gene.
  • the demethylated region in the FOXP3 gene is the Treg specific demethylated region (TSDR).
  • the Thl7 cells are identified as cells in blood that have a demethylated region in the IL17 A gene.
  • Nonlimiting exemplary methods of identifying and quantifying T cells, Treg cells, and/or Thl7 cells using epigenetic analysis are described, e.g., in Wieczorek et al., 2009, Cancer Res. 69: 599-608; Sehouli et al, 2011, Epigenetics 6: 236-246; Turbachova et al , 2013, Epigenetics, 8: 11, 1226-1235; PCT Publication No. WO 2013/057202; and European Publication No. EP 1 826 279.
  • genomic DNA may be isolated from a sample, for example, whole blood or peripheral blood mononucleocytes (PBMCs).
  • PBMCs peripheral blood mononucleocytes
  • the genomic DNA is subject to bisulfite conversion, which converts unmethylated C to U (which is typically replaced by T during amplification), but leaves methylated C as C.
  • Kits for bisulfite conversion of genomic DNA are commercially available from various suppliers, and bisulfite conversion is described, e.g., in Olek et al, 1996, Nulc. Acids Res. , 54: 5064-5066.
  • sequence of the unmethylated genomic DNA is altered relative to the sequence of the methylated DNA.
  • Sequence-specific quantitative PCR may be used to quantify the number of copies of unmethylated genomic DNA. Since most genes are present at two copies per cell (for example, if located on an autosome), the number of cells having the demethylated gene will be 1 ⁇ 2 the number of copies of the gene detected. In some embodiments, if the gene is located on the X chromosome, it will be present at two copies per cell in women and one copy per cell in men. Accordingly, for an X-linked gene, the correlation between the number of cells having the demethylated gene and the number of copies of the demethylated gene detected will be 1 : 1 for men and 1 :2 for women.
  • the number of copies is determined by comparison to a standard curve.
  • Quantitative PCR is described, e.g., in “PCR Protocols, A Guide to Methods and Applications,” (M.A. Innis et al, eds., Academic Press, Inc., 1990); “Current Protocols in Molecular Biology” (F. M. Ausubel et al, eds., 1987, and periodic updates); and “PCR: The Polymerase Chain Reaction", (Mullis et al, eds., 1994).
  • quantitative PCR is also used to quantify the total number of cells in the sample, for example, by quantifying the number of copies of a control gene, independent of methylation state.
  • the total number of ceils is determined by quantifying the number of copies of a housekeeping gene.
  • housekeeping genes that may be used to quantity the total number of cells in a sample include, but are not limited to, GAPDH, ACTB, and UBC.
  • primers and probes for detecting bisulfite-converted demethylated genes are discussed below. It is understood that the sequences of the primers and/or probes may differ from the genomic such that they are specific for the bisulfite-converted sequence.
  • Nonlimiting exemplary primers and probe for detecting bisulfite-converted demethylated FOXP3 gene include a forward primer corresponding to a region within chromosomal region X:49004163-49004191 : 1 and a reverse primer corresponding to a region within chromosomal region NCBI36:X:49004227 -49004251 : 1.
  • a nonlimiting exemplar ⁇ 7 probe corresponds to a region within chromosomal region X: 49004200-49004222: 1. See, e.g., Wieczorek et al. , 2009, Cancer Res. 69: 599-608.
  • Nonlimiting exemplar ⁇ ' primers and probe for detecting bisulfite-converted demethylated FOXP3 gene include a forward primer corresponding to a region within chromosomal region X: 491 17219-49117246: 1 and a reverse primer corresponding to a region within chromosomal region X:491 17283-49117307: 1.
  • a nonlimiting exemplar ⁇ ' probe corresponds to a region within chromosomal region X:49117256-
  • Nonlimiting exemplar ⁇ " primers and probe for detecting bisulfite-converted demethylated ILI 7A gene include:
  • IL17A forward primer A 5 ' - ATTCTTCTATAACCTC ATTAAAAGC A-3 ' (SEQ ID NO: 33);
  • IL17A forward primer IL17A-B 5 ' -TTCTTCTATAACCTCATTAAAAGCAA-3 ' (SEQ ID NO: 34);
  • IL17A reverse primer A 5 ' -GAGATGGATAAAATGTAGTGTTATT-3 ' (SEQ ID NO: 35);
  • IL17A reverse primer B 5 ' -GATGGATAAAATGTAGTGTTATTG-3 ' (SEQ ID NO: 36);
  • 1L17A probe 5'-CCACTACAACACACCACATAAAT-3' (SEQ ID NO: 37).
  • detecting bisulfite-converted demethylated IL17A gene comprises IL17A forward primer A, IL17A reverse primer B, and IL17A probe. See, e.g., PCT
  • Nonlimiting exemplar ⁇ ' primers and probe for detecting bisulfite-converted demethylated CD3D/G gene include a forward primer corresponding to a region within chromosomal region 11 : 1 18213632- 1 J 8213653: 1 and a reverse primer corresponding to a region within chromosomal region 11 : 118213686-118213709: 1.
  • a nonlimiting exemplar ⁇ ' probe corresponds to a region within chromosomal region 1 1 : 118213664-1 18213690: 1. See, e.g., Sehouli et al, 201 1 , Epigenetics, 6: 236-246.
  • Nonlimiting exemplary primers and probe for detecting GAPDH include a forward primer corresponding to a region within chromosomal region 12:6644378-6644399: 1 and a reverse primer corresponding to a region within chromosomal region 12:6644456-6644476: 1.
  • a nonlimiting exemplary probe corresponds to a region within chromosomal region
  • a biological sample may be obtained using certain methods known to those skilled in the art.
  • Biological samples may be obtained from vertebrate animals, and in particular, mammals.
  • a biological sample is whole blood or peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • a blood sample may contain a preservative and/or anti-coagulant, including, but not limited to, EDTA, citrate, and/or heparin.
  • the blood sample may also be a fresh sample or a previously frozen sample.
  • about 1 ml of whole blood is used for analysis.
  • about 500 ⁇ , or about 400 ⁇ , or about 300 ⁇ , or about 200 ⁇ , or about 100 ⁇ , or about 50 ⁇ of whole blood is used for analysis.
  • an effective amount of an appropriate therapeutic agent may be administered to the subject to treat the particular disease in the subject, e.g., UC or Crohn's disease.
  • Clinical diagnosis in mammals of the various pathological conditions described herein can be made by the skilled practitioner.
  • Clinical diagnostic techniques are available in the art which allow, e.g., for the diagnosis or detection of inflammatory bowel diseases in a mammal, e.g., ulcerative colitis and Crohn's disease.
  • kits or articles of manufacture are also provided.
  • Such kits may comprise a carrier means being
  • one of the container means may comprise a probe that is or can be detectably labeled.
  • probe may be a polynucleotide specific for a demethylated region of one or more genes that is indicative of a cell type.
  • probe may be a
  • kits may also have containers containing nucleotide(s) for amplification of the target nucleic acid sequence and/or a container comprising a reporter means, such as a biotin- binding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, florescent, or radioisotope label.
  • a reporter means such as a biotin- binding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, florescent, or radioisotope label.
  • Kits will typically comprise the container described above and one or more other containers comprising materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • a label may be present on the container to indicate that the composition is used for a specific therapy or non-therapeutic application, and may also indicate directions for either in vivo or in vitro use, such as those described above.
  • Other optional components in the kit include one or more buffers (e.g.
  • substrate e.g., chromogen
  • control samples positive and/or negative controls
  • control slide(s) etc.
  • the invention herein also encompasses a method for marketing a therapeutic agent or a pharmaceutically acceptable composition thereof comprising promoting to, instructing, and/or specifying to a target audience, the use of the agent or pharmaceutical composition thereof for treating a patient or patient population with a particular disease, e.g., UC or Crohn's disease, from which a sample has been obtained showing levels of biomarkers disclosed herein.
  • a particular disease e.g., UC or Crohn's disease
  • Marketing is generally paid communication through a non-personal medium in which the sponsor is identified and the message is controlled.
  • Marketing for purposes herein includes publicity, public relations, product placement, sponsorship, underwriting, and sales promotion. This term also includes sponsored informational public notices appearing in any of the print communications media designed to appeal to a mass audience to persuade, inform, promote, motivate, or otherwise modify behavior toward a favorable pattern of purchasing, supporting, or approving the invention herein.
  • the marketing of the diagnostic method herein may be accomplished by any means.
  • marketing media used to deliver these messages include television, radio, movies, magazines, newspapers, the intemet, and billboards, including commercials, which are messages appearing in the broadcast media.
  • the type of marketing used will depend on many factors, for example, on the nature of the target audience to be reached, e.g., hospitals, insurance companies, clinics, doctors, nurses, and patients, as well as cost considerations and the relevant jurisdictional laws and regulations governing marketing of medicaments and diagnostics.
  • the marketing may be individualized or customized based on user characterizations defined by service interaction and/or other data such as user demographics and geographical location.
  • RhuMAb Beta7 (etrolizumab) is a humanized monoclonal antibody based on the human IgGl subgroup III VH, K subgroup-I VL consensus sequences and is directed specifically against the ⁇ 7 subunit of the integrin heterodimer. See Figs. 1 A and B. It has been shown to bind with high affinity to ⁇ 4 ⁇ 7 (Kd of about 116 pM) and ⁇ 7 (Kd of about 1800 pM).
  • This recombinant antibody has two heavy chains (446 residues) and two light chains (214 residues) that are covalently linked by interchain and intrachain disulfide bonds typical of IgGl antibodies.
  • it was produced in Chinese hamster ovary (CHO) cells.
  • the molecular mass of the intact, non glycosylated rhuMAb Beta7 molecule was approximately 144 kDa.
  • Each heavy chain of rhuMAb Beta7 has one conserved N linked glycosylation site at Asn297.
  • the oligosaccharides present at this site were typical of those observed in recombinant antibodies expressed in CHO cells, with the predominant glycoforms being the asialo, biantennary GO, and Gl glycans.
  • the mass of the most prevalent rhuMAb Beta7 form containing two GO glycans and no C terminal lysine residues was approximately 147 kDa.
  • RhuMAb Beta7 drug product and placebo were prepared by Genentech. They were clear to slightly opalescent, colorless to slightly yellow aqueous solutions. Both solutions were sterile and preservative-free liquid intended for IV and SC administration. Study Design
  • This Phase II study was a randomized, double-blind, placebo-controlled multicenter study to evaluate the efficacy and safety across two rhuMAb Beta7 dose levels compared with placebo in patients with moderate to severe UC.
  • the primary efficacy endpoint was evaluated at Week 10 (2 weeks after the final dose of study drug was administered) with a secondary efficacy endpoint at Week 6.
  • the dose values provided in the preceding paragraph are nominal doses.
  • the phase II dose administration used a vial and a syringe with a vial concentration of 150 mg/ml.
  • 0.7 ml was the selected volume per subcutaneous (SC) injection.
  • the actual drug amount therefore, for the nominal 100 mg dose arm was 105 mg (1 x 0.7 ml SC injection) and for the nominal 300 mg dose was 315 mg (3 x 0.7 ml SC injections).
  • the actual loading dose of 420 mg was 420 mg (4 x 0.7 ml SC injections). All SC injections were administered into the abdomen. Accordingly, a dose of "100 mg" and a dose of "105 mg” are used interchangeably herein.
  • the daily bleeding score represents the most severe bleeding of the day.
  • the physician's global assessment acknowledges the three other criteria, the patient's daily recollection of abdominal discomfort and general sense of well-being, and other observations, such as physical findings and the patient's performance status.
  • azathioprine [AZA], 6-mercaptopurine [6-MP], or methotrexate) doses must have been kept stable for at least 4 weeks prior to randomization on Day 1. Patients who were receiving topical 5-ASA or corticosteroids must have discontinued 2 weeks before randomization on Day 1. Oral corticosteroid doses must have been kept stable for 2 weeks prior to randomization on Day 1. Patients receiving high-dose steroids must have had the dose reduced to ⁇ 20 mg/day for 2 weeks prior to randomization on Day 1.
  • immunosuppressants must have been commenced at Week 8, and patients must have completely discontinued immunosuppressants by Week 10. Patients who have previously received anti-TNF therapy must have discontinued therapy for a minimum of 8 weeks prior to randomization to receive study drug on Day 1. If patients experienced persisting or increasing disease activity at any time during the study, rescue therapy in the form of an increase in steroids and or immunosuppressant dose may be increased or initiated according to the investigator's clinical judgment. Patients who required rescue therapy were permitted to remain in the study but discontinued study treatment and, during data analysis, were classified as having experienced treatment failure.
  • TNF-IR loss of response and/or intolerance and/or inadequate response to anti-TNF agents
  • UC disease activity was assessed using the MCS at Screening (and this was considered the baseline MCS), Week 6 (2 weeks after dosing at Week 4), and Week 10 (2 weeks after the final dose of study drug). Biopsies of the colon were obtained during the flexible sigmoidoscopy conducted at these same time points. Partial MCS was also collected throughout the study. Patient Reported Outcomes (PROs) were also assessed by using a Short Inflammatory Bowel Disease Questionnaire (SIBDQ) and MCS, which were to be completed by patients at Day 1 and at Weeks 6 and 10.
  • SIBDQ Short Inflammatory Bowel Disease Questionnaire
  • disease activity, daily symptoms, and impact of UC were collected in a patient diary, to be completed daily by patients from screening (approximately 7 days prior to and up to Day 1) and at least 7 days prior to and up to the study visits at Weeks 6 and 10.
  • Serum and fecal samples were also obtained for biomarker analysis. Stool was obtained at screening and Weeks 6, 10, and 28 for measurement of biomarkers. Exemplary biomarkers that were considered for measurement include, but are not limited to, lipocalin, calprotectin, and lactorferrin.
  • Serum and plasma were obtained at screening, at Day 1, and at Weeks 4, 6, 10, 16, and 28 for measurement of exploratory biomarkers.
  • the primary efficacy outcome measure was clinical remission at Week 10.
  • the exploratory outcome measure for this study were the time to flare of UC in patients who achieved response or remission.
  • a flare is defined as a 2 point increase in partial MCS accompanied by 3 days of continuous rectal bleeding and an endoscopy score of 2 on flexible sigmoidoscopy.
  • peripheral levels of all three cell types were relatively stable over time in the placebo group, whereas etrolizumab-treated patients showed a trend towards an increase for all three cell types.
  • the median increase for T cell, Treg, and Thl7 cells for the 300 mg dose group compared to placebo was significant at day 29 (P ⁇ 0.05).
  • Figure 6 shows the change from baseline (study day 0) in the percentage of T cells (Fig.
  • C7 are QQGNSLP
  • C8 is selected from the group antibody consisting of N, V, W, Y, R, S, T, A, F, H, I L, and M
  • C9 is T
  • Fl anti-beta7 F2-F11 or Fl-Fl l
  • F2 is R, M, A, E, G, Q, S
  • F3 to F10 is antibody TGSSGYFD
  • Fl 1 is selected from the group consisting of F and Y

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • Microbiology (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

L'invention porte sur des biomarqueurs prédictifs de la réactivité à des antagonistes de l'intégrine bêta 7, y compris à des anticorps de sous-unités de l'intégrine anti-bêta 7, et sur des procédés d'utilisation de tels biomarqueurs. De plus, l'invention concerne des méthodes de traitement de troubles inflammatoires gastro-intestinaux, tels que les maladies inflammatoires de l'intestin, y compris la colite ulcéreuse et la maladie de Crohn. L'invention concerne également des méthodes d'utilisation de tels biomarqueurs prédictifs pour le traitement de maladies inflammatoires de l'intestin, y compris de la colite ulcéreuse et de la maladie de Crohn.
PCT/US2016/044022 2015-07-27 2016-07-26 Méthodes permettant de diagnostiquer et de traiter une maladie inflammatoire de l'intestin WO2017019673A2 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2018503499A JP2018521661A (ja) 2015-07-27 2016-07-26 炎症性腸疾患の診断及び治療方法
CN201680055933.8A CN108139385A (zh) 2015-07-27 2016-07-26 用于诊断和治疗炎症性肠病的方法
EP16750314.3A EP3329276A2 (fr) 2015-07-27 2016-07-26 Méthodes de diagnostic et de traitement de la maladie intestinale inflammatoire

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562197293P 2015-07-27 2015-07-27
US62/197,293 2015-07-27
US201562241331P 2015-10-14 2015-10-14
US62/241,331 2015-10-14

Publications (2)

Publication Number Publication Date
WO2017019673A2 true WO2017019673A2 (fr) 2017-02-02
WO2017019673A3 WO2017019673A3 (fr) 2017-06-08

Family

ID=56618256

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/044022 WO2017019673A2 (fr) 2015-07-27 2016-07-26 Méthodes permettant de diagnostiquer et de traiter une maladie inflammatoire de l'intestin

Country Status (4)

Country Link
EP (1) EP3329276A2 (fr)
JP (1) JP2018521661A (fr)
CN (1) CN108139385A (fr)
WO (1) WO2017019673A2 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110283889B (zh) * 2019-07-22 2020-10-13 中南大学 一种双基因监控反应体系、试剂盒及其应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2322556A1 (fr) * 2004-09-03 2011-05-18 Genentech, Inc. Antagonistes anti-béta 7 humanisés et utilisations associées
WO2013037970A1 (fr) * 2011-09-15 2013-03-21 Nogra Pharma Limited Méthodes pour surveiller la réactivité à un traitement anti-smad7
WO2014160753A1 (fr) * 2013-03-27 2014-10-02 Genentech, Inc. Utilisation de biomarqueurs pour évaluer le traitement de troubles gastro-intestinaux inflammatoires par des antagonistes de l'intégrine bêta7

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2322556A1 (fr) * 2004-09-03 2011-05-18 Genentech, Inc. Antagonistes anti-béta 7 humanisés et utilisations associées
WO2013037970A1 (fr) * 2011-09-15 2013-03-21 Nogra Pharma Limited Méthodes pour surveiller la réactivité à un traitement anti-smad7
WO2014160753A1 (fr) * 2013-03-27 2014-10-02 Genentech, Inc. Utilisation de biomarqueurs pour évaluer le traitement de troubles gastro-intestinaux inflammatoires par des antagonistes de l'intégrine bêta7

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ANIKA FISCHER ET AL: "Differential effects of [alpha]4[beta]7 and GPR15 on homing of effector and regulatory T cells from patients with UC to the inflamed gut in vivo", GUT, vol. 65, no. 10, 24 July 2015 (2015-07-24), UK, pages 1642 - 1664, XP055306066, ISSN: 0017-5749, DOI: 10.1136/gutjnl-2015-310022 *
DATABASE BIOSIS [online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; 2003, HART AILSA ET AL: "INTESTINAL HOMING MEMORY T LYMPHOCYTES PRODUCE INCREASED TUMOUR NECROSIS FACTOR-ALPHA DURING RELAPSE OF ULCERATIVE COLITIS: PROSPECTIVE STUDY OF PATIENTS IN REMISSION AND AT RELPASE.", XP002768300, Database accession no. PREV200300572617 *
DATABASE BIOSIS [online] BIOSCIENCES INFORMATION SERVICE, PHILADELPHIA, PA, US; April 2015 (2015-04-01), LORD JAMES D ET AL: "Effect of Vedolizumab on Circulating Lymphocyte Subsets in IBD James D. Lord, Donna Shows", XP002768299, Database accession no. PREV201500715500 *
FRANKLIN FUH: "Inflammatory Bowel Disease", AMERICAN JOURNAL OF GASTROENTEROLOGY, vol. 110, 21 October 2015 (2015-10-21), US, pages S766 - S847, XP055306057, ISSN: 0002-9270, DOI: 10.1038/ajg.2015.276 *

Also Published As

Publication number Publication date
CN108139385A (zh) 2018-06-08
JP2018521661A (ja) 2018-08-09
WO2017019673A3 (fr) 2017-06-08
EP3329276A2 (fr) 2018-06-06

Similar Documents

Publication Publication Date Title
US11091551B2 (en) Methods for diagnosing and treating inflammatory bowel disease
US11261493B2 (en) Methods for diagnosing and treating inflammatory bowel disease
AU2012236304C1 (en) Methods of administering beta7 integrin antagonists
WO2017019673A2 (fr) Méthodes permettant de diagnostiquer et de traiter une maladie inflammatoire de l'intestin
US20220403033A1 (en) Methods of administering beta7 integrin antagonists
US20210238288A1 (en) Methods of administering beta7 integrin antagonists

Legal Events

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

Ref document number: 16750314

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2018503499

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2016750314

Country of ref document: EP