US20040009169A1 - Administration of agents for the treatment of inflammation - Google Patents

Administration of agents for the treatment of inflammation Download PDF

Info

Publication number
US20040009169A1
US20040009169A1 US10/372,111 US37211103A US2004009169A1 US 20040009169 A1 US20040009169 A1 US 20040009169A1 US 37211103 A US37211103 A US 37211103A US 2004009169 A1 US2004009169 A1 US 2004009169A1
Authority
US
United States
Prior art keywords
alpha
integrin
natalizumab
disease
patient
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/372,111
Other languages
English (en)
Inventor
Julie Taylor
Theodore Yednock
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biogen MA Inc
Original Assignee
Julie Taylor
Yednock Theodore A.
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27767592&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20040009169(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US10/372,111 priority Critical patent/US20040009169A1/en
Application filed by Julie Taylor, Yednock Theodore A. filed Critical Julie Taylor
Publication of US20040009169A1 publication Critical patent/US20040009169A1/en
Priority to US11/540,640 priority patent/US7807167B2/en
Priority to US12/846,350 priority patent/US20110064729A1/en
Assigned to BIOGEN IDEC INTERNATIONAL HOLDING LTD. reassignment BIOGEN IDEC INTERNATIONAL HOLDING LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ELAN PHARMA INTERNATIONAL LIMITED
Assigned to BIOGEN IDEC INTERNATIONAL HOLDING LTD. reassignment BIOGEN IDEC INTERNATIONAL HOLDING LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ELAN PHARMACEUTICALS, INC.
Assigned to BIOGEN IDEC MA INC. reassignment BIOGEN IDEC MA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BIOGEN IDEC INTERNATIONAL HOLDING LTD.
Priority to US14/613,821 priority patent/US20150152182A1/en
Priority to US15/586,724 priority patent/US20170306026A1/en
Priority to US16/411,042 priority patent/US11248051B2/en
Priority to US17/578,294 priority patent/US20220162321A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • C07K16/2842Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily against integrin beta1-subunit-containing molecules, e.g. CD29, CD49
    • 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
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4737C-reactive protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70546Integrin superfamily, e.g. VLAs, leuCAM, GPIIb/GPIIIa, LPAM
    • G01N2333/7055Integrin beta1-subunit-containing molecules, e.g. CD29, CD49

Definitions

  • This invention relates generally to agents that specifically bind to and inhibit an integrin receptor comprising an alpha-4 ( ⁇ 4) subunit, and therapeutic uses of the same.
  • Inflammation is a response of vascularized tissues to infection or injury and is affected by adhesion of leukocytes to the endothelial cells of blood vessels and their infiltration into the surrounding tissues.
  • the infiltrating leukocytes release toxic mediators to kill invading organisms, phagocytize debris and dead cells, and play a role in tissue repair and the immune response.
  • infiltrating leukocytes are over-responsive and can cause serious or fatal damage. See, e.g., Hickey, Psychoneuroimmunology II (Academic Press 1990).
  • the integrins are a family of cell-surface glycoproteins involved in cell-adhesion, immune cell migration and activation.
  • Alpha-4 integrin is expressed by all circulating leukocytes except neutrophils, and forms heterodimeric receptors in conjunction with either the beta1 or beta7 integrin subunits; both alpha-4 beta-1 ( ⁇ 4 ⁇ 1) and alpha-4 beta-7 ( ⁇ 4 ⁇ 7) dimers play a role in the migration of leukocytes across the vascular endothelium (Springer et al., 1994 Cell 76: 301-14; and Butcher et al., 1996 Science 272: 60-6) and contribute to cell activation and survival within the parenchyma (Damle et al., 1993 J. Immunol. 151: 2368-79; Koopman et al., 1994 J. Immunol. 152: 3760-7; and Leussink et al., 2002 Acta Neuropathol. 103:131-136).
  • alpha-4 beta-1 also known as very late antigen-4 [VLA-4]
  • VLA-4 very late antigen-4
  • VCAM-1 vascular cell adhesion molecule-1
  • the alpha-4 beta-7 dimer interacts with mucosal addressin cell adhesion molecule (MAdCAM-1), and mediates homing of lymphocytes to the gut (Farstad et al., 1997 Am. J. Pathol. 150: 187-99; and Issekutz et al., 1991 J. Immunol. 147: 4178-84).
  • MAdCAM-1 mucosal addressin cell adhesion molecule
  • IBD inflammatory bowel disease
  • Adhesion molecules such as alpha-4 integrins are potential targets for therapeutic agents.
  • the VLA-4 receptor of which alpha-4 integrin is a subunit, is an important target because of its interaction with a ligand residing on brain endothelial cells. Diseases and conditions resulting from brain inflammation have particularly severe consequences.
  • the alpha-4 beta-7 integrin dimer is an important target due to its involvement in lymphocyte homing and pathological inflammation in the gastrointestinal tract.
  • Alpha-4 beta-1 integrin is expressed on the extracellular surface of activated lymphocytes and monocytes, which have been implicated in the pathogenesis of acute inflammatory brain lesions and blood brain barrier (BBB) breakdown associated with multiple sclerosis (MS) (Coles et al., 1999 Ann. Neurol. 46(3): 296-304).
  • BBB blood brain barrier
  • MS multiple sclerosis
  • Agents against alpha-4 integrin have been tested for their anti-inflammatory potential both in vitro and in vivo in animal models. See Yednock et al., 1992 Nature 356: 63-66; U.S. Pat. No. 5,840,299 issued to Bendig, et al. on Nov. 24, 1998, and U.S. Pat. No. 6,001,809 issued to Thorsett et al.
  • Crohn's disease is a chronic, incurable, relapsing, transmural inflammation of the intestinal tract.
  • the disease is characterized by inappropriate immune cell migration and activation in the intestinal mucosa involving T cells, macrophages and neutrophils (Schreiber et al., 1991 Gastroenterology 101: 1020-30).
  • First-line medical therapies for Crohn's disease include 5-aminosalicylates (5-ASAs), which have low efficacy, and corticosteroids, which have various short- and long-term side effects (Munkholm et al., 1994 Gut 35: 360-2).
  • the present invention provides methods of chronically reducing pathological inflammation in a patient via the chronic administration of an agent that selectively binds to alpha-4 integrin.
  • the chronic dosage regime of an alpha-4 agent is designed such that (1) the agent specifically binds to alpha-4 integrin or an integrin, dimer comprising alpha-4 integrin, and (2) the agent is administered repeatedly to maintain alpha-4 integrin receptor saturation at a sufficient level to suppress pathological inflammation.
  • the agent of the invention can be useful in the suppression of inflammation via binding and inhibition of all integrin dimers comprising the alpha-4 subunit, or it can be designed to bind to a specific dimer, e.g., alpha-4 beta-1.
  • the efficacy of a chronic dosage regime can be determined by the measurement of saturation of a specific integrin dimer.
  • a specific integrin dimer it is believed that alpha-4 beta-1 integrin dimer is involved in multiple sclerosis, and thus the level of saturation required for an efficacious chronic administration regime can be measured via measurement of the saturation of the alpha-4 beta-1 dimer receptor.
  • saturation of a combination of dimer receptors can be measured to determine the efficacy of a chronic administration regime.
  • both the alpha-4 beta-1 and the alpha-4 beta-7 dimers are believed to have involvement in pathological inflammation associated with inflammatory bowel disease.
  • measurement of both alpha-4 beta-1 and alpha-4 beta-7 saturation levels may be beneficial in determining the efficacy of a particular chronic administration regime.
  • the success of a chronic dosage regime can be determined by assessing a physiological marker of the pathological inflammation.
  • a physiological marker of the pathological inflammation For example, in a chronic dosage regime administered for MS, the success of the dosage regime can be confirmed by detection of the levels of brain lesions using an imaging technique, e.g., magnetic resonance imaging (MRI).
  • MRI magnetic resonance imaging
  • the success of the dosage regime can be confirmed by detection of the levels of serum C-reactive protein levels in a patient.
  • the success of the dosage regime can be determined using a set group of criteria associated with wellness, e.g., reduction in CDAI in a CD patient.
  • Another embodiment of the invention contemplates the treatment of an inflammatory disease of the gastrointestinal tract (e.g., Crohn's disease, ulcerative colitis and inflammatory bowel disease) of a subject comprising administering a therapeutically effective amount of a composition comprising natalizumab sufficient to treat or ameliorate said inflammatory disease of the gastrointestinal tract in said subject.
  • an inflammatory disease of the gastrointestinal tract e.g., Crohn's disease, ulcerative colitis and inflammatory bowel disease
  • the invention features a dosage regime wherein repeated administration of an agent is provided to provide a level of alpha-4 integrin receptor saturation of 65-100% in a patient, thereby providing chronic suppression of pathological inflammation in the patient.
  • the agent is repeatedly administered to provide levels of at least about 75-100% in a patient.
  • the agent in repeatedly administered to provide levels of at least about 80-100% in a patient.
  • a feature of the invention is that undesirable effects of pathological inflammation can be suppressed long-term in a patient, e.g., over a period of six months, one year, two years, or more.
  • An aspect of the method of the invention is that sufficient levels of an anti-alpha-4 integrin agent are maintained over long periods to suppress pathological inflammation over those periods.
  • a feature of the invention is that the dosage form provides lower levels of pathological inflammation over longer periods as compared to a single dose.
  • An advantage of the invention is that agents used in the methods of the invention are well tolerated and have low toxicity.
  • FIG. 1 is a line graph illustrating the cumulative mean number of new Gd-enhancing lesions in MS patients following dosing with natalizumab.
  • FIG. 2 shows the percentage of active scans at each time point during the natalizumab MS study. Active scans are those containing one or more new Gd-enhancing lesions.
  • FIGS. 3 A-C and 4 A-C show the serum concentrations of natalizumab following the dosage regime time points in an MS study.
  • FIGS. 3 A-C show the levels for the 3-mg/kg study;
  • FIGS. 4 A-C show the levels for the 6-mg/kg study.
  • FIGS. 5 A-F illustrates the levels of receptor saturation maintained in the MS study. Levels shown are percentage values.
  • FIGS. 6 and 7 show the percentage of Patients Achieving Predefined Criteria of Clinical Response (FIG. 6) or Remission (FIG. 7) following dosing in the CD study.
  • FIG. 8 illustrates mean changes in serum C-reactive protein in a subset of patients who had elevated C-reactive protein at base-line in the CD study.
  • FIGS. 9 A-B demonstrate respectively the effects of natalizumab on circulating eosinophils in patients with active Crohn's disease (CD) and ulcerative colitis (UC).
  • FIG. 9B shows that administration of natalizumab significantly increased monocyte counts in active Crohn's disease and ulcerative colitis patients after 3 mg/kg natalizumab infusion.
  • FIGS. 10 A-D demonstrate the impact of natalizumab administration on TCR ⁇ + cells expressing activation antigen.
  • Panel A demonstrates the effect of natalizumab in patients with active Crohn's disease, i.e., a significant increase of TCR ⁇ + cells expressing CD26, HLA-DR, CD8CR and CD8 CD28 to at least four weeks post 3 mg/kg natalizumab infusion.
  • Panel B demonstrates the effects of natalizumab on TCR ⁇ + cells expressing activation antigens in patients with activated ulcerative colitis, i.e., significant increase in TCR ⁇ + cells expressing CD26, HLA-DR, CD8DR and CD8CD28 to at least four weeks post 3 mg/kg natalizumab in fusion.
  • Panel C demonstrates the effects of natalizumab administration on TCR ⁇ + cells expressing activation antigens and memory and naive markers in patients with active Crohn's disease, i.e., significant increase of memory (CD45RO) and naive (CD45RA) TCR ⁇ + cells to at least four weeks post-natalizumab infusion.
  • Panel B shows the effects of natalizumab on TCR ⁇ + cells expressing activation antigens, memory and naive markers in patients with active ulcerative colitis, i.e., significant increase of memory (CD45RO), naive (CD45RA), CD69 and CD38 TCR ⁇ + cells at one week post natalizumab administration.
  • FIGS. 11 A-B demonstrate the effect of natalizumab on circulating TCR ⁇ + and NK-type cells in patients with active Crohn's disease and with ulcerative colitis respectively.
  • anti-alpha-4 agent refers to any agent, which binds specifically to an integrin comprising an alpha-4 subunit and inhibits activity of the integrin.
  • agents that specifically bind to alpha-4 integrin, as well as agents that bind to an integrin dimer that comprises the alpha-4 integrin, e.g., alpha-4 beta-1 ( ⁇ 4 ⁇ 1) or alpha-4 beta-7 ( ⁇ 4 ⁇ 7).
  • agents is meant to include synthetic and recombinant molecules (e.g., antibodies, small molecules, peptides, or other synthetically produced molecules or compounds, as well as recombinantly produced gene products) as well as naturally-occurring compounds (e.g., polypeptides, antibodies, and the like).
  • efficacy refers to the effectiveness of a particular treatment regime. Efficacy can be measured based on change the course of the disease in response to an agent of the present invention. For example, in the treatment of MS, efficacy can be measured by the frequency of relapses in relapsing-remitting MS, and by the presence or absence of new lesions in the central nervous system as detected using methods such as MRI.
  • uccess refers to the effectiveness of a particular treatment regime. This includes a balance of efficacy, toxicity (e.g., side effects and patient tolerance of a formulation or dosage unit), patient compliance, and the like. For a chronic administration regime to be considered “successful” it must balance different aspects of patient care and efficacy to produce the most favorable patient outcome.
  • the terms “specifically binds” or “binds specifically” as used herein refer to the situation in which one member of a specific binding pair will not show any significant binding to molecules other than its specific binding partner (e.g., an affinity of about 1,000 ⁇ or more for its binding partner).
  • the anti-alpha-4 integrin agent will not show significant binding to any polypeptide other than an alpha-4 integrin or a receptor comprising an alpha-4 integrin.
  • antibodies used in the methods of the invention that bind to an alpha-4 integrin with a binding affinity of 10 7 mole/l or more, preferably 10 8 mole/liters or more, are said to bind specifically to an alpha-4 integrin.
  • substantially homologous as used herein is intended to mean any polypeptide that has an alteration in the sequence such that a functionally equivalent amino acid is substituted for one or more amino acids in the polypeptide, thus producing a change that has no or relatively little effect on the binding properties of the polypeptide.
  • one or more amino acid residues within the sequence can be substituted by another amino acid of a similar polarity.
  • the terms “elicits an immune response” and “elicits a host immune response” as used herein refer to the production of an immunological response to a receptor comprising an alpha-4 integrin in a subject upon introduction of an agent of the invention to the subject.
  • An immune response in the subject can be characterized by serum reactivity with an alpha-4 integrin receptor that is at least twice that of an untreated subject, more preferably three times the reactivity of an untreated subject, and even more preferably at least four times the reactivity of an untreated subject, with serum immunoreactivity measured using a serum dilution of approximately 1:00.
  • excipient material is intended to mean any compound forming a part of the formulation that is intended to act merely as a carrier, i.e. not intended to have biological activity itself.
  • adjuvant refers to a composition additive that augments the immune response to an agent of the invention but which will not on its own elicit an immune response.
  • adjuvants may augment the immune response using a variety of biological mechanisms, including but not limited to lymphocytic recruitment, T cell stimulation, B cell stimulation, and macrophage stimulation.
  • the terms “treating”, and “treatment” and the like are used herein to generally mean obtaining a desired pharmacological and physiological effect.
  • the effect may be prophylactic in terms of preventing or partially preventing a disease, symptom or condition thereof and/or may be therapeutic in terms of a partial or complete cure of a disease, condition, symptom or adverse effect attributed to the disease.
  • treatment covers any treatment of a disease in a mammal, particularly a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; or (c) relieving the disease, i.e., causing regression of the disease and/or its symptoms or conditions.
  • the invention is directed towards treating a patient's suffering from disease related to pathological inflammation.
  • the present invention is involved in preventing, inhibiting, or relieving adverse effects attributed to pathological inflammation over long periods of time and/or are such caused by the physiological responses to inappropriate inflammation present in a biological system over long periods of time.
  • pathological inflammation refers to an inappropriate and chronic inflammation associated with disorders including, but not limited to, asthma, atherosclerosis, AIDS dementia, diabetes, inflammatory bowel disease, rheumatoid arthritis, transplant rejection, graft versus host disease, multiple sclerosis (especially to inhibit further demyelination), tumor metastasis, nephritis, atopic dermatitis, psoriasis, myocardial ischemia, chronic prostatitis, complications from sickle cell anemia, lupus erythematosus, and acute leukocyte mediated lung injury.
  • Such inflammation is characterized by a heightened response of inflammatory cells, including infiltrating leukocytes. Over time, such pathological inflammation often results in damage to tissue in the region of inappropriate inflammation.
  • Antegren® is meant to include the antibody also known as AN100226 (antibody code number) or natalizumab (USAN name).
  • Antegreng is a recombinant, humanized anti-alpha-4 integrin antibody.
  • the disease or condition being treated in the mammal is one which is modulated when a therapeutically effective dose of Antegren® is administered.
  • the present invention is based on the surprising result that chronic administration of an emerging class of new compounds known as selective adhesion molecule inhibitors (SAMIs) is sufficient to provide the maintenance of chronic suppression of inflammation in disorders involving integrin dimers.
  • SAMIs selective adhesion molecule inhibitors
  • Previous treatments of inflammatory inhibitors have approached the dosage regimes quite differently, in the belief that the administration of an inflammatory inhibitor would cause a reaction of the body's own response system, which would in turn lead to a recognition of the inflammation as pathological and a resulting chronic relief of the pathological inflammation.
  • a chronic dosage regime is not only more effective than a short-term dosage regime, but in fact it is required to maintain the suppression of pathological inflammation.
  • the levels of an anti-alpha-4 integrin agent need to be maintained over a number of months or even years.
  • the present invention is based on the results of a large, randomized, placebo-controlled trial of an anti-alpha-4 integrin antibody, natalizumab, in patients with relapsing MS or with moderate to severely active CD.
  • Natalizumab is a recombinant, humanized, monoclonal antibody antagonist against alpha-4 integrin. Results from these two trials have shown that treatment with natalizumab improved the signs and symptoms of patients with MS and CD.
  • the invention is also intended to include other chimeric antibodies, including primatizedTM antibodies.
  • the method of the invention does not involve any particular mode of administration, since the mode of administration is dependent upon the form of the active agent and the formulation developed to administer the active agent(s).
  • the specific examples described here were obtained using parenteral administration of natalizumab.
  • the present invention is described using an antibody that specifically binds to alpha-4 integrin, it is also intended to include chronic administration of, for example, bivalent or multivalent antibodies that recognize both partners of an integrin dimer, provided the dimer comprises an alpha-4 integrin.
  • the general concept of the invention relates to introducing relatively constant amounts of an active agent to a patient's circulatory system over a period of months or years.
  • This chronic introduction of an agent that selectively binds to alpha-4 integrin or a dimer comprising alpha-4 integrin results in suppression of pathological inflammation being maintained at a constant level over a period of time.
  • pathological inflammation can be chronically suppressed in the patient.
  • the invention involves obtaining and maintaining a receptor saturation level in a human patient of a dimer comprising alpha-4 integrin in a range of from about 65% to about 100%, more preferably between about 75% to about 100%, and even more preferably between about 80% to about 100%. These receptor saturation levels are maintained at these levels chronically (e.g., over a period of 6 months or so) to allow for continued suppression of pathological inflammation.
  • Agents that Selectively Bind to Alpha-4 Integrins are maintained at these levels chronically (e.g., over a period of 6 months or so) to allow for continued suppression of pathological inflammation.
  • agents with the ability to bind to and inhibit alpha-4 integrin activity can be used in the practice of the invention. Many such agents have been identified and characterized, and specific agents are described below. Given the teachings disclosed herein, it is well within the skill of one in the art to identify other agents that will be able to inhibit the alpha-4-comprising integrin dimers in a manner that biologically mimics or is similar to the specifically described agents, and the present invention is intended to include the chronic administration of such agents and combinations of such agents.
  • the agents of the invention are antibodies or immunologically active fragments thereof that selectively bind to an alpha-4 integrin or a dimer comprising alpha-4, such as alpha-4 beta-1 or alpha-4 beta-7.
  • the agent of the invention is an antibody
  • a monoclonal antibody is preferred.
  • each monoclonal antibody is directed against a single epitope on the antigen.
  • a second advantage of monoclonal antibodies is that they are synthesized by means that are uncontaminated by other immunoglobulins, e.g., by phage display or isolation from a hybridoma.
  • monoclonal antibodies are preferred as they are highly specific, and the invention is thus discussed primarily in terms of monoclonal antibodies.
  • antibodies of the present invention can be produced using phage display technology. Antibody fragments that selectively bind to an alpha-4 integrin or a dimer comprising an alpha-4 integrin are then isolated. Exemplary preferred methods for producing such antibodies via phage display are disclosed in U.S. Pat. Nos. 6,225,447; 6,180,336; 6,172,197; 6,140,471; 5,969,108; 5,885,793; 5,872,215; 5,871,907; 5,858,657; 5,837,242; 5,733,743; and 5,565,332.
  • Monoclonal antibodies can also be produced using the conventional hybridoma methods. These methods have been widely applied to produce hybrid cell lines that secrete high levels of monoclonal antibodies against many specific antigens, and can also be used to produce monoclonal antibodies of the present invention.
  • mice e.g., Balb/c mice
  • mice can be immunized with an antigenic alpha-4 integrin epitope by intraperitoneal injection. After sufficient time has passed to allow for an immune response, the mice are sacrificed, and the spleen cells obtained and fused with myeloma cells, using techniques well known in the art.
  • hybridomas are then grown in a selective medium, and the surviving cells grown in such medium using limiting dilution conditions.
  • hybridomas can be isolated that secrete antibodies (for example, of the IgG or IgM class or IgG1 subclass) that selectively bind to the target, alpha-4 integrin or a dimer comprising an alpha-4 integrin.
  • the isolated monoclonal can then be used to produce chimeric and humanized antibodies.
  • Antibodies of the invention include, but are not limited to, polyclonal, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies (e.g., scFv), Fab fragments, F(ab′) fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), and epitope-binding fragments of any of the above.
  • the antibodies are human antigen-binding antibody fragments of the present invention and include, but are not limited to, Fab, Fab′ and F(ab′) 2 , Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain.
  • Antigen-binding antibody fragments, including single-chain antibodies may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: hinge region, CH1, CH2, and CH3 domains. Also included in the invention are antigen-binding fragments that can comprise any combination of variable region(s) with a hinge region, CH1, CH2, and CH3 domains.
  • the antibodies of the invention may be from any animal origin including birds and mammals.
  • the antibodies are human, murine (e.g., mouse and rat), donkey, sheep, monkey, rabbit, goat, guinea pig, pig, camel, horse, or chicken (or other avian).
  • “human” antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulins and that do not express endogenous immunoglobulins, as described infra and, for example in, U.S. Pat. No. 5,939,598 by Kucherlapati et al.
  • Chimeric and humanized antibodies can be produced from non-human antibodies, and can have the same or similar binding affinity as the antibody from which they are produced.
  • Techniques for producing chimeric antibodies include splicing the genes from, e.g., a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity; such antibodies are within the scope of this invention.
  • a nucleic acid encoding a variable (V) region of a mouse monoclonal antibody can be joined to a nucleic acid encoding a human constant (C) region, e.g., IgG1 or IgG4.
  • C human constant
  • the resulting antibody is thus a species hybrid, generally with the antigen binding domain from the non-human antibody and the C or effector domain from a human or primate antibody.
  • Humanized antibodies are antibodies with variable regions that are primarily from a human antibody (i.e., the acceptor antibody), but which have complementarity determining regions substantially from a non-human antibody (the donor antibody). See, e.g., Queen et al., Proc. Nat'l. Acad. Sci USA 86: 10029-10033 (1989); WO 90/07861, U.S. Pat. Nos. 6,054,297; 5,693,761; 5,585,089; 5,530,101; and 5,224,539.
  • the constant region or regions of these antibodies are generally also from a human antibody.
  • the human variable domains are typically chosen from human antibodies having sequences displaying a high homology with the desired non-human variable region binding domains.
  • the heavy and light chain variable residues can be derived from the same antibody, or a different human antibody.
  • the sequences can be chosen as a consensus of several human antibodies, such as described in WO 92/22653.
  • a “primatizedTM antibody” is a recombinant antibody containing primate variable sequences or antigen binding portions, and human constant domain sequences. See Newman, Biotechnology, 1992, 10: 1455-60. Primatization of antibodies results in the generation of antibodies that contain monkey variable domains and human constant sequences. For more details see U.S. Pat. No. 6,113,898. This technique modifies antibodies such that they are not rejected upon administration in humans because they are antigenic. This technique relies on immunization of cynomolgus monkeys with human antigens or receptors. This technique was developed to create high affinity monoclonal antibodies directed to human cell surface antigens.
  • Specific amino acids within the human variable region are selected for substitution based on the predicted conformation and antigen binding properties. This can be determined using techniques such as computer modeling, prediction of the behavior and binding properties of amino acids at certain locations within the variable region, and observation of effects of substitution. For example, when an amino acid differs between a non-human variable region and a human variable region, the human variable region can be altered to reflect the amino acid composition of the non-human variable region.
  • the antibodies used in the chronic dosage regime of the present invention are humanized antibodies as disclosed in U.S. Pat. No. 5,840,299, which is incorporated herein by reference.
  • transgenic mice containing human antibody genes can be immunized with an antigenic alpha-4 integrin structure and hybridoma technology can be used to generate human antibodies that selectively bind to alpha-4 integrin.
  • Chimeric, human and/or humanized antibodies can be produced by using recombinant expression, e.g., expression in human hybridomas (Cole et al., Monoclonal Antibodies and Cancer Therapy , Alan R. Liss, p. 77 (1985)), in myeloma cells or in Chinese hamster ovary (CHO) cells.
  • antibody coding sequences can be incorporated into transgenes for introduction into the genome of a transgenic animal and subsequent expression in the milk of the transgenic animal. See e.g., U.S. Pat. Nos. 6,197,946; 5,849,992; 5,565,362; 5,336,894; and 5,304,489.
  • Suitable transgenes include transgenes having a promoter and/or enhancer from a mammary gland specific gene, for example casein or ⁇ -lactoglobulin.
  • Small molecules for use in the present invention encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 50 and less than about 4,000 Daltons.
  • Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of functional chemical groups.
  • the candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Candidate agents are also found among biomolecules including, but not limited to: peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof.
  • the small molecules can be obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides and oligopeptides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs.
  • the methods of the invention can be performed with any peptide that is capable of binding to an alpha-4 integrin or a dimer comprising an alpha-4 subunit. Included in the methods of the invention are peptides that are substantially homologous to a region of the extracellular matrix or a natural ligand of the specific alpha-4 integrin receptor or receptors targeted.
  • peptides can be used that comprise at least a portion of the fibronectin IIICS region (e.g., peptides comprising at least a portion of the CS-I peptide sequence or a sequence substantially homologous to the CS-1 sequence) can be used to bind to a receptor and inhibit the activity of the alpha-4 comprising integrin. See for example U.S. Ser. No. 08/452,098, which is incorporated by reference in its entirety.
  • the agents of the invention are peptides or peptidomimetics that comprise an immunogenic fragment of an alpha-4 integrin.
  • An immunogenic fragment is any fragment that comprises an epitope of alpha-4, and generally has at least 3, 5, 7, 10, 15, 17 or 20 contiguous amino acids from a naturally occurring mammalian alpha-4 protein.
  • GenBank accesion Nos. AA59613 and NP — 034706).
  • One skilled in the art can readily design a peptide agent of the invention based on the amino acid sequence of alpha-4 or using the wild-type nucleotides that encode them (e.g., GenBank Accession Nos.
  • an appropriate peptide can be initially screened against antibodies known to have the desired immunogenic activity, e.g., antibodies that selectively bind to an alpha-4 structure and are characterized by the ability to inhibit the activity of an integrin comprising alpha-4.
  • the immunogenic fragment may also be designed to have amino acid analogs or other structural elements that will enhance the immunogenic response.
  • peptide fragments may have altered C- or N-terminal ends that enhance overall immunogenicity of the molecules while not impeding their ability to elicit an immune response.
  • analogs include, but are not limited to: alpha, alpha-disubstituted amino acids, N-alkyl amino acids, lactic acid, 4-hydroxyproline, gamma-carboxyglutamate, gamma-N,N,N,-trimethyllysine, gamma-N-acetyllysine, O-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, and 5-hydroxylysine.
  • Other useful analogs can be found in Sigma, Biochemicals and Reagents , Sigma-Aldrich (2001).
  • the fragment may also be detectably labeled to allow for tracking of the molecule within a subject following administration to a subject.
  • Peptides, analog structures, peptidomimetics and the like can be isolated from natural sources, and then optionally processed (e.g., via peptide cleavage) or alternatively synthesized by conventional techniques known in the art such as solid phase synthesis or recombinant expression. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Press, New York, 2nd edition 1989). Automatic peptide synthesis can be performed using a commercially available apparatus from manufacturers such as Applied Biosystems (Foster City, Calif.), and methods of doing so are well established.
  • Recombinant production of the proteins may be in prokaryotic, such as phage or bacterial cells or eukaryotic systems, such as yeast, insect, or mammalian cells.
  • proteins can be produced using cell-free in vitro systems known in the art.
  • phage peptide display libraries can be used to express large numbers of peptides that can be screened in vitro to identify peptides that specifically bind to alpha-4 or a dimer comprising an alpha-4 integrin.
  • Phage display technology provides a means for expressing a diverse population of random or selectively randomized peptides.
  • Various methods of phage display and methods for producing diverse populations of peptides are well known in the art. For example, Ladner et al. (U.S. Pat. No. 5,223,409), describes methods for preparing diverse populations of binding domains on the surface of a phage. Ladner et al.
  • phage vectors useful for producing a phage display library as well as methods for selecting potential binding domains and producing randomly or selectively mutated binding domains.
  • Screening of a phage display library generally involves in vitro panning of the library using a purified target molecule. Phage that bind the target molecule can be recovered; individual phage can be cloned, and the peptide expressed by a cloned phage can be determined.
  • the molecules of a peptide phage display library also can be present as a conjugate, which can facilitate recovery or identification of the peptide of interest.
  • conjugate means a peptide or peptidomimetic of the library linked to a physical, chemical or biological moiety such as but not limited to a solid substrate, a plastic microbead, an oligonucleotide or a bacteriophage, and the like. The moiety can provide a means to identify or recover an agent.
  • a peptide agent can be linked to a suitable carrier to facilitate an immune response.
  • suitable carriers include serum albumins, keyhole limpet hemocyanin (KLH), immunoglobulin molecules, thyroglobulin, ovalbumin, tetanus toxoid, or a toxoid from other pathogenic bacteria, such as diphtheria, E. coli , cholera, or H. pylori , or an attenuated toxin derivative.
  • T-cell epitopes that bind to multiple MHC alleles, e.g., at least 75% of all human MHC alleles. Such carriers are sometimes known in the art as “universal T-cell epitopes.”
  • Examples of universal T-cell epitopes include: Influenza Hemagluttinin: HA 307-319 PKYVKQNTLKLAT (SEQ ID NO: 2) PADRE AKXVAAWTLKAAA, (SEQ ID NO: 3) where X is preferably cyclohexylalanine, tyrosine, or phenylalanine Malaria CS: T3 epitope EKKIAKMEKASSVFNV (SEQ ID NO: 4) Hepatitis B surface antigen: HBsAg 19-28 FFLLTRILTI (SEQ ID NO: 5) Heat Shock Protein 65: hsp65 153-171 DQSIGDLIAEAMDKVGNEG (SEQ ID NO: 6) bacille Calmette-
  • cytokines such as IL-1, IL-1 ⁇ and ⁇ peptides, IL-2, ⁇ -INF, IL-10, GM-CSF, and chemokines, such as macrophage inflammatory protein (MIP)1 ⁇ and ⁇ and RANTES (i.e., regulation upon activation normal T cell expressed and secreted).
  • MIP macrophage inflammatory protein
  • RANTES RANTES
  • Immunogenic agents can be linked to carriers by chemical cross-linking.
  • Techniques for linking an agent to a carrier include but are not limited to the formation of disulfide linkages using N-succinimidyl-3-(2-pyridyl-thio) propionate (SPDP) and succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC) (if the peptide lacks a sulfhydryl group, this can be provided by addition of a cysteine residue).
  • SPDP N-succinimidyl-3-(2-pyridyl-thio) propionate
  • SMCC succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate
  • reagents create a disulfide linkage between themselves and a peptide cysteine that resides on one protein and an amide linkage through the ⁇ -amino on a lysine, or other free amino group in other amino acids.
  • disulfide/amide-forming agents are described in Immun. Rev. 62: 185 (1982).
  • Other bifunctional coupling agents form a thioether rather than a disulfide linkage.
  • Many of these thio-ether-forming agents are commercially available and include reactive esters of 6-maleimidocaproic acid, 2-bromoacetic acid, 2-iodoacetic acid, and 4-(N-maleimido-methyl)cyclohexane-1-carboxylic acid.
  • the carboxyl groups can be activated by combining them with succinimide or 1-hydroxyl-2-nitro-4-sulfonic acid sodium salt.
  • Peptide agents can also be expressed as fusion proteins with carriers (i.e., heterologous peptides).
  • the peptide agent can be linked at its amino terminus, its carboxyl terminus, or both to a carrier.
  • multiple repeats of the immunogenic peptide can be present in the fusion protein.
  • an immunogenic peptide can be linked to multiple copies of a heterologous peptide, for example, at both the N- and C-termini of the peptide.
  • Some carrier peptides serve to induce a helper T-cell response against the carrier peptide.
  • the induced helper T-cells in turn induce a B-cell response against the immunogenic peptide linked to the carrier peptide.
  • the agent is an antibody, polypeptide, peptide, small molecule or other pharmaceutically useful compound according to the present invention that is to be given to an individual
  • administration is via a chronic dosage regime.
  • the actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g., decisions on dosage etc., is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of the techniques and protocols mentioned above can be found in Remington's Pharmaceutical Sciences, 18th edition, Osol, A. (ed.), 1990.
  • the present invention also provides pharmaceutical compositions for the reduction of chronic pathological inflammation in a subject susceptible to such and/or suffering from a disorder associated with pathological inflammation.
  • compositions of the invention preferably contain an agent in a concentration from about 0.1 to about 10% of the formulation. They may also be used in appropriate association with other pharmaceutically active compounds.
  • the following methods and excipients are merely exemplary and are in no way meant to be limiting.
  • the agents can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • the formulation is preferably administered in a parenteral dosage form.
  • the preferred form depends on the intended mode of administration and therapeutic application.
  • the compositions can also include, depending on the formulation desired, pharmaceutically acceptable, non-toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration.
  • the diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution.
  • the pharmaceutical composition or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, non-immunogenic stabilizers and the like.
  • carrier molecules such as proteoglycans.
  • Specific examples of such carrier molecules include, but are not limited to, glycosaminoglycans such as heparin sulfate, hyaluronic acid, keratan-sulfate, chondroitin 4-sulfate, chondroitin 6-sulfate, heparan sulfate and dermatin sulfate, perlecan, and pentopolysulfate.
  • Antibodies of the invention can be administered as injectionable dosages of a solution or as a suspension of the substance in a physiologically acceptable diluent with a pharmaceutical carrier that can be a sterile liquid such as water and oils with or without the addition of a surfactant.
  • a pharmaceutical carrier that can be a sterile liquid such as water and oils with or without the addition of a surfactant.
  • Other pharmaceutically preparations are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, and mineral oil.
  • glycols such as propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions.
  • the agents of this invention can be administered in a sustained release form, for example a depot injection, implant preparation, or osmotic pump, which can be formulated in such a manner as to permit a sustained release of the active ingredient.
  • agents of the invention that are antibodies may be provided by administering a polynucleotide encoding a whole or partial antibody (e.g., a single chain Fv) to a subject.
  • the polynucleotide is administered to a subject in an appropriate vehicle to allow the expression of the antibody in the subject in a therapeutically effective amount.
  • the agents of the invention can be formulated into preparations for injections by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol.
  • aqueous or nonaqueous solvent such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol.
  • the formulations may also contain conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • the agents can be utilized in aerosol formulation to be administered via inhalation or pulmonary delivery.
  • the agents of the present invention can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like.
  • the agents can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • the agents of the present invention can be administered rectally via a suppository.
  • the suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature.
  • an agent of the invention may be accomplished by any convenient means, including parenteral injection, and may be systemic or localized in delivery.
  • the agents of this invention can be incorporated into a variety of formulations for therapeutic administration. More particularly, the agents of the present invention can be formulated into pharmaceutical compositions by combination with appropriate pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • administration of the agents can be achieved in various ways, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intratracheal, intranasal, gastric, intramuscular, intracranial, subdermal etc., administration.
  • the active agent may be systemic after administration or may be localized by the use of regional administration, intramural administration, or use of an implant that acts to retain the active dose at the site of implantation.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more agents of the present invention.
  • unit dosage forms for injection or intravenous administration may comprise the agent of the present invention in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • Implants for sustained release formulations are well known in the art. Implants are formulated as microspheres, slabs, etc. with biodegradable or non-biodegradable polymers. For example, polymers of lactic acid and/or glycolic acid form an erodible polymer that is well tolerated by the host.
  • the implant is placed in proximity to the site of protein deposits (e.g., the site of formation of amyloid deposits associated with neurodegenerative disorders), so that the local concentration of active agent is increased at that site relative to the rest of the body.
  • a typical dosage unit for administration of a subject includes, but is not limited to: a solution suitable for intravenous administration; a tablet taken from two to six times daily; or a one time-release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient, etc.
  • the time-release effect may be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure or by any other known means of controlled release.
  • Certain agents of the invention are sometimes administered in combination with an adjuvant.
  • adjuvants can be used in combination with an anti alpha-4 integrin agent to elicit an immune response.
  • Preferred adjuvants augment the intrinsic response to an agent without causing conformational changes in the agent that affect the qualitative form of the response.
  • Preferred adjuvants include aluminum hydroxide and aluminum phosphate, 3 De-O-acylated monophosphoryl lipid A (MPLTM) (see GB 2220211 (RIBI ImmunoChem Research Inc., Hamilton, Mont., now part of Corixa).
  • StimulonTM QS-21 is a triterpene glycoside or saponin isolated from the bark of the Quillaja Saponaria Molina tree found in South America (see Kensil et al., in Vaccine Design: The Subunit and Adjuvant Approach (eds. Powell & Newman, Plenum Press, NY, 1995); U.S. Pat. No. 5,057,540, (Aquila BioPharmaceuticals, Framingham, Mass.).
  • Other adjuvants are oil in water emulsions (such as squalene or peanut oil), optionally in combination with immune stimulants, such as monophosphoryl lipid A (see Stoute et al., 1997 N. Engl. J. Med. 336: 86-91).
  • Adjuvants can be administered as a component of a therapeutic composition with an active agent or can be administered separately, before, concurrently with, or after administration of the therapeutic agent.
  • a preferred class of adjuvants for administration is aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, and aluminum sulfate. Such adjuvants can be used with or without other specific immunostimulating agents such as MPL or 3-DMP, QS-21, polymeric or monomeric amino acids such as polyglutamic acid or polylysine.
  • Another class of adjuvants is oil-in-water emulsion formulations.
  • Such adjuvants can be used with or without other specific immunostimulating agents such as muramyl peptides (e.g., N-acetylmuramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1′-2′dipalmitoyl-sn-glycero-3-hydroxyphosphoryloxy)-ethylamine (MTP-PE), N-acetylglucsaminyl-N-acetylmuramyl-L-Al-D-isoglu-L-Ala-dipalmitoxy propylamide (DTP-DPP) theramideTM, or other bacterial cell wall components.
  • muramyl peptides e
  • Oil-in-water emulsions include (a) MF59 (WO 90/14837), containing 5% Squalene, 0.5% Tween 80, and 0.5% Span 85 (optionally containing various amounts of MTP-PE) formulated into submicron particles using a microfluidizer such as Model 110Y microfluidizer (Microfluidics, Newton Mass.), (b) SAF, containing 10% Squalene, 0.4% Tween 80, 5% pluronic-blocked polymer L121, and thr-MDP, either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) RibiTM adjuvant system (RAS), (Ribi Immunochem, Hamilton, Mont.) containing 2% squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphoryl lipid A, trehalose dimycolate (TDM), and cell wall
  • adjuvants are saponin adjuvants, such as StimulonTM (QS-21; Aquila, Framingham, Mass.) or particles generated therefrom, such as ISCOMs (immunostimulating complexes) and ISCOMATRIX.
  • Other adjuvants include Complete and Incomplete Freund's Adjuvant (IFA), cytokines, such as interleukins (IL-1, IL-2, and IL-12), macrophage colony stimulating factor (M-CSF), and tumor necrosis factor (TNF).
  • IFA Complete and Incomplete Freund's Adjuvant
  • cytokines such as interleukins (IL-1, IL-2, and IL-12
  • M-CSF macrophage colony stimulating factor
  • TNF tumor necrosis factor
  • An adjuvant can be administered with an agent as a single composition, or can be administered before, concurrent with or after administration of the agent.
  • the agent and an adjuvant can be packaged and supplied in the same vial or can be packaged in separate vials and mixed before use.
  • the agent and adjuvant are typically packaged with a label indicating the intended therapeutic application. If the agent and adjuvant are packaged separately, the packaging typically includes instructions for mixing before use.
  • the choice of an adjuvant and/or carrier depends on such factors as the stability of the formulation containing the adjuvant, the route of administration, the dosing schedule, and the efficacy of the adjuvant for the species being vaccinated. In humans, a preferred pharmaceutically acceptable adjuvant is one that has been approved for human administration by pertinent regulatory bodies.
  • Examples of such preferred adjuvants for humans include alum, MPL and QS-21.
  • two or more different adjuvants can be used simultaneously.
  • Preferred combinations include alum with MPL, alum with QS-21, MPL with QS-21, and alum, QS-21 and MPL together.
  • Incomplete Freund's adjuvant can be used (Chang et al., 1998 Advanced Drug Delivery Reviews 32: 173-186), optionally in combination with any of alum, QS-21, and MPL and all combinations thereof.
  • the chronic treatment regimes of the present invention provides anti-alpha-4 integrin agent at a level that will maintain sufficient receptor saturation to suppress pathological inflammation in a patient in need of such.
  • the methods of the invention entail administration once per every two weeks or once a month to once every two months, with repeated dosings taking place over a period of at least six months, and more preferably for a year or longer.
  • the methods of the invention involve obtaining and maintaining a receptor saturation level in a human patient of a dimer comprising alpha-4 integrin (e.g., VLA-4) in a range of from about 65% to 100%, more preferably between about 75% to about 100%, and even more preferably between about 80% to about 100%.
  • alpha-4 integrin e.g., VLA-4
  • These receptor saturation levels are maintained at these levels chronically (e.g., over a period of 6 months or so) to allow for continued suppression of pathological inflammation.
  • the anti-alpha-4 agent is an antibody, preferably a humanized or human antibody (e.g., natalizumab), and the dosing is on a monthly basis.
  • Levels of receptor saturation can be monitored to determine the efficacy of the dosing regime, and physiological markers measured to confirm the success of the dosage regime.
  • serum levels of the antibody can be monitored to identify clearance of the antibody and to determine the potential effect of half-life on the efficacy of the treatment.
  • the amount of agent administered in a dosage unit may depend on whether adjuvant is also administered, with higher dosages generally being required in the presence of adjuvant.
  • the dosage ranges from about 0.0001 to about 100 mg/kg, and more usually about 0.01 to 5 mg/kg, of the host body weight.
  • dosages can be about 1 mg/kg body weight or about 10 mg/kg body weight.
  • Dosage and frequency may vary depending on the half-life of the agent in the patient.
  • the dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic.
  • each dosing injection is generally between about 2.0 to about 8.0 mg/kg.
  • effective dosages can be monitored by obtaining a fluid sample from the patient, generally a blood serum or cerebrospinal fluid sample, and determining the integrin receptor saturation using methods well known in the art.
  • a sample is taken prior to initial dosing; subsequent samples are taken and measured prior to and/or after each immunization.
  • a particularly preferred amount is a 3 mg per kg of patient per month of natalizumab or an immunologically active fragment equivalent thereof.
  • the dosage level is increased in accordance with the particular adjuvant and the level of immunogenicity of the anti-alpha-4 agent.
  • Doses for individual agents, selected in accordance with the present invention are determined according to standard dosing methods, taken in conjunction with the teachings provided herein.
  • an anti-alpha-4 agent can be administered as a sustained release formulation, provided the dosage is such that the levels of receptor saturation remain sufficient to suppress inflammation.
  • controlled release systems can be used to chronically administer an anti-alpha-4 agent within the scope of this invention. Discussions of appropriate controlled release dosage forms may be found in Lesczek Krowczynski, Extended - Release Dosage Forms, 1987 (CRC Press, Inc.).
  • Controlled release technologies include, but are not limited to physical systems and chemical systems.
  • Physical systems include, but are not limited to, reservoir systems with rate-controlling membranes, such as microencapsulation, macroencapsulation, and membrane systems; reservoir systems without rate-controlling membranes, such as hollow fibers, ultra microporous cellulose triacetate, and porous polymeric substrates and foams; monolithic systems, including those systems physically dissolved in non-porous, polymeric, or elastomeric matrices (e.g., non-erodible, erodible, environmental agent ingression, and degradable), and materials physically dispersed in non-porous, polymeric, or elastomeric matrices (e.g., non-erodible, erodible, environmental agent ingression, and degradable); laminated structures, including reservoir layers chemically similar or dissimilar to outer control layers; and other physical methods, such as osmotic pumps, or
  • Chemical systems include, but are not limited to, chemical erosion of polymer matrices (e.g., heterogeneous, or homogeneous erosion), or biological erosion of a polymer matrix (e.g., heterogeneous, or homogeneous). Additional discussion of categories of systems for controlled release may be found in Agis F. Kydonieus, Controlled Release Technologies: Methods, Theory and Applications, 1980 (CRC Press, Inc.).
  • the methods of the invention can be used to treat a patient that is affected with a disorder involving or arising from pathological inflammation, or to prophylactically treat a patient at risk for a particular disorder.
  • the dosage regimes that are necessary for prophylactic versus therapeutic treatment can vary, and will need to be designed for the specific use and disorder treated.
  • two or more agents are administered simultaneously, in which case the dosage of each agent administered falls within the ranges indicated. Intervals can also be irregular as indicated by measuring receptor saturation levels or by following other indicia of the disease process.
  • dose levels can vary as a function of the specific agent, the severity of the symptoms and the susceptibility of the subject to side effects. Some of the specific agents are more potent than others. Preferred dosages for a given agent are readily determinable by those of skill in the art by a variety of means. A preferred means is to measure the physiological potency of a given agent.
  • the controlled release formulations of the present invention can be used to obtain a wide range of desirable effects. Particularly the formulations of the invention are useful in treating essentially any disease state or symptom that is treatable by long term administration of anti-inflammatories that target pathological inflammation.
  • the invention also provides methods of treatment that exploit the ability of anti-alpha-4 integrin agents to block alpha-4-dependent interactions.
  • the alpha-4-dependent interaction with the VCAM-1 ligand on endothelial cells is an early event in many inflammatory responses, including those of the central nervous system.
  • Undesired diseases and conditions resulting from inflammation and having acute and/or chronic clinical exacerbations include multiple sclerosis (Yednock et al., 1992 Nature 356: 63; Baron et al., 1993 J. Exp. Med.
  • nephritis Rabb et al., 1995 Springer Semin. Immunopathol. 16: 417-25
  • retinitis atopic dermatitis
  • psoriasis myocardial ischemia
  • chronic prostatitis complications from sickle cell anemia, lupus erythematosus
  • acute leukocyte-mediated lung injury such as occurs in adult respiratory distress syndrome.
  • CD Crohn's disease
  • Ulcerative colitis is an inflammatory response limited largely to the colonic mucosa and submucosa. Lymphocytes and macrophages are numerous in lesions of inflammatory bowel disease and may contribute to inflammatory injury.
  • Asthma is a disease characterized by increased responsiveness of the tracheobronchial tree to various stimuli potentiating paroxysmal constriction of the bronchial airways.
  • the stimuli cause release of various mediators of inflammation from IgE-coated mast cells including histamine, eosinophilic and neutrophilic chemotactic factors, leukotrines, prostaglandin and platelet activating factor. Release of these factors recruits basophils, eosinophils and neutrophils, which cause inflammatory injury.
  • Atherosclerosis is a disease of arteries (e.g., coronary, carotid, aorta and iliac).
  • the basic lesion, the atheroma consists of a raised focal plaque within the intima, having a core of lipid and a covering fibrous cap.
  • Atheromas compromise arterial blood flow and weaken affected arteries.
  • Myocardial and cerebral infarcts are a major consequence of this disease. Macrophages and leukocytes are recruited to atheromas and contribute to inflammatory injury.
  • Rheumatoid arthritis is a chronic, relapsing inflammatory disease that primarily causes impairment and destruction of joints. Rheumatoid arthritis usually first affects the small joints of the hands and feet but then may involve the wrists, elbows, ankles and knees. The arthritis results from interaction of synovial cells with leukocytes that infiltrate from the circulation into the synovial lining of joints. See e.g., Paul, Immunology (3d ed., Raven Press, 1993).
  • CD8 + cells, CD4 + cells and monocytes are all involved in the rejection of transplant tissues.
  • Antibodies directed to alpha-4 integrin are useful, inter alia, to block alloantigen-induced immune responses in the donee thereby preventing such cells from participating in the destruction of the transplanted tissue or organ. See, e.g., Paul et al., 1996 Transplant International 9: 420-425; Georczynski et al., 1996 Immunol. 87: 573-580); Georcyznski et al., 1995 Transplant. Immunol. 3: 55-61); Yang et al., 1995 Transplantation 60: 71-76); Anderson et al., 1994 APMIS 102: 23-27.
  • GVHD graft versus host disease
  • GVHD is a potentially fatal disease that occurs when immunologically competent cells are transferred to an allogeneic recipient. In this situation, the donor's immunocompetent cells may attack tissues in the recipient. Tissues of the skin, gut epithelia and liver are frequent targets and may be destroyed during the course of GVHD.
  • the disease presents an especially severe problem when immune tissue is being transplanted, such as in bone marrow transplantation; but less severe GVHD has also been reported in other cases as well, including heart and liver transplants.
  • the therapeutic agents of the present invention are used, inter alia, to block activation of the donor T-cells thereby interfering with their ability to lyse target cells in the host.
  • a further use of anti alpha-4 agents of the invention is inhibiting tumor metastasis.
  • Several tumor cells have been reported to express alpha-4 integrin and antibodies to alpha-4 integrin have been reported to block adhesion of such cells to endothelial cells (Steinback et al., 1995 Urol. Res. 23: 175-83); Orosz et al., 1995 Int. J. Cancer 60: 867-71); Freedman et al., 1994 Leuk. Lymphoma 13: 47-52); Okahara et al., 1994 Cancer Res. 54: 3233-6).
  • MS Multiple sclerosis
  • Multiple sclerosis is a progressive neurological autoimmune disease that affects an estimated 250,000 to 350,000 people in the United States. Multiple sclerosis is thought to be the result of a specific autoimmune reaction in which certain leukocytes attack and initiate the destruction of myelin, the insulating sheath covering nerve fibers.
  • murine monoclonal antibodies directed against alpha-4 beta-1 integrin have been shown to block the adhesion of leukocytes to the endothelium, and thus prevent inflammation of the central nervous system and subsequent paralysis in the animals.
  • MS The onset of MS may be dramatic or so mild as to not cause a patient to seek medical attention.
  • the most common symptoms include weakness (in one or more limbs, visual blurring due to optic neuritis, sensory disturbances, diplopia and ataxia.
  • the course of disease may be stratified into three general categories: (1) relapsing MS, (2) chronic progressive MS, and (3) inactive MS.
  • Relapsing MS is characterized by recurrent attacks of neurologic dysfunction. MS attacks generally evolve over days to weeks and may be followed by complete, partial or no recovery. Recovery from attacks generally occurs within weeks to several months from the peak of symptoms, although rarely some recovery may continue for 2 or more years.
  • Chronic progressive MS results in gradually progressive worsening without periods of stabilization or remission. This form develops in patients with a prior history of relapsing MS, although in 20% of patients, no relapses can be recalled. Acute relapses also may occur during the progressive course.
  • inactive MS is characterized by fixed neurologic deficits of variable magnitude. Most patients with inactive MS have an earlier history of relapsing MS.
  • the course of MS is also dependent on the age of the patient.
  • favourable prognostic factors include early onset (excluding childhood), a relapsing course and little residual disability 5 years after onset.
  • poor prognosis is associated with a late age of onset (i.e., age 40 or older) and a progressive course.
  • These variables are interdependent, since chronic progressive MS tends to begin at a later age that relapsing MS.
  • Disability from chronic progressive MS is usually due to progressive paraplegia or quadriplegia in individual patients.
  • patients will preferably be treated when the patient is in remission rather then in a relapsing stage of the disease.
  • adrenocorticotropic hormone e.g., oral prednisone or intravenous methylprednisolone
  • oral corticosteroids e.g., oral prednisone or intravenous methylprednisolone
  • Newer therapies for MS include treating the patient with interferon beta-1b, interferon beta-1a, and copaxone® (formerly known as copolymer 1). These three drugs have been shown to significantly reduce the relapse rate of the disease. These drugs are typically self-administered intramuscularly or subcutaneously.
  • MS demyelination or MS.
  • One aspect of the invention contemplates treating MS with agents disclosed herein either alone or in combination with other standard treatment modalities.
  • Standard treatment modalities include but are not limited to the following. Additional treatment modalities not discussed herein for use in treating MS in combination with the methods and compositions disclosed herein depending on the state of disease in the patient would be evident to the skilled practitioner. Such additional treatment modalities for MS other pathological inflammation would include other immunomodulators or immunosupressants.
  • compositions discussed supra can be chronically administered for prophylactic and/or therapeutic treatments of the previously listed inflammatory disorders, including multiple sclerosis, inflammatory bowel disease, asthma, atherosclerosis, rheumatoid arthritis, organ or graft rejection and graft versus host disease.
  • compositions are administered to a patient suspected of, or already suffering from such a disease in an amount sufficient to cure, or at least partially arrest, the symptoms of the disease and its complications. An amount adequate to accomplish this is defined as a therapeutically- or pharmaceutically-effective dose.
  • compositions are chronically administered to a patient susceptible to, or otherwise at risk of, a particular disease in an amount sufficient to eliminate or reduce the risk or delay the outset of the disease. Such an amount is defined to be a prophylactically effective dose.
  • risk may be assessed by NMR imaging or, in some cases, by presymptomatic indications observed by the patient.
  • Effective dosage regimes of the compositions of the present invention, for the treatment of the above described conditions will vary depending upon many different factors, including means of administration, target site, physiological state of the patient, and other medicaments administered. Thus, treatment dosages will need to be titrated to optimize safety and efficacy.
  • each administration of the dosage regime will range from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg of the host body weight.
  • One preferred dosage regimen is 300 mg administered once per month for a period of at least 6 months, more preferably 12 months and perhaps over the course of several years.
  • Another dosage regimen that is preferred is a 3 mg per kilogram of patient weight per month. Such a regimen may be preferable for pediatric or adolescent patients in need of therapy.
  • the anti-alpha-4 agents of the invention can be used with effective amounts of other therapeutic agents against acute and chronic inflammation.
  • agents include other antagonists of adhesion molecules (e.g., other integrins, selectins, and immunoglobulin (Ig) super family members (see Springer, 1990 Nature 346: 425-433; Osborn, 1990 Cell 62: 3; Hynes, 1992 Cell 9: 11).
  • Integrins are heterodimeric transmembrane glycoproteins consisting of an a chain (120-180 kDa) and a ⁇ chain (90-110 kDa), generally having short cytoplasmic domains.
  • LFA-1 ⁇ L ⁇ 2
  • ICAM-1 Ig-family member counter-receptor
  • Mac-1 ( ⁇ M ⁇ 2 ) is distributed on neutrophils and monocytes, and also binds to ICAM-1.
  • the selectins consist of L-selectin, E-selectin and P-selectin.
  • anti-inflammatory agents that can be used in combination with the anti-alpha-4 agents include antibodies and other antagonists of cytokines, such as interleukins IL-1 through IL-13, tumor necrosis factors ⁇ and ⁇ , interferons ⁇ , ⁇ and ⁇ , tumor growth factor beta (TGF- ⁇ ), colony stimulating factor (CSF) and granulocyte monocyte colony stimulating factor (GM-CSF).
  • cytokines such as interleukins IL-1 through IL-13, tumor necrosis factors ⁇ and ⁇ , interferons ⁇ , ⁇ and ⁇ , tumor growth factor beta (TGF- ⁇ ), colony stimulating factor (CSF) and granulocyte monocyte colony stimulating factor (GM-CSF).
  • TGF- ⁇ tumor growth factor beta
  • CSF colony stimulating factor
  • GM-CSF granulocyte monocyte colony stimulating factor
  • Other anti-inflammatory agents include antibodies and other antagonists of chemokines such as MCP-1, MIP-1 ⁇ , MIP-1 ⁇ , RANTES
  • Additional agents for use in combination with agents which mediate alpha-4 integrin or dimers comprising alpha-4 integrin and treat inflammatory bowel disease (IBD), Crohn's Disease (CD) and ulcerative colitis (UC), include but are not limited to 5-aminosalicylates, glucocorticoids, thioguanine derivatives, methotrexate (MTX), cyclosporine, antibiotics, and infliximab.
  • 5-Aminosalicylates include sulfasalazine (also known as Azulfidine) which is a conjugate of mesalamine linked to sulfapyridine by a diazo bond and is usually administered in an amount of 500 mg/day to about 6 g/day.
  • 5-Aminosalicylates can also be co-administered with a glucocorticoid.
  • a 5-aminosalicylate is used in combination therapy with one of the other agents discussed herein to treat ulcerative colitis, however it can also be used to treat Crohn's disease.
  • Non-sulfonamide containing formulations of mesalamine include but are not limited to ASACOL®, CLAVERSA, SALOFALK, PENTASA®, DIPENTUM®, COLAZIDE and ROWASA®.
  • Glucocorticoids have been a mainstay of treatment for acute severe exacerbations of IBD since 1955, when they first where shown to be efficacious in UC.
  • Oral prednisone can be administered in conjunction with any of the agents disclosed herein. Typically, 20 to 40 mg of oral prednisone is administered once a day.
  • Glucocorticoids can also be administered intravenously and via enemas in combination with or concurrently with or within a short time before/after an anti-alpha-4 integrin agent is administered.
  • hydrocortisone is available as a retention enema (100 mg/60 mL) and the usual dose is one 60-mL enema per night for 2 to 3 weeks.
  • steroids that can be used include, but are not limited to, prednisolone methasulfobenzoate, tixocortol pivalate, fluticasone propionate, beclomethasone dipropionate, and budesonide.
  • Thioguanine derivatives are also useful in the treatment of IBD, CD and UC. These include but are not limited to 6-mercaptopurine (6-MP) and azathioprine (IMURAN).
  • 6-MP 6-mercaptopurine
  • IMURAN azathioprine
  • the two drugs can be used interchangeably in combination with any of the alpha-4 integrin modulating agents discussed herein.
  • Methotrexate is also contemplated for use in combination with the alpha-4 integrin regulatory agents discussed herein.
  • MTX is administered via intramuscular injection (i.m.) to the subject in combination with an anti-alpha-4 integrin agent.
  • MTX is effective in steroid-dependent CD, but not as useful in UC.
  • MTX can be administered in amounts of about 15 to about 25 mg per week per subject or as necessary as determined by the artisan of ordinary skill.
  • Cyclosporines e.g., SANDIMMUNE®, NEORAL®
  • SANDIMMUNE® can also be used in combination with the alpha-4 integrin modulating agents discussed herein to treat pathological inflammation of the bowel. This can be used to treat acute, severe UC, which does not respond to glucocorticoids.
  • Infliximab i.e., REMICADE®
  • REMICADE® can also be used to treat CD in combination with the alpha-4 integrin modulating agents indicated herein.
  • Infliximab is an immunoglobulin that binds to TNF and thereby neutralizes its activity.
  • Other anti-TNF antibodies such as CDP571, can also be used in combination with the alpha-4 integrin modulating agents disclosed herein.
  • Antibiotics are also contemplated for use in combination with the alpha-4 integrin modulating agents indicated herein to modulate UC, IBD and CD.
  • patients can be treated with metronidazole or ciprofloxacin (or pharmacological equivalents thereof) in combination with an alpha-4 integrin mediating agent or in the form of an admixture.
  • Supportive therapies include, but are not limited to, analgesics, anticholinergic and antidiarrheal agents. Combining such supportive therapies can be useful in the beginning of a treatment regimen in reducing a patient's symptoms and improving their quality of life. Supportive therapies include administering oral iron, folate, and vitamin B 12 .
  • Antidiarrheal agents include, but are not limited to diphenoxylate, codeine, loperamide, and anticholinergics (or pharmacological equivalents thereof), which can be administered to patients with mild disease to reduce the frequency of bowel movements and relive rectal urgency.
  • Cholestyramine can be used in patients to prevent bile salt-induced colonic secretion in patients who have already undergone limited ileocolic resections prior to treatment with the chronic regimens described herein.
  • Anticholinergic agents include, but are not limited to, clidinium bromide, dicyclomine hydrochloride, tincture of belladonna and the like, and are useful to reduce abdominal cramps, pain and rectal urgency.
  • the anti-alpha-4 integrin agents e.g., anti-alpha-4 integrin antibodies, small compound alpha-4 integrin antagonists and the like
  • agents utilized to treat, ameliorate or palliate symptoms associated with MS include but are not limited to: muscle relaxants (e.g., Diazepam, cyclobenzaprine, Clonazepam, clonidine, primidone, and the like), anticholinergics (e.g., propantheline, dicyclomine, and the like), central nervous system stimulants (e.g., Pemoline), non-steroidal anti-inflammatory agents (NS such as ibuprofen, naproxen and ketoprofen), interferons, immune globulin, glatiramer (Copaxone®), mitoxantrone (Novantrone®), misoprostol, tumor necrosis factor-alpha inhibitors (e.g., Pirfenidone, infliximab and the like) and corticosteroids (e.g., glucocorticoids and mineralocorticoids).
  • muscle relaxants e.g., Diazepam, cyclo
  • Common agents for treating multiple sclerosis include interferon beta-1b (Betaserong), interferon beta-1a (Avonex®), high-dose interferon beta-1a (Rebif), Glatiramer (Copaxone®), immune globulin, mitoxantrone (Novantrone®), corticosteroids (e.g., prednisone, methylprednisolone, dexamethasone and the like). Other corticosteroids may also be used and include but are not limited to cortisol, cortisone, fludrocortisone, prednisolone, 6 ⁇ -methylprednisolone, triamcinolone, and betamethasone.
  • corticosteroids e.g., prednisone, methylprednisolone, dexamethasone and the like.
  • corticosteroids e.g., prednisone, methylprednisolone, dexamethasone and
  • interferons are typically administered as follows: Interferon beta-1a (Avonex®) is administered 30 ⁇ g once a week; interferon beta-1a is administered at about 22 ⁇ g or 44 ⁇ g three times a week; and interferon beta-1b (Betaseron®) is administered at 250 ⁇ g on alternate days (Durelli et al., Lancet 359: 1453-60, 2002). Typically the interferons are administered for relapsing or remitting multiple sclerosis.
  • Interferon beta-1a Avonex®
  • interferon beta-1a is administered at about 22 ⁇ g or 44 ⁇ g three times a week
  • interferon beta-1b Betaseron®
  • the interferons are administered for relapsing or remitting multiple sclerosis.
  • preferred ranges of interferons may include about 0.1 ⁇ g to about 250 ⁇ g and more preferably about 0.5 ⁇ g to about 50 ⁇ g, depending on the manner in which the agent is administered in conjunction with the other anti-alpha-4 integrin compounds and compositions disclosed herein.
  • Non-selective COX inhibitors include but are not limited to salicylic acid derivatives (e.g., aspirin, sodium salicylates, choline magnesium trisalicylate, salsalate, diflunisal, sulfasalazine, and olsalazine), para-aminophenol derivatives (e.g., acetaminophen), indole and indene acetic acids (e.g., tolmetin, diclofenac, and ketorolac), heteroaryl acetic acids (e.g., abuprofen, naproxen, flurbiprofen, ketoprofen, fenprofen, and oxaprozin), anthranilic acids or fenamates (e.g., mefenamic acid and me
  • Selective COX-2 inhibitors include diaryl-substituted furanones (e.g., rofecoxib), diaryl-substituted pyrazoles (e.g., celecoxib), indole acetic acids (e.g., etodolac), and sulfonanilides (e.g., nimesulide).
  • NS are oftentimes administered in combination with interferon to lessen the flu-like symptoms experienced by patients receiving, for example, Avonex®.
  • Common NS agents include naproxen, ibuprofen and ketoprofen. Paracetamol is also frequently administered to patients. See, Reess et al., 2002 Mult. Scler. 8: 15-8.
  • Glatiramer acetate (GA, Copaxone®) is a synthetic molecule that inhibits activation of myelin basic protein-reactive T cells and induces a T-cell repertoire characterized by anti-inflammatory effects. Moreover, Glatiramer can access the central nervous system (CNS), whereas interferon-beta cannot (Dhib-Jalbut, 2002 Neurology 58: S3-9; Weinstock-Guttman et al., 2000 Drugs 59: 401-10).
  • CNS central nervous system
  • Mitoxantrone is an anthracenedione synthetic agent, which has been shown to be effective for treating secondary progressive multiple sclerosis (SP-MS). However, use of this drug is again limited by its cumulative cardiotoxicity (Weinstock-Guttman et al., 2000).
  • Tumor necrosis factor-alpha may be a key cytokine in demyelination (Walker et al., 2001 Mult. Scler. 7: 305-12).
  • agents that antagonize TNF- ⁇ function or inhibit its synthesis may be useful in combination with the agents and compounds disclosed herein.
  • This can include anti-TNF- ⁇ antibodies (e.g., infliximab) as well as agents such as pirfenidone.
  • Pirfenidone is a non-peptide drug, which has been shown to decrease synthesis of TNF- ⁇ and to block receptors for TNF- ⁇ . Id.
  • ACTH ACTH
  • glucocorticoids corticoid steroids
  • corticoid steroids glucocorticoids and corticoid steroids. These agents are used for their anti-edema and anti-inflammatory effects.
  • ACTH is commonly administered to a subject at 80 U given intravenously in 500 mL of 5% dextrose and water over 6-8 hours for 3 days. It may also be administered at 40 U/ml intramuscularly at a dose of 40 U every 12 hours for 7 days, with the dose then reduced every 3 days. See, S. Hauser, “Multiple sclerosis and other demyelinating diseases,” in Harrison 's Principles of Internal Medicine 2287-95 (13th ed., Isselbacher et al., ed.
  • Methylprednisolone is typically administered slowly in 500 ml D5W over 6 hours, preferably in the morning. Common dosages include 1000 mg daily for 3 days, 500 mg daily for 3 days and 250 mg daily for 3 days. Id. A methylprednisolone-prednisone combination is also commonly administered. Typically about 1,000 mg of intravenous methylprednisolone is administered over three days followed by oral prednisone at 1 mg/kg per day for 14 days. Thus, for use in combination with the compounds and compositions disclosed herein, the steroids may be administered in amounts ranging from about 1 to about 1,000 mg/kg over about 1 to 14 days, as needed.
  • Agents such as amantadine hydrochloride and pemoline have been frequently used to treat fatigue associated with MS (Geisler et al., 1996 Arch. Neurol. 53: 185-8).
  • the benefit of such combination therapies is that it may lessen the class-specific and agent-specific side effects currently encountered with some of the drugs.
  • Class-specific side effects of interferon-beta include fever, chills, myalgias, arthralgias and other flu-like symptoms beginning 2-6 hours after injection and typically resolving 24 hours post injection. Occasionally interferon-beta also induces transient worsening of preexisting MS symptoms.
  • Agent specific side effects include injection-site reactions with interferon beta-1b. Management of these effects can be accomplished by tailoring the dose and time of administration, prescribing appropriate combinations of acetaminophen, non-steroidal anti-inflammatory drugs (NS or NSAIDS) and steroids. See Munschauer et al., 1997 Clin. Ther. 19: 883-93.
  • combinations of drugs that can lessen the quantity of a particular drug administered may reduce adverse side effects experienced by a patient.
  • the small compound alpha-4-integrin antagonists may be administered in the same formulation as these other compounds or compositions, or in a separate formulation.
  • the anti-alpha-4-antibodies are generally administered in a separate formulation than the other compounds and compositions.
  • the anti-alpha-4 agents may be administered prior to, following, or concurrently with the other compounds and compositions used to treat, ameliorate, or palliate symptoms.
  • Eligible subjects were required to be age 18 through 65 years, with Poser criteria defined clinically or laboratory supported definite MS, either relapsing-remitting or secondary progressive, (Poser et al., 1983 Ann. Neurol. 13: 227-31; Lublin et al., 1996 Neurology 46: 907-11), a history of at least two relapses within the previous two years, a base-line Kurtzke Expanded Disability Status Score (EDSS) (Kurtzke, 1983 Neurology 33: 1444-52) between 2 and 6.5, and a minimum of three lesions on T 2 -weighted brain MRI. Patients were excluded if they received immunosuppressive or immunomodulating treatments within the past 3 months, or experienced a relapse, or received systemic corticosteroids within the past 30 days.
  • Poser criteria defined clinically or laboratory supported definite MS, either relapsing-remitting or secondary progressive, (Poser et al., 1983 Ann. Neurol. 13: 227
  • Patients were randomly assigned to one of three treatment groups: 3 mg/kg natalizumab, 6 mg/kg natalizumab, or placebo according to a computer-generated block randomization schedule. Patients received six intravenous infusions at 28-day intervals and then had six months of safety follow-up. The investigator, all other study personnel, and patients were blinded to treatment assignment.
  • Unenhanced proton density T 2 -weighted and Gd-enhanced T 1 -weighted MRI brain scans were obtained during the screening phase (month-1), immediately before each treatment (month 0-5), and one month after the last treatment (month 6).
  • Follow-up MRI scans were obtained at months 9 and 12.
  • Forty-six contiguous, 3-mm thick, axial slices through the brain were acquired.
  • MRI analysis was performed by a single center blinded to patient treatment and history. Lesions were identified on hard copy images by two experienced clinicians working by consensus.
  • the prospective primary outcome measure was the number of new Gd-enhancing lesions over the 6-month treatment period, defined as the period following the first infusion to one month after the last infusion.
  • Other MRI parameters evaluated included: the number of persistent Gd-enhancing lesions (enhancing lesions that had also enhanced on the previous monthly scan); the volume of Gd-enhancing lesions (measured by a semiautomated local thresholding method; Grimaud et al., 1996 Magn. Reson. Imaging 14: 495-505); the number of new active lesions (i.e., new Gd-enhancing lesions plus new or enlarging, non-enhancing T2 lesions); and the number of active scans (i.e., containing one or more new Gd-enhancing lesions).
  • Clinical endpoints included relapse frequency and changes in EDSS, and a self-reported global assessment using a visual-analog scale (VAS). All adverse events were recorded. Patients were examined at scheduled quarterly intervals, and at unscheduled visits for suspected relapses, by the treating and evaluating neurologists who were both unaware of the patient's treatment assignment. The treating neurologist performed a medical history and examination, and recorded adverse events. The evaluating neurologist assessed neurological status and assigned an EDSS score without knowledge of the patient's history or prior EDSS scores.
  • VAS visual-analog scale
  • An objective relapse was defined as the occurrence of an acute episode of new or worsening MS symptoms lasting at least 48 hours following a stable period of at least 30 days. It was also accompanied by an increase of at least one point in the EDSS score, an increase of at least one point on two functional system scores (FSS), or an increase of at least two points on one FSS compared with base-line, as determined by the evaluating neurologist. Neurological symptoms that did not meet the above criteria for relapse, but were assessed by the treating neurologist to constitute a relapse, were also recorded (total relapses).
  • VAS visual analog scale
  • Sample size estimates were based on the number of new Gd-enhancing lesions observed during the first 12 weeks following the first infusion in a previous clinical trial of natalizumab (Tubridy et al., 1999 Neurology 53: 466-72). Based on the results of this previous trial, and using sample size methodology appropriate for a two-sided, two-group comparison at the 5 percent level of significance, based on the Wilcoxon-Mann-Whitney statistic (Noether, 1987 J. Amer. Stat. Assoc. 82: 645-7), it was calculated that approximately 73 patients were needed in each group for 80 percent power.
  • Serum samples were collected from patients at each visit and analyzed quantitatively for antibodies directed specifically against natalizumab using an enzyme linked immunosorbant assay (ELISA). Natalizumab serum levels and receptor occupancy by natalizumab were also measured in a subgroup of 12 to 14 patients per treatment group before each infusion and at 2 hours, 24 hours, 1, 2, and 3 weeks after the first and last infusions.
  • ELISA enzyme linked immunosorbant assay
  • Serum antibody concentrations were determined using an ELISA assay.
  • a 2.0 ⁇ g/mL solution of a capture antibody that binds specifically to natalizumab was prepared in a solution containing sodium bicarbonate to pH 8.3.
  • 100 ⁇ L of the antibody solution was added to each well of a Costar 96-well microtiter plate. The plate was covered with plate sealing tape and incubated at ambient temperature for 12-26 hours. The plate was aspirated, 200 ⁇ L of blocking buffer (0.25% casein in PBS, pH 7.4) added to each well, and incubated for an additional hour at ambient temperature.
  • blocking buffer 0.25% casein in PBS, pH 7.4
  • Plates were then either dessicated and stored for later use, or washed once with 300 ⁇ L wash buffer (TBS, pH 7.5 containing 0.05% Tween-20). If plates were dessicated, they were rehydrated just prior to use by adding 300 ⁇ L wash buffer to each well and incubating for 1-2 minutes. Plates were aspirated and inverted onto tissue paper to absorb excess moisture.
  • 300 ⁇ L wash buffer TBS, pH 7.5 containing 0.05% Tween-20.
  • Test samples were diluted in casein diluent prior to the ELISA (0.25% casein in PBS, with 0.05% Tween 20, pH 7.4). Typically, two or three dilutions of each sample are tested to ensure the natalizumab values were accurate. Dilution control samples were also prepared using known quantities of natalizumab to monitor the accuracy of the remaining steps.
  • VLA-4 saturation was determined by an indirect immunoassay using flow cytometry.
  • the supernatant was then very carefully removed, 0.5 mL of a fixative solution (Ortho's Fixative pH 7.6) added, and the tubes immediately vortexed to ensure that cells were resuspended in the fixative.
  • the tubes were covered with aluminum foil until FACS assay.
  • Each sample was then for VLA-4 receptor saturation in the samples with and without analyzed natalizumab using the FACS Calibur flow cytometer and CellQuestTM software.
  • the CellQuestTM software allows acquisition and analysis of data from the flow cytometer.
  • Receptor saturation levels are shown in FIG. 5.
  • the levels of receptor saturation for months 1-4 were determined prior to that month's dosing.
  • the levels of receptor saturation produced by one month dosage intervals were consistently fairly high, and these chronic levels were sufficient to maintain a suppression of the pathological inflammation and associated physiological hallmarks of the disease.
  • the levels of saturation were maintained for a month at a mean of at least 67% and a median of at least 75%. These levels were sufficient for the suppression of brain lesions in the treated patients (see FIG. 1).
  • the minimum level of saturation determined in the study preinfusion from month 2 to month 5 (and week 21 following administration at month 5) is particularly low as compared to the mean and median values due to a single patient with an antibody response to natalizumab.
  • SAEs serious adverse events
  • 7 placebo-treated patients reported 11 SAEs, 5 patients receiving natalizumab 3 mg/kg reported 5 SAES, and 3 patients receiving natalizumab 6 mg/kg reported 4 SAES.
  • four were considered to be immune-mediated and related to study drug.
  • serum sickness one in each group including placebo. Only one event was accompanied by a change in complement levels and all occurred at the same investigative site. Overall, these events complicated fewer than one in 250 infusions.
  • natalizumab has demonstrated strong effects on clinically meaningful parameters in this placebo-controlled trial in patients with relapsing MS. Therapy was well tolerated during this six-month trial.
  • the beneficial effects of natalizumab on the appearance of new inflammatory CNS lesions, the occurrence of clinical relapses and improvement in patient well-being observed in this study indicate the potential for observing effects on disability in the longer term studies currently in progress.
  • CD Crohn's Disease Activity Index
  • QOL health-related quality of life
  • serum C-reactive protein levels serum C-reactive protein levels
  • Natalizumab increased the rates of clinical remission and clinical response, and improved QOL in patients with active CD, while demonstrating a safety profile acceptable for treatment of this disease.
  • each center screened male and female patients of at least 18 years of age who had clinical evidence of moderate to severely active CD defined as a CDAI of at least 220 but less than or equal to 450.
  • CDAI moderate to severely active CD
  • 248 were randomized at thirty-five study centers in Belgium, the Czech Republic, Denmark, Germany, Israel, the Netherlands, Sweden, and the United Kingdom from September 1999 to August 2000. All patients gave informed, written consent.
  • Patients who received methotrexate, cyclosporin, or any investigational agents within 3 months were excluded; patients receiving azathioprine or 6-mercaptopurine were required to have been on a stable dose for at least 4 months.
  • exclusions included prior antibody treatment; current use of oral prednisolone at a dose greater than 25 mg/day; current use of an elemental diet or parenteral nutrition; infectious or neoplastic diseases of the bowel; bowel surgery within 3 months; presence of a colostomy, ileostomy, or a colorectostomy with ileorectal anastomosis; symptoms due mainly to the presence of fibrotic strictures; and clinical impression that the patient was likely in the near term to require emergency abdominal surgery.
  • Eligible patients were randomly assigned to one of four treatment regimens according to a computer-generated block randomization schedule. Each group received two intravenous infusions spaced by a 4-week interval. The four treatment regimens were two infusions of placebo; one infusion of natalizumab at 3 mg/kg followed by placebo infusion; and two infusions of natalizumab at 3 or 6 mg/kg. The investigator, all other study personnel, and patients were blinded to treatment assignment.
  • the primary efficacy endpoint was the proportion of patients in remission (CDAI ⁇ 150) at week 6.
  • the CDAI incorporates eight related variables: number of liquid or very soft stools per day, severity of abdominal pain or cramping, general well-being, presence of extraintestinal manifestations of disease, presence of an abdominal mass, use of antidiarrheal drugs, hematocrit, and body weight (Best et al., 1976 Gastroenterology 70: 439-441; and Summers et al., 1979 Gastroenterology 77: 847-69). Scores less than 150 indicated remission; scores between 150 and 219 indicated mildly active disease, between 220 and 450 indicated moderately active disease, and scores greater than 450 indicated severe activity.
  • Additional prospective endpoints were the proportion of patients demonstrating clinical response (i.e., at least 70 point reduction in CDAI), health-related quality of life, as measured by an inflammatory bowel disease specific questionnaire, the IBDQ (Irvine et al., 1994 Gastroenterology 106: 287-96), and serum levels of C-reactive protein.
  • the sample size of 60 subjects per group was calculated to provide 80 percent power at a 5 percent significance level to detect a difference in response rates assuming a 40 percent response rate in the natalizumab groups and a 15 percent response rate in the placebo group.
  • C-reactive protein and IBDQ data were analyzed using the Wilcoxon-Mann-Whitney test to compare the change from base-line between each of the three active treatment groups and placebo.
  • the C-reactive protein levels were compared for patients with a value above the upper limit of the normal range of 8 mg/l at base-line (week 0).
  • base-line variables that may predict remission or clinical response.
  • the variables examined included site of disease, duration of disease, base-line CDAI scores, concomitant use of oral steroids, concomitant use of azathioprine or 6-mercaptopurine, and extraintestinal symptoms.
  • Base-line CDAI score was a significant predictor of remission (P ⁇ 0.001); patients with a higher base-line CDAI were less likely to achieve remission. However, base-line CDAI did not predict the likelihood of response. All other variables analyzed were not significant predictors of remission or response.
  • Anti-natalizumab antibodies were detected in 13 natalizumab-treated patients (7 percent) at week 12. Overall, patients with detectable anti-natalizumab antibodies were no more likely to experience an adverse event or serious adverse event than those who had no detectable anti-natalizumab antibodies.
  • a patient in the 3+3 mg/kg natalizumab group experienced symptoms of mild itching and erythema, and was subsequently found to be positive for anti-natalizumab antibodies.
  • a patient in the 6+6 mg/kg natalizumab group experienced symptoms of mild itching and coughing. These symptoms resolved without treatment, and the patient was subsequently found to be negative for detectable anti-natalizumab antibodies.
  • the dosage levels of receptor saturation in the CD trial are comparable to the dosages of the MS trial, and thus receptor saturation levels should be comparable in the CD trial.
  • Receptor saturation levels in the CD trial are thus associated with decreased levels of inflammation, as demonstrated by C-reactive protein levels and an improvement in overall patient well-being, as demonstrated by the CDAI.
  • alpha-4 integrin antagonism is an effective mechanism for treating CD raises the possibility that this modality may be more broadly applicable to the treatment of chronic autoimmune and inflammatory diseases.
  • IBD inflammatory bowel disease
  • alpha-4 integrins are key mediators of leukocyte migration across the vascular endothelium, being that they are expressed on all leucocytes except neutrophils, the effects of a single 3 mg/kg natalizumab (Antegren®) infusion on basic circulating leukocyte subsets, natural killer (NK) cells and activated T cells in active inflammatory bowel disease patients (IBD).
  • natalizumab had also previously been shown that a single 3 mg/kg infusion of natalizumab produced a sustained rise in circulating peripheral blood leucocytes in animals and healthy volunteers (“A six-month weekly intravenous toxicity study with AntegrenTM in cynomolgous monkeys with a six-week recovery,” Athena Report 1998, No. 723-013-098). However, it was unknown whether natalizumab could have differential effects on leukocyte subsets, given that all leucocytes except neutrophils express alpha-4 integrins.
  • Venous blood was taken immediately prior to natalizumab/placebo infusion and again at one, two, four, eight and twelve weeks post-infusion.
  • the LymphoprepTM method (Nycomed, Denmark) was used to isolate the peripheral blood lymphocytes (PBLs) prior to analysis by multi-color fluorescence-activated cell sorter (FACS; Becton Dickenson, Oxford, UK) in conjunction with Consort 30 software (Amlot et al., 1996 Clin. Exp. Immunol. 105: 176-82).
  • the percentages of PBLs expressing the following markers were measured using FACS analysis: CD19 (B cell), TCR ⁇ (T cell), CD3 (pan-T cell), TCR ⁇ (T cell), CD4 (helper/Th-1 T cell), CD8 (cytotoxic/suppressor T cell) and CD16 (NK cell).
  • TCR ⁇ cells expressing the activation antigens CD38, CD25 (a chain of interleukin-2 receptor), CD26, CD69 and HLA-DR were measured, in addition to naive (CD45RA) and memory (CD45RO) T cell subsets and “NK-T cells” (CD57 + /CD3 + ).
  • CD45RA naive
  • CD45RO memory
  • NK-T cells CD57 + /CD3 +
  • CD8 + cytotoxic/suppressor T cells expressing the activation antigens CD28 and HLA-DR were also measured.
  • Eosinophil, monocyte, B and T cell counts were all significantly raised for ⁇ I week post-natalizumab.
  • Neutrophil and basophil counts were unchanged.
  • T cells expressing the activation markers CD25, CD26, HLA-DR, CD8DR, CD8, CD28, CD45RO and CD45RA were significantly raised compared to baseline at ⁇ 4 and 1 week in UC and CD patients respectively.
  • CD38 + and CD69 + T cells were raised at ⁇ 1 week in UC patients only.
  • NK cells were unchanged post-infusion in all patients and NK-type T cells (CD57 + ) were raised at 1 week in CD patients only.
  • FIGS. 9A and 9B The effect of natalizumab on circulating eosinophils and monocytes are shown in FIGS. 9A and 9B.
  • FIGS. 10 and 11 demonstrate the effects of natalizumab administration on specific circulating T cell subsets and natural killer cells in Crohn's disease and ulcerative colitis patients. The error bars denote standards of deviation in each chart.
  • a single 3 mg/kg natalizumab infusion produced increased circulating levels of most, but not all, leukocyte subsets in patients with active IBD. Circulating eosinophil, monocyte and lymphocyte counts were significantly elevated above baseline values for at least four weeks post-infusion in most patients. A wide range of circulating T cell subsets were significantly increased above pretreatment values, particularly those expressing activation antigens. However, NK cell counts (CD16 + /CD3 ⁇ ) were not affected by natalizumab, and CD57 + T cells were affected to a much lesser extent by natalizumab than other T cell subsets. Lymphocytes expressing the ⁇ T cell receptor were also not affected by natalizumab, suggesting that alpha-4 integrins are either not expressed, or are expressed at a lower level on these cells.
  • natalizumab may limit trafficking and maintain in circulation many leukocyte and activated lymphocyte subsets.
  • the NK cells, ⁇ cells, neutrophils and basophils appear unaffected by the administration of natalizumab, which may suggest that they are less important mediators in the trafficking of these cell types.
US10/372,111 2002-02-25 2003-02-25 Administration of agents for the treatment of inflammation Abandoned US20040009169A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US10/372,111 US20040009169A1 (en) 2002-02-25 2003-02-25 Administration of agents for the treatment of inflammation
US11/540,640 US7807167B2 (en) 2002-02-25 2006-10-02 Administration of agents for the treatment of inflammation
US12/846,350 US20110064729A1 (en) 2002-02-25 2010-07-29 Administration of agents for the treatment of inflammation
US14/613,821 US20150152182A1 (en) 2002-02-25 2015-02-04 Administration of agents for the treatment of inflammation
US15/586,724 US20170306026A1 (en) 2002-02-25 2017-05-04 Administration of agents for the treatment of inflammation
US16/411,042 US11248051B2 (en) 2002-02-25 2019-05-13 Administration of agents for the treatment of inflammation
US17/578,294 US20220162321A1 (en) 2002-02-25 2022-01-18 Administration of agents for the treatment of inflammation

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US36013402P 2002-02-25 2002-02-25
US37450102P 2002-04-23 2002-04-23
US10/372,111 US20040009169A1 (en) 2002-02-25 2003-02-25 Administration of agents for the treatment of inflammation

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/540,640 Division US7807167B2 (en) 2002-02-25 2006-10-02 Administration of agents for the treatment of inflammation

Publications (1)

Publication Number Publication Date
US20040009169A1 true US20040009169A1 (en) 2004-01-15

Family

ID=27767592

Family Applications (7)

Application Number Title Priority Date Filing Date
US10/372,111 Abandoned US20040009169A1 (en) 2002-02-25 2003-02-25 Administration of agents for the treatment of inflammation
US11/540,640 Expired - Lifetime US7807167B2 (en) 2002-02-25 2006-10-02 Administration of agents for the treatment of inflammation
US12/846,350 Abandoned US20110064729A1 (en) 2002-02-25 2010-07-29 Administration of agents for the treatment of inflammation
US14/613,821 Abandoned US20150152182A1 (en) 2002-02-25 2015-02-04 Administration of agents for the treatment of inflammation
US15/586,724 Abandoned US20170306026A1 (en) 2002-02-25 2017-05-04 Administration of agents for the treatment of inflammation
US16/411,042 Expired - Lifetime US11248051B2 (en) 2002-02-25 2019-05-13 Administration of agents for the treatment of inflammation
US17/578,294 Abandoned US20220162321A1 (en) 2002-02-25 2022-01-18 Administration of agents for the treatment of inflammation

Family Applications After (6)

Application Number Title Priority Date Filing Date
US11/540,640 Expired - Lifetime US7807167B2 (en) 2002-02-25 2006-10-02 Administration of agents for the treatment of inflammation
US12/846,350 Abandoned US20110064729A1 (en) 2002-02-25 2010-07-29 Administration of agents for the treatment of inflammation
US14/613,821 Abandoned US20150152182A1 (en) 2002-02-25 2015-02-04 Administration of agents for the treatment of inflammation
US15/586,724 Abandoned US20170306026A1 (en) 2002-02-25 2017-05-04 Administration of agents for the treatment of inflammation
US16/411,042 Expired - Lifetime US11248051B2 (en) 2002-02-25 2019-05-13 Administration of agents for the treatment of inflammation
US17/578,294 Abandoned US20220162321A1 (en) 2002-02-25 2022-01-18 Administration of agents for the treatment of inflammation

Country Status (28)

Country Link
US (7) US20040009169A1 (no)
EP (6) EP3674322A1 (no)
JP (3) JP2005526045A (no)
KR (1) KR20040105740A (no)
CN (1) CN1310678C (no)
AR (1) AR038605A1 (no)
AT (1) ATE512163T1 (no)
AU (2) AU2003213231A1 (no)
BR (1) BR0307975A (no)
CA (1) CA2477178A1 (no)
CO (1) CO5611176A2 (no)
CY (1) CY1111787T1 (no)
DK (2) DK1485127T3 (no)
ES (1) ES2531852T3 (no)
HK (1) HK1073997A1 (no)
IL (3) IL163725A0 (no)
MX (1) MXPA04008267A (no)
MY (1) MY157727A (no)
NO (1) NO336235B1 (no)
NZ (3) NZ591635A (no)
PL (1) PL215263B1 (no)
PT (2) PT1485127E (no)
RU (1) RU2359697C2 (no)
SA (1) SA03240276A (no)
SI (2) SI2336184T1 (no)
TW (2) TWI346558B (no)
WO (1) WO2003072040A2 (no)
ZA (1) ZA200407406B (no)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050232917A1 (en) * 2004-01-09 2005-10-20 Nicholas Pullen Antibodies to MAdCAM
WO2005099776A2 (en) 2004-04-01 2005-10-27 Elan Pharmaceuticals, Inc. Steroid sparing agents and methods of using same
US20050255118A1 (en) * 2004-02-06 2005-11-17 Nancy Wehner Methods and composition for treating tumors and metastatic disease
WO2006023629A2 (en) * 2004-08-20 2006-03-02 Biogen Idec Ma Inc. Treatment of pediatric multiple sclerosis
WO2006055871A3 (en) * 2004-11-19 2006-08-03 Biogen Idec Inc Treatment for multiple sclerosis
WO2007007160A2 (en) * 2005-07-11 2007-01-18 Pfizer Limited Anti-madcam antibodies to treat fever
US20070207141A1 (en) * 2006-02-28 2007-09-06 Ivan Lieberburg Methods of treating inflammatory and autoimmune diseases with natalizumab
WO2007103112A2 (en) 2006-03-03 2007-09-13 Elan Pharmaceuticals, Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
US20080044382A1 (en) * 2006-02-28 2008-02-21 Ivan Lieberburg Methods of treating inflammatory and autoimmune diseases with alpha-4 inhibitory compounds
US20080261894A1 (en) * 2005-02-17 2008-10-23 Rivka Kreitman Combination Therapy with Glatiramer Acetate and Rasagiline for the Treatment of Multiple Sclerosis
US20090169477A1 (en) * 2004-12-03 2009-07-02 Michael Panzara Delaying or preventing onset of multiple sclerosis
US20100266587A1 (en) * 2009-04-17 2010-10-21 Biogen Idec Ma Inc. Compositions and Methods to Treat Acute Myelogenous Leukemia
US20120258093A1 (en) * 2009-08-20 2012-10-11 Institut National De La Sante Et De La Recherche Medicale (Inserm) Vla-4 as a biomarker for prognosis and target for therapy in duchenne muscular dystrophy
AU2011232775B2 (en) * 2004-08-20 2013-11-28 Biogen Ma Inc. Extended Treatment of Multiple Sclerosis
US20140120084A1 (en) * 2011-03-31 2014-05-01 Genentech, Inc. Methods of administering beta7 integrin antagonists
US20140271532A1 (en) * 2005-02-17 2014-09-18 Teva Pharmaceutical Industries, Ltd. Combination therapy with glatiramer acetate and rasagiline for the treatment of multiple sclerosis
AU2012202823B2 (en) * 2005-04-04 2014-09-25 Biogen Ma Inc. Methods and Products for Evaluating an Immune Response to a Therapeutic Protein
US8871449B2 (en) 2005-04-04 2014-10-28 Biogen Idec Ma Inc. Methods and products for evaluating an immune response to a therapeutic protein
AU2012202575B2 (en) * 2004-11-19 2014-11-20 Biogen Ma Inc. Treatment for Multiple Sclerosis
US9663579B2 (en) 2011-05-02 2017-05-30 Millennium Pharmaceuticals, Inc. Formulation for anti-α4β7 antibody
US9763992B2 (en) 2014-02-13 2017-09-19 Father Flanagan's Boys' Home Treatment of noise induced hearing loss
US10040855B2 (en) 2011-05-02 2018-08-07 Millennium Pharmaceuticals, Inc. Formulation for anti-α4β7 antibody
JP2018530593A (ja) * 2015-10-16 2018-10-18 ノースウェスタン ユニバーシティ 統合失調症、双極性障害、認知障害及び大うつ病性障害の治療のための非定型的抗精神病薬ならびにnmda調節剤の医薬品の組み合わせ
US10335485B2 (en) 2010-04-16 2019-07-02 Biogen Ma Inc. Anti-VLA-4 antibodies
US10444234B2 (en) 2010-01-11 2019-10-15 Biogen Ma Inc. Assay for JC virus antibodies
US10677803B2 (en) 2013-05-28 2020-06-09 Biogen Ma Inc. Method of assessing risk of PML
US20210147547A1 (en) * 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
US11104661B1 (en) 2019-10-16 2021-08-31 Morphic Therapeutic, Inc. Inhibiting human integrin (α-4) (β-7)
US20210332139A1 (en) * 2013-10-03 2021-10-28 Leuvas Therapeutics Modulation of leukocyte activity in treatment of neuroinflammatory degenerative disease
US11174228B2 (en) 2018-04-12 2021-11-16 Morphic Therapeutic, Inc. Antagonists of human integrin (α4)(β7)
US11280793B2 (en) 2009-10-11 2022-03-22 Biogen Ma Inc. Anti-VLA-4 related assays
US11287423B2 (en) 2010-01-11 2022-03-29 Biogen Ma Inc. Assay for JC virus antibodies
US11802156B2 (en) 2017-07-14 2023-10-31 Pfizer Inc. Antibodies to MAdCAM

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL215263B1 (pl) 2002-02-25 2013-11-29 Elan Pharm Inc Kompozycja do przewleklego leczenia patologicznego zapalenia i zastosowanie natalizumabu lub jego immunologicznie aktywnego fragmentu do wytwarzania leku
WO2003100033A2 (en) 2002-03-13 2003-12-04 Biogen Idec Ma Inc. ANTI-αvβ6 ANTIBODIES
CA2514117A1 (en) * 2003-01-24 2004-08-12 Elan Pharmaceuticals, Inc. Composition for and treatment of demyelinating diseases and paralysis by administration of remyelinating agents
KR20050110628A (ko) * 2003-02-10 2005-11-23 엘란 파마슈티칼스, 인크. 면역글로불린 제제 및 이의 제조방법
AR044927A1 (es) * 2003-06-25 2005-10-12 Elan Pharm Inc Metodos y composiciones para tratar la artritis reumatoidea
AU2011213878B2 (en) * 2004-04-01 2012-06-14 Biogen Ma Inc. Steroid sparing agents and methods of using same
CA2590471A1 (en) * 2004-12-22 2006-06-29 Laboratoires Serono S.A. Combination treatment for multiple sclerosis
CN102875681A (zh) 2005-07-08 2013-01-16 拜奥根Idec马萨诸塞公司 抗-αvβ6抗体及其用途
BRPI0714381A2 (pt) 2006-07-10 2013-03-05 Biogen Idec Inc composiÇÕes e mÉtodos para inibir crescimento de cÂnceres deficientes em smad4
EP2053961A4 (en) * 2006-08-09 2013-05-01 Biogen Idec Inc METHOD FOR DISPENSING A MEDICAMENT
EP2109480B1 (en) 2006-12-07 2017-06-28 The Government of the United States of America as Represented by the Secretary of the Department of Health and Human Services Use of antagonists of the interaction between hiv gp120 and alpha4beta7 integrin
BRPI0901298A2 (pt) 2009-04-06 2011-01-04 Ems Sa derivados ftalimìdicos de compostos antiinflamatórios não-esteróide e/ou moduladores de tnf-(alfa), processo de sua obtenção, composições farmacêuticas contendo os mesmos e seus usos no tratamento de doenças inflamatórias
EP3722808A1 (en) 2010-10-25 2020-10-14 Biogen MA Inc. Methods for determining differences in alpha-4 integrin activity by correlating differences in svcam and/or smadcam levels
IN2014MN01568A (no) * 2012-01-12 2015-05-08 Pharma Two B Ltd
TWI640298B (zh) * 2012-12-05 2018-11-11 美商提拉諾斯股份有限公司 用於體液採樣之系統、裝置及方法
WO2014144466A1 (en) 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Anti-alpha v beta 6 antibodies and uses thereof
US10035860B2 (en) 2013-03-15 2018-07-31 Biogen Ma Inc. Anti-alpha V beta 6 antibodies and uses thereof
EP3127545B1 (en) * 2014-04-04 2020-06-03 Taiho Pharmaceutical Co., Ltd. Anti-tumor drug containing taxane compound, and anti-tumor effect enhancer
CN106573065A (zh) * 2014-05-09 2017-04-19 诺格尔制药有限公司 用于治疗炎症性肠病的方法
MX2017010336A (es) * 2015-02-26 2017-12-20 Genentech Inc Antagonistas de integrina beta7 y métodos para el tratamiento de la enfermedad de crohn.
MA49645A (fr) * 2017-07-20 2021-03-31 Univ Virginia Patent Foundation Procédés de traitement ou de prévention d'un trouble neurologique du système immunitaire
WO2022160323A1 (zh) * 2021-01-31 2022-08-04 兰州大学 核苷类似物在制备预防或治疗胃肠道疾病药物中的应用
WO2022221458A1 (en) 2021-04-13 2022-10-20 Biogen Ma Inc Compositions and methods for treating chronic active white matter lesions / radiologically isolated syndrome
WO2023035000A1 (en) * 2021-09-03 2023-03-09 Pliant Therapeutics, Inc. Therapeutic modulation of integrins

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5716648A (en) * 1993-06-08 1998-02-10 Farmaceutisk Laboratorium Ferring A/S Compositions for use in the regulation of subnormal pH values in the intestinal tract and for treatment of bowel diseases
US5932214A (en) * 1994-08-11 1999-08-03 Biogen, Inc. Treatment for inflammatory bowel disease with VLA-4 blockers
US20010046496A1 (en) * 2000-04-14 2001-11-29 Brettman Lee R. Method of administering an antibody
US6602885B2 (en) * 1999-05-04 2003-08-05 Schering Corporation Piperidine derivatives useful as CCR5 antagonists
US20050215565A1 (en) * 2003-01-24 2005-09-29 Karlik Stephen J Composition for and treatment of demyelinating diseases and paralysis by administration of remyelinating agents

Family Cites Families (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0832981A1 (en) 1987-02-17 1998-04-01 Pharming B.V. DNA sequences to target proteins to the mammary gland for efficient secretion
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US4912094B1 (en) 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
JPH037237A (ja) * 1989-03-14 1991-01-14 Sandoz Ag シクロスポリン類投与の新規用途および治療手段
HU212924B (en) 1989-05-25 1996-12-30 Chiron Corp Adjuvant formulation comprising a submicron oil droplet emulsion
US5730978A (en) * 1989-09-01 1998-03-24 Fred Hutchinson Cancer Research Center Inhibition of lymphocyte adherence with α4β1-specific antibodies
IL113261A (en) 1989-09-01 1996-10-16 Hutchinson Fred Cancer Res Repression by peptides of lymphocyte binding to vascular endothelium by interaction of extracellular uterine capacitor with connective tissue, and pharmaceutical preparations containing
US5260210A (en) * 1989-09-27 1993-11-09 Rubin Lee L Blood-brain barrier model
US6033665A (en) * 1989-09-27 2000-03-07 Elan Pharmaceuticals, Inc. Compositions and methods for modulating leukocyte adhesion to brain endothelial cells
SE464961B (sv) 1989-11-15 1991-07-08 Volvo Ab Ledprotes, i synnerhet foer fingerleder
EP0501983B1 (de) 1989-11-20 1994-12-28 BECKER, Wolfgang Verfahren zur herstellung einer zahnprothese
ATE356869T1 (de) 1990-01-12 2007-04-15 Amgen Fremont Inc Bildung von xenogenen antikörpern
GB9206318D0 (en) 1992-03-24 1992-05-06 Cambridge Antibody Tech Binding substances
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US6172197B1 (en) 1991-07-10 2001-01-09 Medical Research Council Methods for producing members of specific binding pairs
US5264563A (en) 1990-08-24 1993-11-23 Ixsys Inc. Process for synthesizing oligonucleotides with random codons
JP3854306B2 (ja) 1991-03-06 2006-12-06 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング ヒト化及びキメラモノクローナル抗体
US6225447B1 (en) 1991-05-15 2001-05-01 Cambridge Antibody Technology Ltd. Methods for producing members of specific binding pairs
US5858657A (en) 1992-05-15 1999-01-12 Medical Research Council Methods for producing members of specific binding pairs
DE69230142T2 (de) 1991-05-15 2000-03-09 Cambridge Antibody Tech Verfahren zur herstellung von spezifischen bindungspaargliedern
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
US5224539A (en) 1991-06-14 1993-07-06 Coen Company, Inc. Cooling system for air heaters and the like
LU91067I2 (fr) 1991-06-14 2004-04-02 Genentech Inc Trastuzumab et ses variantes et dérivés immuno chimiques y compris les immotoxines
ES2136092T3 (es) 1991-09-23 1999-11-16 Medical Res Council Procedimientos para la produccion de anticuerpos humanizados.
WO1993011236A1 (en) 1991-12-02 1993-06-10 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
US5872215A (en) 1991-12-02 1999-02-16 Medical Research Council Specific binding members, materials and methods
DE69309906T2 (de) 1992-02-12 1997-11-06 Biogen Inc Behandlung für entzündungserkrankung des darmes
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5336894A (en) 1992-04-21 1994-08-09 The United States Of America As Represented By The Secretary Of The Air Force Universal infrared heat source controller
JP3720353B2 (ja) 1992-12-04 2005-11-24 メディカル リサーチ カウンシル 多価および多重特異性の結合タンパク質、それらの製造および使用
DK0678122T3 (da) 1993-01-12 2000-03-06 Biogen Inc Rekombinante anti-VLA4 antistofmolekyler
US5827690A (en) 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
EP1759709B1 (en) 1994-01-25 2013-02-27 Elan Pharmaceuticals Inc. Humanized antibodies against leukocyte adhesion molecule VLA-4
US5840299A (en) 1994-01-25 1998-11-24 Athena Neurosciences, Inc. Humanized antibodies against leukocyte adhesion molecule VLA-4
GB9406974D0 (en) 1994-04-08 1994-06-01 Pharmaceutical Proteins Ltd Transgenic production
CA2193508A1 (en) 1994-07-11 1996-01-25 Eugene D. Thorsett Inhibitors of leukocyte adhesion
US6001809A (en) 1994-07-11 1999-12-14 Elan Pharmaceuticals, Inc. Inhibitors of leukocyte adhesion
US6113898A (en) 1995-06-07 2000-09-05 Idec Pharmaceuticals Corporation Human B7.1-specific primatized antibodies and transfectomas expressing said antibodies
EP1281718A3 (en) 1995-11-10 2005-01-19 ELAN CORPORATION, Plc Peptides which enhance transport across tissues
GB9712818D0 (en) 1996-07-08 1997-08-20 Cambridge Antibody Tech Labelling and selection of specific binding molecules
IL119989A0 (en) 1997-01-10 1997-04-15 Yeda Res & Dev Pharmaceutical compositions for oral treatment of multiple sclerosis
WO1998040100A1 (en) 1997-03-10 1998-09-17 Ottawa Civic Loeb Research Institute USE OF NUCLEIC ACIDS CONTAINING UNMETHYLATED CpG DINUCLEOTIDE AS AN ADJUVANT
WO1998053817A1 (en) 1997-05-29 1998-12-03 Merck & Co., Inc. Biarylalkanoic acids as cell adhesion inhibitors
EP1148889A2 (en) 1999-01-29 2001-10-31 Millennium Pharmaceuticals, Inc. Method of treating demyelinating inflammatory disease using ccr1 antagonists
MXPA02011163A (es) 2000-05-10 2004-08-19 Mayo Foundation Anticuerpos igm humanos con la capacidad de inducir remielinacion, y usos diagnosticos y terapeuticos de los mismos particularmente en el sistema nervioso central.
PL215263B1 (pl) 2002-02-25 2013-11-29 Elan Pharm Inc Kompozycja do przewleklego leczenia patologicznego zapalenia i zastosowanie natalizumabu lub jego immunologicznie aktywnego fragmentu do wytwarzania leku
KR20050110628A (ko) * 2003-02-10 2005-11-23 엘란 파마슈티칼스, 인크. 면역글로불린 제제 및 이의 제조방법
US7163939B2 (en) 2003-11-05 2007-01-16 Abbott Laboratories Macrocyclic kinase inhibitors
NZ581497A (en) * 2004-12-03 2012-07-27 Biogen Idec Inc Delaying or preventing onset of multiple sclerosis by vla-4 ginding antibody
EP1863940B1 (en) * 2005-03-03 2012-06-06 Seedlings Life Science Ventures, LLC. Method of risk management for patients undergoing natalizumab treatment
EP2645106B1 (en) * 2005-04-04 2017-06-14 Biogen MA Inc. Methods for evaluating an immune response to a therapeutic agent
EP3620469A1 (en) * 2006-02-28 2020-03-11 Biogen MA Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5716648A (en) * 1993-06-08 1998-02-10 Farmaceutisk Laboratorium Ferring A/S Compositions for use in the regulation of subnormal pH values in the intestinal tract and for treatment of bowel diseases
US5932214A (en) * 1994-08-11 1999-08-03 Biogen, Inc. Treatment for inflammatory bowel disease with VLA-4 blockers
US6602885B2 (en) * 1999-05-04 2003-08-05 Schering Corporation Piperidine derivatives useful as CCR5 antagonists
US20010046496A1 (en) * 2000-04-14 2001-11-29 Brettman Lee R. Method of administering an antibody
US20050215565A1 (en) * 2003-01-24 2005-09-29 Karlik Stephen J Composition for and treatment of demyelinating diseases and paralysis by administration of remyelinating agents

Cited By (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9328169B2 (en) 2004-01-09 2016-05-03 Pfizer Inc. Human antibodies that bind human MAdCAM
US10259872B2 (en) 2004-01-09 2019-04-16 Pfizer, Inc. Antibodies to MAdCAM
US20070166308A1 (en) * 2004-01-09 2007-07-19 Nicholas Pullen Antibodies to MAdCAM
US20050232917A1 (en) * 2004-01-09 2005-10-20 Nicholas Pullen Antibodies to MAdCAM
US7932372B2 (en) 2004-01-09 2011-04-26 Amgen Fremont Inc. Antibodies to MAdCAM
US20080124339A1 (en) * 2004-01-09 2008-05-29 Nicholas Pullen Antibodies to MAdCAM
USRE45847E1 (en) 2004-01-09 2016-01-19 Pfizer Inc. Antibodies to MAdCAM
US20050255118A1 (en) * 2004-02-06 2005-11-17 Nancy Wehner Methods and composition for treating tumors and metastatic disease
JP2013166765A (ja) * 2004-04-01 2013-08-29 Elan Pharmaceuticals Inc ステロイド節約剤およびそれを使用する方法
WO2005099776A3 (en) * 2004-04-01 2006-05-18 Elan Pharm Inc Steroid sparing agents and methods of using same
TWI462744B (zh) * 2004-04-01 2014-12-01 Biogen Idec Inc 類固醇節約劑及彼之使用方法
JP2007531761A (ja) * 2004-04-01 2007-11-08 エラン ファーマシューティカルズ,インコーポレイテッド ステロイド節約剤およびそれを使用する方法
WO2005099776A2 (en) 2004-04-01 2005-10-27 Elan Pharmaceuticals, Inc. Steroid sparing agents and methods of using same
WO2006023629A3 (en) * 2004-08-20 2006-09-28 Biogen Idec Inc Treatment of pediatric multiple sclerosis
WO2006023651A3 (en) * 2004-08-20 2006-09-21 Biogen Idec Inc Extended treatment of multiple sclerosis
AU2011232775B2 (en) * 2004-08-20 2013-11-28 Biogen Ma Inc. Extended Treatment of Multiple Sclerosis
WO2006023649A3 (en) * 2004-08-20 2006-06-22 Biogen Idec Inc Treatment of severe multiple sclerosis
WO2006023629A2 (en) * 2004-08-20 2006-03-02 Biogen Idec Ma Inc. Treatment of pediatric multiple sclerosis
WO2006036371A3 (en) * 2004-08-20 2006-12-21 Biogen Idec Inc Combination therapy
WO2006023649A2 (en) * 2004-08-20 2006-03-02 Biogen Idec Ma Inc. Treatment of severe multiple sclerosis
AU2005277343B2 (en) * 2004-08-20 2011-07-07 Biogen Ma Inc. Extended treatment of multiple sclerosis
US20090010926A1 (en) * 2004-08-20 2009-01-08 Biogen Idec Ma Inc. Extended Treatment of Multiple Sclerosis
WO2006036371A2 (en) * 2004-08-20 2006-04-06 Biogen Idec Ma Inc. Combination therapy
EP2783701A3 (en) * 2004-08-20 2014-12-17 Biogen Idec MA Inc. Extended treatment of multiple sclerosis
US20080260728A1 (en) * 2004-08-20 2008-10-23 Biogen Idec Ma Inc. Treatment of Severe Multiple Sclerosis
EP2808033A1 (en) * 2004-11-19 2014-12-03 Biogen Idec MA Inc. Treatment for multiple sclerosis
CN101102792A (zh) * 2004-11-19 2008-01-09 比奥根艾迪克Ma公司 治疗多发性硬化
US20090202527A1 (en) * 2004-11-19 2009-08-13 Biogen Idec Ma Inc. Treatment for multiple sclerosis
WO2006055871A3 (en) * 2004-11-19 2006-08-03 Biogen Idec Inc Treatment for multiple sclerosis
AU2012202575B2 (en) * 2004-11-19 2014-11-20 Biogen Ma Inc. Treatment for Multiple Sclerosis
US20090169477A1 (en) * 2004-12-03 2009-07-02 Michael Panzara Delaying or preventing onset of multiple sclerosis
AU2005311635B2 (en) * 2004-12-03 2012-02-02 Biogen Ma Inc. Delaying or preventing onset of multiple sclerosis
US20080261894A1 (en) * 2005-02-17 2008-10-23 Rivka Kreitman Combination Therapy with Glatiramer Acetate and Rasagiline for the Treatment of Multiple Sclerosis
US20140271532A1 (en) * 2005-02-17 2014-09-18 Teva Pharmaceutical Industries, Ltd. Combination therapy with glatiramer acetate and rasagiline for the treatment of multiple sclerosis
AU2012202823B2 (en) * 2005-04-04 2014-09-25 Biogen Ma Inc. Methods and Products for Evaluating an Immune Response to a Therapeutic Protein
US9709575B2 (en) 2005-04-04 2017-07-18 Biogen Ma Inc. Methods and products for evaluating an immune response to a therapeutic protein
US10705095B2 (en) 2005-04-04 2020-07-07 Biogen Ma Inc. Methods and products for evaluating an immune response to a therapeutic protein
US11686734B2 (en) 2005-04-04 2023-06-27 Biogen Ma Inc. Methods and products for evaluating an immune response to a therapeutic protein
US8871449B2 (en) 2005-04-04 2014-10-28 Biogen Idec Ma Inc. Methods and products for evaluating an immune response to a therapeutic protein
WO2007007160A3 (en) * 2005-07-11 2007-05-18 Pfizer Ltd Anti-madcam antibodies to treat fever
WO2007007160A2 (en) * 2005-07-11 2007-01-18 Pfizer Limited Anti-madcam antibodies to treat fever
US20080044382A1 (en) * 2006-02-28 2008-02-21 Ivan Lieberburg Methods of treating inflammatory and autoimmune diseases with alpha-4 inhibitory compounds
US10233245B2 (en) 2006-02-28 2019-03-19 Biogen Ma Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
US11292845B2 (en) * 2006-02-28 2022-04-05 Biogen Ma Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
US8410115B2 (en) 2006-02-28 2013-04-02 Elan Pharmaceuticals, Inc. Methods of treating inflammatory and autoimmune diseases with alpha-4 inhibitory compounds
US20070207141A1 (en) * 2006-02-28 2007-09-06 Ivan Lieberburg Methods of treating inflammatory and autoimmune diseases with natalizumab
US9493567B2 (en) 2006-02-28 2016-11-15 Biogen Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
US20100196318A1 (en) * 2006-02-28 2010-08-05 Elan Pharmaceuticals, Inc. Methods of Treating Inflammatory and Autoimmune Diseases with Natalizumab
WO2007103112A2 (en) 2006-03-03 2007-09-13 Elan Pharmaceuticals, Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
WO2007103112A3 (en) * 2006-03-03 2007-11-29 Elan Pharm Inc Methods of treating inflammatory and autoimmune diseases with natalizumab
EP3360900A1 (en) * 2006-03-03 2018-08-15 Biogen MA Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
AU2007224228B2 (en) * 2006-03-03 2013-09-19 Biogen Ma Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
US20070231319A1 (en) * 2006-03-03 2007-10-04 Yednock Theodore A Methods of treating inflammatory and autoimmune diseases with natalizumab
EP1991267B1 (en) 2006-03-03 2017-12-20 Biogen MA Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
US11333670B2 (en) 2006-03-03 2022-05-17 Biogen Ma Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
US10466251B2 (en) 2006-03-03 2019-11-05 Biogen Ma Inc. Methods of treating inflammatory and autoimmune diseases with natalizumab
EA016626B1 (ru) * 2006-03-03 2012-06-29 Элан Фамэсьютикэлс, Инк. Способ лечения натализумабом воспалительного и/или аутоиммунного заболевания (варианты)
US20100266587A1 (en) * 2009-04-17 2010-10-21 Biogen Idec Ma Inc. Compositions and Methods to Treat Acute Myelogenous Leukemia
US20120258093A1 (en) * 2009-08-20 2012-10-11 Institut National De La Sante Et De La Recherche Medicale (Inserm) Vla-4 as a biomarker for prognosis and target for therapy in duchenne muscular dystrophy
US11280793B2 (en) 2009-10-11 2022-03-22 Biogen Ma Inc. Anti-VLA-4 related assays
US10444234B2 (en) 2010-01-11 2019-10-15 Biogen Ma Inc. Assay for JC virus antibodies
US11287423B2 (en) 2010-01-11 2022-03-29 Biogen Ma Inc. Assay for JC virus antibodies
US11571477B2 (en) 2010-04-16 2023-02-07 Biogen Ma Inc. Anti-VLA-4 antibodies
US10335485B2 (en) 2010-04-16 2019-07-02 Biogen Ma Inc. Anti-VLA-4 antibodies
US11083791B2 (en) 2010-04-16 2021-08-10 Biogen Ma Inc. Anti-VLA-4 antibodies
US20140120084A1 (en) * 2011-03-31 2014-05-01 Genentech, Inc. Methods of administering beta7 integrin antagonists
US9764033B2 (en) 2011-05-02 2017-09-19 Millennium Pharmaceuticals, Inc. Formulation for anti-α4β7 antibody
US10040855B2 (en) 2011-05-02 2018-08-07 Millennium Pharmaceuticals, Inc. Formulation for anti-α4β7 antibody
US9663579B2 (en) 2011-05-02 2017-05-30 Millennium Pharmaceuticals, Inc. Formulation for anti-α4β7 antibody
TWI664978B (zh) * 2011-05-02 2019-07-11 美商千禧製藥公司 抗-α4β7抗體之用途
US11560434B2 (en) 2011-05-02 2023-01-24 Millennium Pharmaceuticals, Inc. Formulation for anti-α4β7 antibody
US10004808B2 (en) 2011-05-02 2018-06-26 Millennium Pharmaceuticals, Inc. Methods of treating ulcerative colitis
US10143752B2 (en) 2011-05-02 2018-12-04 Millennium Pharmaceuticals, Inc. Methods of treating ulcerative colitis
US10677803B2 (en) 2013-05-28 2020-06-09 Biogen Ma Inc. Method of assessing risk of PML
US11280794B2 (en) 2013-05-28 2022-03-22 Biogen Ma Inc. Method of assessing risk of PML
US20210332139A1 (en) * 2013-10-03 2021-10-28 Leuvas Therapeutics Modulation of leukocyte activity in treatment of neuroinflammatory degenerative disease
US9763992B2 (en) 2014-02-13 2017-09-19 Father Flanagan's Boys' Home Treatment of noise induced hearing loss
JP2018530593A (ja) * 2015-10-16 2018-10-18 ノースウェスタン ユニバーシティ 統合失調症、双極性障害、認知障害及び大うつ病性障害の治療のための非定型的抗精神病薬ならびにnmda調節剤の医薬品の組み合わせ
US11802156B2 (en) 2017-07-14 2023-10-31 Pfizer Inc. Antibodies to MAdCAM
US11174228B2 (en) 2018-04-12 2021-11-16 Morphic Therapeutic, Inc. Antagonists of human integrin (α4)(β7)
US20210147547A1 (en) * 2018-04-13 2021-05-20 Novartis Ag Dosage Regimens For Anti-Pd-L1 Antibodies And Uses Thereof
US11370773B1 (en) 2019-10-16 2022-06-28 Morphic Therapeutic, Inc. Inhibiting human integrin (alpha-4) (beta-7)
US11104661B1 (en) 2019-10-16 2021-08-31 Morphic Therapeutic, Inc. Inhibiting human integrin (α-4) (β-7)

Also Published As

Publication number Publication date
ATE512163T1 (de) 2011-06-15
BR0307975A (pt) 2005-01-11
US20170306026A1 (en) 2017-10-26
KR20040105740A (ko) 2004-12-16
US20070025989A1 (en) 2007-02-01
PT2336184E (pt) 2015-03-09
EP2289941A2 (en) 2011-03-02
WO2003072040A2 (en) 2003-09-04
NO336235B1 (no) 2015-06-22
EP2360185A3 (en) 2012-01-11
US11248051B2 (en) 2022-02-15
CO5611176A2 (es) 2006-02-28
DK2336184T3 (en) 2015-02-16
RU2359697C2 (ru) 2009-06-27
JP2011140493A (ja) 2011-07-21
EP1485127A2 (en) 2004-12-15
DK1485127T3 (da) 2011-10-03
HK1073997A1 (en) 2005-10-28
RU2004128454A (ru) 2005-05-10
SI1485127T1 (sl) 2011-09-30
US20110064729A1 (en) 2011-03-17
TW200400047A (en) 2004-01-01
PL215263B1 (pl) 2013-11-29
US20150152182A1 (en) 2015-06-04
EP2336184A2 (en) 2011-06-22
EP2360185A2 (en) 2011-08-24
PL372306A1 (en) 2005-07-11
SI2336184T1 (sl) 2015-04-30
IL163725A (en) 2013-07-31
ES2531852T3 (es) 2015-03-20
NZ579583A (en) 2011-04-29
ZA200407406B (en) 2005-11-30
EP3173426A1 (en) 2017-05-31
AU2009201587B2 (en) 2012-01-19
SA03240276A (ar) 2005-12-03
EP2289941A3 (en) 2012-01-18
JP2015042629A (ja) 2015-03-05
MY157727A (en) 2016-07-15
IL163725A0 (en) 2005-12-18
US7807167B2 (en) 2010-10-05
US20200148773A1 (en) 2020-05-14
NZ535320A (en) 2008-05-30
CN1646163A (zh) 2005-07-27
NZ591635A (en) 2012-06-29
EP3674322A1 (en) 2020-07-01
US20220162321A1 (en) 2022-05-26
EP1485127B1 (en) 2011-06-08
WO2003072040A3 (en) 2004-05-21
EP1485127A4 (en) 2006-02-01
TWI346558B (en) 2011-08-11
PT1485127E (pt) 2011-09-07
AR038605A1 (es) 2005-01-19
AU2003213231A1 (en) 2003-09-09
EP2336184B1 (en) 2014-12-03
EP2336184A3 (en) 2012-01-11
JP2005526045A (ja) 2005-09-02
MXPA04008267A (es) 2004-11-10
NO20044054L (no) 2004-11-24
CN1310678C (zh) 2007-04-18
AU2009201587A1 (en) 2009-05-14
TW201125584A (en) 2011-08-01
CY1111787T1 (el) 2015-10-07
IL203515A (en) 2015-10-29
CA2477178A1 (en) 2003-09-04

Similar Documents

Publication Publication Date Title
US11248051B2 (en) Administration of agents for the treatment of inflammation
US20150056205A1 (en) Steroid sparing agents and methods of using same
AU2014265076B2 (en) Administration of agents for the treatment of inflammation
AU2012202155B2 (en) Administration of agents for the treatment of inflammation
ES2367480T3 (es) Administración de agentes para el tratamiento de inflamación.
AU2011213878B2 (en) Steroid sparing agents and methods of using same
TW202311293A (zh) 免疫療法之組合及其用途

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: BIOGEN IDEC INTERNATIONAL HOLDING LTD., BERMUDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ELAN PHARMA INTERNATIONAL LIMITED;REEL/FRAME:030155/0822

Effective date: 20130402

AS Assignment

Owner name: BIOGEN IDEC MA INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BIOGEN IDEC INTERNATIONAL HOLDING LTD.;REEL/FRAME:031214/0500

Effective date: 20130819

Owner name: BIOGEN IDEC INTERNATIONAL HOLDING LTD., BERMUDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ELAN PHARMACEUTICALS, INC.;REEL/FRAME:031214/0424

Effective date: 20130724