AU2009200156B2 - Method of administering an antibody - Google Patents

Method of administering an antibody Download PDF

Info

Publication number
AU2009200156B2
AU2009200156B2 AU2009200156A AU2009200156A AU2009200156B2 AU 2009200156 B2 AU2009200156 B2 AU 2009200156B2 AU 2009200156 A AU2009200156 A AU 2009200156A AU 2009200156 A AU2009200156 A AU 2009200156A AU 2009200156 B2 AU2009200156 B2 AU 2009200156B2
Authority
AU
Australia
Prior art keywords
days
immunoglobulin
day
human
dose
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
AU2009200156A
Other versions
AU2009200156A1 (en
Inventor
David Edward Allison
Lee R. Brettman
Judith A. Fox
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.)
Genentech Inc
Millennium Pharmaceuticals Inc
Original Assignee
Genentech Inc
Millennium Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech Inc, Millennium Pharmaceuticals Inc filed Critical Genentech Inc
Priority to AU2009200156A priority Critical patent/AU2009200156B2/en
Publication of AU2009200156A1 publication Critical patent/AU2009200156A1/en
Application granted granted Critical
Publication of AU2009200156B2 publication Critical patent/AU2009200156B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Classifications

    • 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
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Peptides Or Proteins (AREA)

Abstract

METHOD OF ADMINISTERING AN ANTIBODY Abstract Disclosed is a method for treating a human having a disease associated with leukocyte infiltration of mucosal tissues, comprising administering to said human an 5 effective amount of a human or humanized immunoglobulin or antigen-binding fragment thereof having binding specificity for ax4p7 integrin. Preferably, no more than about 8 mg immunoglobulin or fragment per kg body weight are administered during a period of about one month.

Description

S&F Ref: 611851D2 AUSTRALIA PATENTS ACT 1990 COMPLETE SPECIFICATION FOR A STANDARD PATENT Name and Address Millennium Pharmaceuticals, Inc., of 75 Sidney Street, of Applicants: Cambridge, Massachusetts, 02139, United States of America Genentech Inc., of 1 DNA Way, South San Francisco, California, 94080-4990, United States of America Actual Inventor(s): David Edward Allison, Lee R. Brettman, Judith A. Fox Address for Service: Spruson & Ferguson St Martins Tower Level 35 31 Market Street Sydney NSW 2000 (CCN 3710000177) Invention Title: Method of administering an antibody The following statement is a full description of this invention, including the best method of performing it known to me/us: 5845c(1 928523_1) METH OD OF ADMIMSTERING AN ANTIBODY RELATED APPLICATIONS This application is a continuation of U.S. Application No. 09/748,960, filed on December 27, 2000, which is a continuation of U.S. Application No. 09/550,082,, 5 filed on April 14, 2000. The entire teachings of each of the above applications are incorporated herein by reference. BACKGROUND OF TFE INVENTION Integrin receptors are important for regulating both lymphocyte recirculation and recruitment to sites of inflammation (Carlos, T.M. and Harlan, J.M., Blood, 10 84:2068-2101 (1994)). The human a4p7 integrin has several ligands, one of which is the mucosal vascular addressin MAdCAM-1 (Berlin, C., et al., Cell 74:185-195 (1993); Erle, D.J., et al., J Immunol. 153:517-528 (1994)) expressed on high endothelial venules in mesenteric lymph nodes and Peyer's patches (Streeter, P.R., et al., Nature 331:41-46 (1988)). As such, the a4p7 integrin acts as a homing 15 receptor that mediates lymphocyte migration to intestinal mucosal lymphoid tissue (Schweighoffer, T., et al., J Immunol. 151:717-729 (1993)). In addition, the a4p7 integrin interacts with fibronectin and vascular cell adhesion molecule-I (VCAM-1). Inflammatory bowel disease (IBD), such as ulcerative colitis and Crohn's disease, for example, can be a debilitating and progressive disease involving 20 inflammation of the gastrointestinal tract. Affecting an estimated two million people in the United States alone, symptoms include abdominal pain, cramping, diarrhea and rectal bleeding. IBD treatments have included anti-inflammatory drugs (such as, corticosteroids and sulfasalazine), immunosuppressive drugs (such as, 6-mercaptopurine, cyclosporine and azathioprine) and surgery (such as, colectomy).
2 Podolsky, New Engl. J. Med., 325: 928-937 (1991) and Podolsky, New Engl. J. Med., 325: 1008-1016 (1991). However, such therapeutic agents have not been effective in maintaining remission of IBD. Antibodies against human a4p7 integrin, such as murine monoclonal antibody 5 Act-1 (mAb Act-1), interfere with a4p7 integrin binding to mucosal addressin cell adhesion molecule-1 (MAdCAM-1) present on high endothelial venules in mucosal lymph nodes. Act-I was originally isolated by Lazarovits, A. I., et al., JImmunol. 133: 1857-1862 (1984), from mice immunized with human tetanus toxoid-specific T lymphocytes and was reported to be a mouse IgG1/K antibody. More recent analysis of 1o the antibody by Schweighoffer, T., et al., J. Immunol. 151: 717-729 (1993) demonstrated that it can bind to a subset of human memory CD4+ T lymphocytes which selectively express the cA47 integrin. However, a serious problem with using murine antibodies for therapeutic applications in humans is that they are highly immunogenic in humans and quickly induce a human anti-murine antibody response (HAMA), which reduces the is efficacy of the mouse antibody in patients and can prevent continued administration. The HAMA response results in rapid clearance of the mouse antibody, severely limiting any therapeutic benefit. Thus, a need exists for improved therapeutic approaches to inflammatory bowel diseases and other inflammatory disorders of mucosal tissues. 20 Summary of the Invention According to a first aspect of the invention, there is provided a method for treating a human having a disease associated with leukocyte infiltration of mucosal tissues, comprising administering to said human an effective amount of a human immunoglobulin or antigen-binding fragment thereof having binding specificity for a4p7 integrin, wherein 25 said immunoglobulin or antigen-binding fragment thereof specifically binds a4p7 but does not bind a4pl, wherein said immunoglobulin or fragment is administered in an initial dose followed by one or more subsequent doses and the minimum interval between any two doses is a period of at least about I day, and wherein no more than about 8 mg immunoglobulin or antigen-binding fragment per kg body weight are administered during 30 a period of about one month.
2a According to a second aspect of the invention, there is provided a method for treating a human having inflammatory bowel disease, comprising administering to said human an effective amount of a human immunoglobulin or antigen-binding fragment thereof having binding specificity for a407 integrin, wherein said immunoglobulin or 5 antigen-binding fragment thereof specifically binds a4p7 but does not bind a4plI, wherein said immunoglobulin or fragment is administered in an initial dose followed by one or more subsequent doses and the minimum interval between any two doses is a period of at least about 1 day, and wherein no more than about 8 mg immunoglobulin or fragment per kg body weight are administered during a period of about one month. 10 According to a third aspect of the invention, there is provided a method for inhibiting relapse and/or recurrence of quiescent inflammatory bowel disease in a human, comprising administering to said human an effective amount of a human immunoglobulin or antigen-binding fragment thereof having binding specificity for a4p7 integrin, wherein said immunoglobulin or antigen-binding fragment thereof specifically binds a4p7 but 15 does not bind a4pl, wherein said immunoglobulin or fragment is administered in doses and the minimum interval between doses is a period of at least about 7 days, and wherein no more than about 8 mg immunoglobulin or fragment per kg body weight are administered during a period of about 30 days. The invention relates to a method of administering an antibody (e. g., humanized 20 antibody, human antibody). In one aspect the invention is a method of treating a human having a disease associated with leukocyte infiltration of mucosal tissues comprising administering to the human an effective amount of an immunoglobulin having binding specificity for ax4p7 integrin. In preferred embodiments no more than about 8 mg immunoglobulin per kg body weight is administered in a period of about one month. In 25 particular embodiments, the immunoglobulin can include one or more complementarity determining regions (CDRs) having the amino acid sequence of a CDR of murine Act-i mAb. LDP-02 is -3 a preferred antibody for administration. The immunoglobulin can be administered in multiple doses and the interval between doses can be at least 1 day or longer. In particular embodiments, the interval between doses can be at least about 7, 14 or 21 days or about one month. In one embodiment, the amount of immunoglobulin 5 administered per dose can be an amount which is sufficient to achieve about 50% or greater saturation of a4P7 binding sites on circulating lymphocytes and/or about 50% or greater inhibition of a4$7 integrin expression on the surface of circulating lymphocytes for a period of at least about 10 days following administration of the dose. In another embodiment, the amount of immunoglobulin administered per dose 10 can be an amount which is sufficient to achieve and maintain a serum concentration of said immunoglobulin of at least about I pig/mL for a period of about 10 days following administration of the dose. The immunoglobulin can be administered alone or together with one or more . other agents to treat a disease associated with leukocyte infiltration of mucosal 15 tissues. For example, the immunoglobulin can be administered with steroids, immunosuppressive agents, non-steroidal anti-inflammatory agents or immunomodulators. In a preferred embodiment, immunoglobulin is administered to treat a human having an inflammatory bowel disease, such as Crohn's disease or ulcerative colitis. 20 BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 is an illustration of the nucleotide sequence (SEQ ID NO: 1) and deduced amino acid sequence (SEQ ID NO:2) of the mouse (Mus musculus) Act-I light chain variable region joined to the mouse Act-1 light chain signal peptide sequence. 25 FIG. 2 is an illustration of the nucleotide sequence (SEQ ID NO:3) and amino acid sequence (SEQ ID NO:4) of the mouse Act-I antibody heavy chain variable region. The nucleotide sequence of the variable region is joined to a nucleotide sequence which encodes a deduced mouse Act-I heavy chain signal peptide sequence, to yield a composite sequence. (The identity of the primer which 30 amplified the heavy chain region was deduced from the degenerate sequence, and an -4 amino acid sequence for the signal peptide was derived from the primer, downstream sequence and sequences of other signal peptides. The signal peptide shown may not be identical to that of the Act-I hybridoma.) FIG. 3 is an illustration of the nucleotide sequence (SEQ ID NO:5) and 5 amino acid sequence (SEQ ID NO:6) of a portion of the heavy chain of a humanized Act-I antibody (LDP-02) with a heavy chain signal peptide. FIG. 4 is an illustration of the nucleotide sequence (SEQ ID NO:7) and amino acid sequence (SEQ ID NO:8) of a portion of the light chain of a humanized Act-I antibody (LDP-02) with a light chain signal peptide. 10 FIG. 5 is an illustration of the amino acid sequence of the light chain complementarity determining regions (CDR1, SEQ ID NO: 9; CDR2, SEQ ID. NO:10; CDR3, SEQ ID NO: 11) and heavy chain complementarity determining regions (CDR1, SEQ ID NO: 12; CDR2, SEQ ID NO:13; CDR3, SEQ ID NO:14) of murine antibody Act-1 and LDP-02. 15 FIG. 6 is a graph showing mean serun LDP-02 levels (pg/mL) in healthy men over time following a single administration of LDP-02. Mean serum LDP-02 levels became negligible by day 36 following administration of 0.15 mg/kg by intravenous (IV)(-*- ) or subcutaneous (SC)(- I- ) injection and following administration of 0.5 mg/kg by intravenous injection (- A-). However serum 20 LDP-02 was still measurable beyond day 36 following administration of 1.5 mg/kg (-x-) or 2.5 mg/kg (- * -) by intravenous injection. FIG. 7 is a graph showing persistent loss of a4p7 signal (detected with Act-i mAb) following administration of LDP-02. About 90% of a4p7 signal was rapidly lost (MESF =10%) after administration of LDP-02 and persisted following 25 administration of all LDP-02 doses. Between about day 7 and day 22, a4p7 signal started to return to baseline for the 0.15 mg/kg IV dose group (- U- ) and for the 0.15 mg/kg SC dose group (-+-). Between day 22 and day 36, a4P7 signal started to return to baseline for the 0.5 mg/kg IV (- A-) dose group. At the higher doses of LDP-02 studied (1.5 mg/kg (-x-) and 2.5 mg/kg (- *-)), loss of a4p7 signal persisted 30 for longer than 36 days following single IV doses. For the 2.5 mg/kg dose group -5 (- *-), loss of a4b7 signal persisted up to about Day 70 (data provided herein in Appendix to Study L297-007). MESF: mean equivalent soluble fluorescence. FIG. 8 is a graph showing mean serum LDP-02 levels (pg/mL) in patients with ulcerative colitis over time following a single administration of LDP-02. Mean 5 serum LDP-02 levels rose rapidly following administration of LDP-02. The concentration of serum LDP-02 fell to below 1.0 gg/mL in patients administered LDP-02 at 0.15 mg/kg by intravenous (- A-) or subcutaneous (-*-) injection by 10 days following dosing. However, serum LDP-02 concentrations remained above 1.0 pg/mL for about 20 days following administration of 0.5 mg/kg by intravenous 10 injection (- 0- ). The serum concentration of LDP-02 remained above I Jig/mL for about 60 days following administration of 2.0 mg/kg by intravenous injection (-Y-). FIG. 9 is a graph showing persistent loss of a4P7 signal (detected with Act-1 mAb) following administration of LDP-02. About 90% of a4p7 signal was rapidly - lost (MESF =10%) after administration of LDP-02 and the duration of signal loss 15 was dependent upon dose. Starting at about Day 10, a4p7 signal started to return to baseline for the group administered 0.15 mg/kg of LDP-02 by IV (- U-) or SC (-+- ) injection. However, a4p7 signal started to return to baseline between day 30 and day 60 for the group administered 0.5 mg/kg (- A-) intravenously, and after day 60 for the group administered 2.0 mg/kg (-x-) intravenously (data provided herein in 20 Appendix to Study L297-006). MESF: mean equivalent soluble fluorescence. DETAILED DESCRIPTION OF THE INVENTION The present invention relates to a method of administering an antibody (immunoglobulin) to a subject. In one aspect, the antibody to be administered is a human or humanized antibody having binding specificity for a4p7 integrin (e.g., 25 mammalian a4P7 (e.g., human (Homo sapiens) a4p7). Preferably, the human or humanized immunoglobulins can bind a4p7 integrin with an affinity of at least about 10' M", preferably at least about 10 M4, and more preferably at least about I O'M4. In one embodiment, the humanized .immunoglobulin includes an antigen binding region of nonhuman origin which binds a4p7 integrin and a constant region -6 derived from a human constant region. In another embodiment, the humanized immunoglobulin which binds a4p7 integrin comprises a complementarity determining region of nonhuman origin and a variable framework region of human origin, and if desired, a constant region of human origin. For example, the 5 humanized immunoglobulin can comprise a heavy chain and a light chain, wherein the light chain comprises a complementarity determining region derived from an antibody of nonhuman origin which binds a407 integrin and a framework region derived from a light chain of human origin, and the heavy chain comprises a complementarity determining region derived from an antibody of nonhuman origin 10 which binds a4p7 integrin and a framework region derived from a heavy chain of human origin. Naturally occurring immunoglobulins have a common core structure in which two identical light chains (about 24 kD) and two identical heavy chains (about 55 or 70 kD) form a tetramer. The amino-terminal portion of each chain is known as 15 the variable (V) region and can be distinguished from the more conserved constant . (C) regions of the remainder of each chain. Within the variable region of the light chain is a C-terminal portion known as the J region. Within the variable region of the heavy chain, there is a D region in addition to the J region. Most of the amino acid sequence variation in immunoglobulins is confined to three separate locations 20 in the V regions known as hypervariable regions or complementarity determining regions (CDRs) which are directly involved in antigen binding. Proceeding f&om the amino-terminus, these regions are designated CDRI, CDR2 and CDR3, respectively. The CDRs are held in place by more conserved framework regions (FRs). Proceeding from the amino-terminus, these regions are designated FRI, FR2, FR3, 25 and FR4, respectively. The locations of CDR and FR regions and a numbering system have been defined by Kabat et al. (Kabat, E.A. et al., Sequences of Proteins ofImmunological Interest, Fifth Edition, U.S. Department of Health and Human Services, U.S. Government Printing Office (1991)). Human immunoglobulins can be divided into classes and subclasses, 30 depending on the isotype of the heavy chain. The classes include IgG, IgM, IgA, IgD and IgE, in which the heavy chains are of the gamma (y), mu (v), alpha (a), -7 delta (6) or epsilon (E) type, respectively. Subclasses include IgG1, IgG2, IgG3, IgG4, IgAl and IgA2, in which the heavy chains are of the y 1, y2, y3, y4, al and a2 type, respectively. Human immunoglobulin molecules of a selected class or subclass may contain either a kappa (K) or lambda (A) light chain. See e.g., Cellular and 5 Molecular Immunology, Wonsiewicz, M.J., Ed., Chapter 45, pp. 41-50, W. B. Saunders Co, Philadelphia, PA (1991); Nisonoff, A., Introduction to Molecular Immunology, 2nd Ed., Chapter 4, pp. 45-65, Sinauer Associates, Inc., Sunderland, MA (1984). The term "immunoglobulin" as used herein includes whole antibodies and 10 biologically functional fragments thereof. Such biologically functional fragments retain at least one antigen binding function of a corresponding full-length antibody (e.g., specificity for a407 of Act-i antibody), and preferably, retain the ability to inhibit the interaction of a 4 07 with one or more of its ligands (e.g., MAdCAM-1, fibronectin). In a particularly preferred embodiment, biologically functional 15 fragments can inhibit binding of a407 to the mucosal addressin (MAdCAM-1). Examples of biologically functional antibody fragments which can be administered as described herein include fragments capable of binding to an a4P7 integrin, such as single chain antibodies, Fv, Fab, Fab' and F(ab') 2 fragments. Such fragments can be produced by enzymatic cleavage or by recombinant techniques. For example, 20 papain or pepsin cleavage can generate Fab or F(ab') 2 fragments, respectively. Other proteases with the requisite substrate specificity can also be used to generate Fab, F(ab') 2 or other antigen-binding fragments. Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site. For example, a chineric 25 gene encoding a F(ab') 2 heavy chain portion can be designed to include DNA sequences encoding the CH, domain and hinge region of the heavy chain. The term "humanized immunoglobulin" as used herein refers to an immunoglobulin (antibody) comprising portions of immunoglobulins of different origin, wherein at least one portion is of human origin. For example, the humanized 30 antibody can comprise portions derived from an immunoglobulin of nonhuman origin with the requisite specificity, such as a mouse, and from immunoglobulin -8 sequences of human origin (e.g., chimeric immunoglobulin), joined together chemically by conventional techniques (e.g., synthetic) or prepared as a contiguous polypeptide using recombinant DNA technology (e.g., DNA encoding the protein portions of the chimeric antibody can be expressed to produce a contiguous 5 polypeptide chain). Another example of a humanized immunoglobulin is an immunoglobulin containing one or more immunoglobulin chains comprising a CDR derived from an antibody of nonhuman origin and a framework region derived from a light and/or heavy chain of human origin (e.g., CDR-grafted antibodies with or without framework changes). Chimeric or CDR-grafted single chain antibodies are 10 also encompassed by the term humanized immunoglobulin. See, e.g., Cabilly et al., U.S. Patent No. 4,816,567; Cabilly et al., European Patent No. 0,125,023 B1; Boss et al., U.S. Patent No. 4,816,397; Boss et al., European Patent No. 0,120,694 B1; Neuberger, M.S. et al., WO 86/01533; Neuberger, M.S. et al., European Patent No. 0,194,276 B1; Winter, U.S. Patent No. 5,225,539; Winter, European Patent No. 15 0,239,400 B 1; Queen et al., European Patent No. 0 451 216 B 1; Padlan, E.A. et al., European Patent Application No. 0,519,596 Al. -See also, Ladner et al., U.S. Patent No. 4,946,778; Huston, U.S. Patent No. 5,476,786; and Bird, R.E. et al., Science, 242: 423-426 (1988)), regarding single chain antibodies. In particular embodiments, the humanized immunoglobulin can include an immunoglobulin chain (e.g., heavy 20 chain) having a variable region of non-human origin (e.g., murine origin) and at least a portion of a human constant region (e.g, Cyl), and an immunoglobulin chain (e.g., light chain) where at least one CDR is of non-human origin (e.g., murine origin) and the framework regions (FRI, FR2, FR3, FR4) and, optionally, the constant region (e.g., CK, CX) are of human origin. 25 The antigen binding region of the humanized immunoglobulin (the nonhuman portion) can be derived from an immunoglobulin of nonhuman origin (referred to as a donor immunoglobulin) having binding specificity for a4p7 integrin. For example, a suitable antigen binding region can be derived from the murine Act-i monoclonal antibody (Lazarovits, A.I. et al., J. Immunol., 133(4): 30 1857-1862 (1984)). Other sources include a4p7 integrin-specific antibodies obtained from nonhuman sources, such as rodent (e.g., mouse, rat), rabbit, pig, goat -9 or non-human primate (e.g., monkey). Other polyclonal or monoclonal antibodies, such as antibodies which bind to the same or similar epitope as the Act-I antibody, or LDP-02, can be made (e.g., Kohler et al., Nature, 256:495-497 (1975); Harlow et al., 1988, Antibodies: A Laboratory Manual, (Cold Spring Harbor, NY); and Current 5 Protocols in Molecular Biology, Vol. 2 (Supplement 27, Summer '94), Ausubel et al., Eds. (John Wiley & Sons: New York, NY), Chapter 11 (1991)). For example, antibodies can be raised against an appropriate immunogen in a suitable mammal (e.g., a mouse, rat, rabbit, sheep). Preparation of immunizing antigen, and polyclonal and monoclonal antibody production can be performed using 10 any suitable technique. A variety of methods have been described (see e.g., Kohler et al., Nature, 256: 495-497 (1975) and Eur. J Immunol. 6: 511-519 (1976); Milstein et al., Nature 266: 550-552 (1977); Koprowski et al., U.S. Patent No. 4,172,124; Harlow, E. and D. Lane, 1988, Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory: Cold Spring Harbor, NY); Current Protocols In 15 Molecular Biology, Vol. 2 (Supplement 27, Summer '94), Ausubel, F.M. et al., Eds., (John Wiley & Sons: New York, NY), Chapter 11, (1991)). For example, suitable immunizing agents include cells bearing a437, membrane fractions containing a4p7, immunogenic fragments of suitable immunogens include a4p7, a P7 peptide conjugated to a suitable carrier and the like. Antibody-producing cells (e.g., a 20 lymphocyte) can be isolated from, for example, the lymph nodes or spleen of an immunized animal. The cells can then be fused to a suitable immortalized cell (e.g., a myeloma cell line (e.g., SP2/0, P3x63Ag8.653)), thereby forming a hybridoma. Fused cells can be isolated employing selective culturing techniques. Cells which produce antibodies with the desired specificity can be selected using a suitable assay 25 (e.g., ELISA). Other suitable methods of producing or isolating antibodies (human antibodies, non-human antibodies) of the requisite specificity can be used, including, for example, methods which select recombinant antibody from a library (e.g., a phage display library). Transgenic animals capable of producing a repertoire of human antibodies (e.g., XenoMouseTM (Abgenix, Fremont, CA) can be produced 30 using suitable methods (see e.g., WO 98/24893 (Abgenix), published June 11, 1998; Kucherlapate, R. and Jakobovits, A., U.S. Patent No. 5,939,598; -10 Jakobovits et al., Proc. Natl. Acad. Sci. USA, 90: 2551-2555 (1993); Jakobovits et al., Nature, 362: 255-258 (1993)). Additional methods for production of transgenic animals capable of producing a repertoire of human antibodies have been described (e.g., Lonberg et al., U.S. Patent No. 5,545,806; Surani et al., U.S. Patent No. 5 5,545,807; Lonberg et aL., W097/13852). In one embodiment, the antigen binding region of the humanized immunoglobulin comprises a CDR of nonhuman origin. In this embodiment, the humanized immunoglobulin having binding specificity for a4P7 integrin comprises at least one CDR of nonhuman origin. For example, CDRs can be derived from the 10 light and heavy chain variable regions of immunoglobulins of nonhuman origin, such that a humanized immunoglobulin includes substantially heavy chain CDRI, CDR2 and/or CDR3, and/or light chain CDRI, CDR2 and/or CDR3, from one or more immunoglobulins of nonhuman origin, and the resulting humanized immunoglobulin has binding specificity for a407 integrin. Preferably, all three 15 CDRs of a selected chain are substantially the same as the CDRs of the corresponding chain of a donor, and more preferably, all six CDRs of the light and heavy chains are substantially the same as the CDRs of the corresponding donor chains. In a preferred embodiment, the one or more CDRs of nonhuman origin have the amino acid sequences of the CDRs of murine Act-I Ab (SEQ ID NOS: 9-14). 20 The portion of the humanized immunoglobulin or immunoglobulin chain which is of human origin (the human portion) can be derived from any suitable human immunoglobulin or immunoglobulin chain. For example, a human constant region or portion thereof, if present, can be derived from the K or I light chains, and/or the y (e.g., yl, y2, y3, y4), ±, a (e.g., al, a2), 6 or E heavy chains of human 25 antibodies, including allelic variants. A particular constant region (e.g., IgG 1), variant or portions thereof can be selected in order to tailor effector function. For example, a mutated constant region (variant) can be incorporated into a fusion protein to minimize binding to Fc receptors and/or ability to fix complement (see e.g., Winter et al., GB 2,209,757 B; Morrison et al., WO 89/07142; Morgan et al., 30 WO 94/29351, December 22, 1994). LDP-02 contains a heavy chain constant region (human y1 heavy chain constant region) that was modified to reduce binding to -11human Fcy receptors. The LDP-02 Fc modifications are at positions 235 and 237 (i.e., Leu 2 -- Ala 3 and G1y"-Ala'). If present, framework regions of human origin (e.g., of the light chain variable region) are preferably derived from a human antibody variable region 5 having sequence similarity to the analogous region (e.g., light chain variable region) of the antigen binding region donor. Other sources of framework regions for portions of human origin of a humanized immunoglobulin include human variable consensus sequences (see e.g., Kettleborough, C.A. et al., Protein Engineering 4:773-783 (1991); Carter et al., WO 94/04679, published March 3, 1994)). For 10 example, the sequence of the antibody or variable region used to obtain the nonhuman portion can be compared to human sequences as described in Kabat, E.A., et aL., Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, U.S. Government Printing Office (1991). In a particularly preferred embodiment, the framework regions of a 15 humanized immunoglobulin chain are derived from a human variable region having at least about 65% overall sequence identity, and preferably at least about 70% overall sequence identity, with the variable region of the nonhuman donor antibody (e.g., mouse Act-i antibody). A human portion can also be derived from a human antibody having at least about 65% sequence identity, and preferably at least about 20 70% sequence identity, within the particular portion (e.g., FR) being used, when compared to the equivalent portion (e.g., FR) of the nonhuman donor. Amino acid sequence identity can be determined using a suitable sequence alignment algorithm, such as the Lasergene system (DNASTAR, Inc., Madison, WI), using the default parameters. 25 In one embodiment, the humanized immunoglobulin comprises at least one of the framework regions (FR) derived from one or more chains of an antibody of human origin. Thus, the FR can include a FR1 and/or FR2 and/or FR3 and/or FR4 derived from one or more antibodies of human origin. Preferably, the human portion of a selected humanized chain includes FR1, FR2, FR3 and FR4 derived 30 from a variable region of human origin (e.g., from a human immunoglobuli-n chain, from a human consensus sequence).
-12 The immunoglobulin portions of nonhuman and human origin for use in preparing humanized antibodies can have sequences identical to immunoglobulins or immunoglobulin portions -from which they are derived or to variants thereof Such variants include mutants differing by the addition, deletion, or substitution of one or 5 more residues. As indicated above, the CDRs which are of nonhuman origin are substantially the same as in the nonhuman donor, and preferably are identical to the CDRs of the nonhuman donor. Changes in the framework region, such as those which substitute a residue of the framework region of human origin with a residue from the corresponding position of the donor, can be made. One or more mutations 10 in the framework region can be made, including deletions, insertions and substitutions of one or more amino acids. For a selected humanized antibody or chain, suitable framework mutations can be designed. Preferably, the humanized immunoglobulins can bind a4p7 integrin with an affinity similar to or better than that of the nonhuman donor. Variants can be produced by a variety of suitable 15 methods, including mutagenesis of nonhuman donor or acceptor human chains. Immunoglobulins (e.g., human and/or humanized immunoglobulins) having binding specificity for human a4p7 integrin include immunoglobulins (including antigen-binding fragments) which can bind determinants (epitopes) of the cx4 chain (e.g., mAb HP1/2 (Pulido, et al., JBiol Chem 266:10241-10245 (1991), murine 20 MAb 21.6 and humanized MAb 21.6 (Bendig et al., U.S. Patent No. 5,840,299)) and/or the 07 chain of the c4p7 heterodimer. For example, in particular embodiments, the human or humanized immunoglobulin can specifically or selectively bind a determinant of the a4P7 complex, but not bind determinants (epitopes) on the a4 chain or the $7 chain. In one embodiment, the human or 25 humanized immunoglobulin can have binding specificity for a combinatorial epitope on the a4p7 heterodimer. Such an immunoglobulin can bind a4p7 and not bind a431, for example. Antibodies which have binding specificity for the a4p'7 complex include, murine Act-I antibody and a humanized Act-I referred to as LDP 02 (see, WO 98/06248 by LeukoSite, Inc., published February 19, 1998 and U.S. 30 Application No. 08/700,737, filed August 15, 1996, the entire teachings of which are both incorporated herein by reference). In a preferred embodiment, the humanized -13 immu'noglobulin has at least one function characteristic of murine Act-i antibody, such as binding function (e.g., having specificity for a407 integrin, having the same or similar epitopic specificity), and/or inhibitory function (e.g., the ability to inhibit a4p7-dependent adhesion in vitro and/or in vivo, such as the ability to inhibit a4p7 5 integrin binding to MAdCAM- 1 in vitro and/or in vivo, or the ability to inhibit the binding of a cell bearing a4p7 integrin to a ligand thereof (e.g., a cell bearing MAdCAM-1)). Thus, preferred humanized immunoglobulins can have the binding specificity of the murine Act-i antibody, the epitopic specificity of murine Act-i antibody (e.g., can compete with murine Act-1, a chimeric Act-1 antibody, or 10 humanized Act-1 (e.g., LDP-02) for binding to a407 (e.g., on a cell bearing a4p7 integrin)), and/or inhibitory function. A particularly preferred humanized Ab for administration in accordance with the method is LDP-02. The binding function of a human or humanized immunoglobulin having binding specificity for a4p7 integrin can be detected by standard immunological 15 methods, for example using assays which monitor formation of a complex between humanized immunoglobulin and a437 integrin (e.g., a membrane fraction comprising a4p7 integrin, on a cell bearing a4P7 integrin, such as a human lymphocyte (e.g., a lymphocyte of the CD4'a4" Pl'* subset), human lymphocyte cell line or recombinant host cell comprising nucleic acid encoding a4 and/or P 7 which 20 expresses a4p7 integrin). Binding and/or adhesion assays or other suitable methods can also be used in procedures for the identification and/or isolation of immunoglobulins (e.g., human and/or humanized immunoglobulins) (e.g., from a library) with the requisite specificity (e.g., an assay which monitors adhesion between a cell bearing an a4p7 integrin and a ligand thereof (e.g., a second cell 25 expressing MAdCAM, an immobilized MAdCAM fusion protein (e.g., MAdCAM Ig chimera)), or other suitable methods. The immunoglobulin portions of nonhuman and human origin for use in preparing humanized immunoglobulins include light chains, heavy chains and portions of light and heavy chains. These immunoglobulin portions can be obtained 30 or derived from immunoglobulins (e.g., by de novo synthesis of a portion), or nucleic acids encoding an inimunoglobulin or chain thereof having the desired -14 property (e.g., binds a407 integrin, sequence similarity) can be produced and expressed. Humanized immunoglobulins comprising the desired portions (e.g., antigen binding region, CDR, FR, constant region) of human and nonhuman origin can be produced using synthetic and/or recombinant nucleic acids to prepare genes 5 (e.g., cDNA) encoding the desired humanized chain. To prepare a portion of a chain, one or more stop codons can be introduced at the desired position. For example, nucleic acid (e.g., DNA) sequences coding for newly designed humanized variable regions can be constructed using PCR mutagenesis methods to alter existing DNA sequences (see e.g., Kamman, M., et al., Nucl. Acids Res. 17:5404 (1989)). 10 PCR primers coding for the new CDRs can be hybridized to a DNA template of a previously humanized variable region which is based on the same, or a very similar, human variable region (Sato, K., et al., Cancer Research 53:851-856 (1993)). If a similar DNA sequence is not available for use as a template, a nucleic acid comprising a sequence encoding a variable region sequence can be constructed from 15 synthetic oligonucleotides (see e.g., Kolbinger, F., Protein Engineering 8:971-980 (1993)). A sequence encoding a signal peptide can also be incorporated into the nucleic acid (e.g., on synthesis, upon insertion into a vector). If the natural signal peptide sequence is unavailable, a signal peptide sequence from another antibody can be used (see, e.g., Kettleborough, C.A., Protein Engineering 4:773-783 (1991)). 20 Using these methods, methods described herein or other suitable methods, variants can be readily produced. In one embodiment, cloned variable regions (e.g., of LDP 02) can be mutagenized, and sequences encoding variants with the desired specificity can be selected (e.g., from a phage library; see e.g., Krebber et al., U.S. 5,514,548; Hoogenboom et al., WO 93/06213, published April 1, 1993)). 25 Human and/or humanized immunoglobulins can be administered (e.g., to a human) for therapeutic and/or diagnostic purposes in accordance with the method of the invention. For example, an- effective amount of a human and/or humanized immunoglobulins having binding specificity for a407 integrin can be administered to a human to treat a disease associated with leukocyte infiltration of mucosal tissues 30 (e.g., inflammatory bowel disease, such as Crohn's disease or ulcerative colitis). Treatment includes therapeutic or prophylactic treatment (e.g., maintenance -15 therapy). According to the method, the disease can be prevented or delayed (e.g., delayed onset, prolonged remission or quiescence) or the severity of disease can be reduced in whole or in part. In one embodiment, no more than about 8 mg of immunoglobulin per kg 5 body weight is administered during a period of about 1 month. In additional embodiments, no more than about 7 or about 6 or about 5 or about 4 or about 3 or about 2 or about 1 mg of immunoglobulin per kg body weight is administered during a period of about I month. As used herein, the term "month" refers to a calendar month and encompasses periods of 28, 29, 30 and 31 days. When an antigen 10 binding fragment of a human or humanized immunoglobulin is to be administered, theamount which is administered during the period of about one month can be adjusted in accordance with the size of the fragment. For example, if the antigen binding fragment is about half the size of the intact antibody by weight (e.g., measured in kDa), the amount administered during a period of about 1 month can be 15 about 4 mg per kg body weight or less. The amount of-immunoglobulin or antigen binding fragment administered can be expressed as mg/kg body weight or using any other suitable units. For example, the amount of immunoglobulin or antigen binding fragment administered can be expressed as moles of antigen binding sites per kg body weight. The number of moles of antigen-binding sites is dependent 20 upon the size, quantity and valency of the immunoglobulin or fragment and can be readily determined. For example, IgG and F(ab')2 fragments thereof are divalent and a dose which comprises I nanomole of IgG or F(ab') 2 fragment comprises 2 nanomoles of antigen-binding sites. The size of an antibody or antigen-binding fragment can be determined using any suitable method (e.g., gel filtration). 25 The human or humanized antibody or antigen-binding fragment can be administered in a single dose or in an initial dose followed by one or more subsequent doses. When multiple doses are desired, the interval between doses and the amount of immunoglobulin or antigen-binding fragment can be adjusted to achieve the desired therapeutic and/or diagnostic effect. For example, each of the 30 doses to be administered can independently comprise up to about 8 mg immunoglobulin or fragment per kg body weight. When a dose comprises about 8 -16 mg immunoglobulin or fragment per kg body weight the minimum interval before a subsequent dose is administered is a period of about 1 month. Preferably, each dose independently comprises about 0.1 to about 8 mg or about 0.1 to about 5 mg immunoglobulin or fragment per kg body weight. More preferably, each dose 5 independently comprises about 0.1 to about 2.5 mg immunoglobulin or fragment per kg body weight. Most preferably, each dose independently comprises about 0.15, about 0.5, about 1.0, about 1.5 or about 2.0 mg immunoglobulin or fragment per kg body weight. The interval between any two doses (e.g., initial dose and first subsequent 10 dose, first subsequent dose and second subsequent dose) can independently vary from a few seconds or minutes to about 120 days or more. For example, the initial dose can be administered and a first subsequent dose can be administered about 1 day later. Thereafter, second and third subsequent doses can be administered at intervals of about I month. Generally the minimum interval between doses is a 15 period of at least about 1 day or at least about 7 days. In particular embodiments, the minimum interval between doses is a period of at least about 14 days, or at least about 21 days or at least about 1 month (e.g., 28, 29, 30, 31 days). In additional embodiments, the interval between doses can be at least about 40, about 50, about 60, about 70, about 80, about 90, about 100, about 110 or about 120 days. 20 The amount of human or humanized immunoglobulin or antigen-binding fragments thereof administered in each dose can be an amount which is sufficient to produce a desired pharmacokinetic or pharmacodynamic effect. A variety of pharmacokinetic and pharmacodynamic parameters of human and/or humanized immunoglobulins or antigen-binding fragments thereof can be measured using 25 suitable methods. For instance, pharmacodynaiic parameters of antibodies and antigen-binding fragments (e.g., antigen saturation, antibody-induced inhibition of expression of antigen) can be measured using a suitable immunoassay. For example, as described herein, a4P7 signal (i.e., binding of labeled antibody to a4p7) following administration of LDP-02 was measured by flow cytometry. The results 30 of the assay revealed that administration of LDP-02 can result in saturation of a4S7 and/or inhibition of expression of a4p7 on the surface of circulating lymphocytes.
17 Accordingly, each dose to be administered can comprise an amount of immunoglobulin or fragment which is sufficient to achieve a) about 50% or greater saturation of a4p7 integrin binding sites on circulating lymphocytes (e.g., CD8* cells) and/or b) about 50% or greater inhibition of a4p7 integrin expression on the 5 cell surface of circulating lymphocytes for a period of at least about 10 days following administration of the dose. In other embodiments, each dose can comprise an amount of immunoglobulin or fragment which is sufficient to achieve and maintain a) about 60% or greater, about 70% or greater, about 80% or greater or about 85% or greater saturation of a4D7 integrin binding sites on circulating 10 lymphocytes and/or b) about 60% or greater, about 70% or greater, about 80% or greater or about 85% or greater inhibition of a407 integrin expression on the cell surface of circulating lymphocytes for a period of at least about 10 days following administration of the dose. In other particular embodiments, each dose can comprise an amount of 15 immunoglobulin or fragment which is sufficient to achieve a desired degree of saturation of a407 integrin binding sites on circulating lymphocytes (e.g., CD8+ cells) and/or inhibit expression of a437 integrin on the cell surface of circulating lymphocytes to the desired degree for a period of at least about 14 days, at least about 20 days, at least about 25 days or at least about one month following 20 administration of the dose. In additional embodiments, each dose can comprise an amount of immunoglobulin or fragment which is sufficient to achieve a desired degree of saturation of a4P7 integrin binding sites on circulating lymphocytes (e.g., CD8+ cells) and/or inhibit expression of u407 integrin on the cell surface of circulating lymphocytes to the desired degree for a period of at least about 40, about 25 50, about 60, about 70, about 80, about 90, about 100, about 110 or about 120 days. Suitable assays for determining the dose of antibody required to achieve a desired serum concentration or to saturate and/or inhibit expression of a target antigen can be readily designed. For example, a flow cytometry based assay can be used to measure a4p7 expression on the surface of cells isolated from a subject 30 following administration of an immunoglobulin (e.g., human, humanized) which binds to a4P7. In one embodiment, a murine antibody which binds human a4p7 -18 can be used. Preferably the murine antibody can bind to an epitope on a4p7 which is distinct from the epitope bound by the human or humanized immunoglobulin and the binding of the murine antibody to a437 is not inhibited (e.g., blocked) by the prior binding of the humanized immunoglobulin. Murine antibodies or other 5 antibodies with these properties can be prepared and selected using the methods described herein or other suitable methods. The level of a4p7 expression on circulating lymphocytes (e.g., CD8+ cells) isolated from a human can be measured or determined using each of the antibodies (i.e., immunoglobulin to be administered, murine antibody) by flow cytometry or other suitable methods. Then, the human or 10 humanized antibody to be administered can be administered to the human, peripheral blood can be drawn at predetermined times following the administration, and lymphocytes can be isolated (e.g., by density gradient centrifugation) for analysis. The peripheral blood lymphocytes (e.g., CD8+ cells) can be stained with each of the antibodies and the amount of x4P7 detected by each antibody can be measured or 15- detected by flow cytometry or other suitable methods. A decrease in the amount of x4p7 integrin measured or determined using the human or humanized immunoglobulin is indicative of a) persistent integrin occupancy by the administered immunoglobulin (e.g., antigen saturation) and/or b) inhibition of c4p7 expression on the surface of the lymphocytes (e.g., down modulation of a4p7, shedding of o4p7). 20 A decrease in the amount of a4p7 integrin measured or detected using the human or humanized immunoglobulin together with no change in the amount of a4p 7 integrin measured or determined using the murine antibody is indicative of persistent occupancy of a4P7 (e.g., saturation) by the human or humanized immunoglobulin administered. A decrease in the amount of a4p7 integrin measured or detected 25 using the human or humanized immunoglobulin together with a decrease in the amount of a4p7 integrin measured or detected using the murine antibody is indicative of inhibition of ax4p7 expression on the surface of circulating lymphocytes. Pharmacokinetic parameters, such as the serum concentration of antibody 30 over time following administration of said antibody can be measured using an -19 immunoassay such as an ELISA or cell-based assay. For example, as described herein, the serum concentration of a humanized anti-cx4p7 immunoglobulin
(LDP
02) at predetermined time points following a single administration of antibody (LDP-02) was measured using a cell-based assay. The results of the assay revealed 5 that the serum concentration of LDP-02 can remain elevated (e.g., at or above I pg/mL) for a period of about 10 days or more following administration of the humanized antibody. The prolonged presence of LDP-02 in the serum can be indicative of superior efficacy as a result of persistent inhibition of a4p7 function, for example persistent inhibition of a4p7 mediated adhesion of leukocytes to 10 MAdCAM. Accordingly, each dose to be administered can comprise an amount of immunoglobulin or fragment which is sufficient to achieve and maintain a serum concentration of at least about 1 ig/mL for a period of at least about 10 days following administration of the dose. In particular embodiments, each dose can 15 _comprise an amount of immunoglobulin or fragment which is sufficient to achieve and maintain a serum concentration of at least about 1 Vtg/mL for a period of at least about 14 days, at least about 20 days, at least about 25 days or at least about one month following administration of the dose. In additional embodiments, each dose can comprise amount of immunoglobulin or fragment which is sufficient to achieve 20 and maintain a serum concentration of at least about 1 4g/mL for a period of at least about 40, about 50, about 60, about 70, about 80, about 90, about 100, about 110 or about 120 days. As discussed herein, antigen-binding fragments of a human or humanized immunoglobulin can be substantially smaller and, therefore, bind more antigen 25 (a4p7) per unit of protein (4g) than intact or native immunoglobulin. Accordingly, the serum concentration of an antigen-binding fragment of a human or humanized immunoglobulin which can be indicative of superior efficacy can be lower than 1 ig/mL. Thus, when administration of an antigen-binding fragment of a human or humanized immunoglobulin is desired, the dose can comprise an amount of antigen 30 binding fragment which is sufficient to achieve a serum concentration which is proportionate to 1 pg/mL for an intact immunoglobulin. For example, if the antigen- -20 binding fragment is about half the size of the intact antibody by weight (e.g., measured in kDa), the dose can comprise an amount sufficient to achieve and maintain a serum concentration of about 0.5 tg/mL for a period of at least about 10 days. The desired serum concentration of immunoglobulin or antigen-binding 5 fragment can be expressed as ig/mL or using any other suitable units. For example, the amount of immunoglobulin or antigen-binding fragment administered can be expressed as moles of antigen binding sites per volume of serum (e.g., M). Human and humanized immunoglobulins can be administered in accordance with the present invention for in vivo diagnostic applications or to modulate a4p7 10 integrin function in therapeutic (including prophylactic) applications. For example, human and humanized immunoglobulins can be used to detect and/or measure the level of an a4P7 integrin in a subject. For example, a humanized immunoglobulin having binding specificity for a4p7 integrin can be administered to a human and antibody-a407 integrin complexes which are formed can be detected using suitable 15 methods. For example, the humanized antibody can be labeled with, for example, radionuclides (121LI, .In, technetium-99m), an epitope label (tag), an affinity label (e.g., biotin, avidin), a spin label, an enzyme, a fluorescent group or a chemiluninescent group and suitable detection methods can be used. In an application of the method, humanized immunoglobulins can be used to analyze 20 normal versus inflamed tissues (e.g., from a human) for a407 integrin reactivity and/or expression (e.g. radiologically) or to detect associations between IBD or other conditions and increased expression of a407 (e.g., in affected tissues). The immunoglobulins described herein can be administered in accordance with the method of the invention for assessment of the presence of a4A7 integrin in normal 25 versus inflamed tissues, through which the presence of disease, disease progress and/or the efficacy of anti-a407 integrin therapy in inflammatory disease can be assessed. Human and humanized immunoglobulins (including antigen-binding fragments) can be administered to an individual to modulate (e.g., inhibit (reduce or 30 prevent)) binding function and/or leukocyte (e.g., lymphocyte, monocyte) infiltration function of a4P7 integrin. For example, human and humanized immunoglobulins which inhibit the binding of a4p7 integrin to a ligand (i.e., one or more ligands) can be administered according to the method for the treatment of diseases associated -21 with leukocyte (e.g., lymphocyte, monocyte) infiltration of tissues (including recruitment and/or accumulation of leukocytes in tissues), particularly of tissues which express the molecule MAdCAM. An effective amount of a human immunoglobulin or antigen-binding fragment thereof, or humanized 5 immunoglobulin or antigen-binding fragment thereof (i.e., one or more immunoglobulins or fragments) is administered to an individual (e.g., a mammal, such as a human or other primate) in order to treat such a disease. For example, inflammatory diseases, including diseases which are associated with leukocyte infiltration of the gastrointestinal tract (including gut-associated endothelium), other 10 mucosal tissues, or tissues expressing the molecule MAdCAM-I (e.g., gut-associated tissues, -such as venules of the lamina propria of the small and large intestine; and mammary gland (e.g., lactating mammary gland)), can be treated according to the present method. Similarly, an individual having a disease associated with leukocyte infiltration of tissues as a result of binding of leukocytes to 15 cells (e.g., endothelial cells) expressing MAdCAM- 1 can be treated according to the present invention. In a particularly preferred embodiment, diseases which can be treated accordingly include inflammatory bowel disease (IBD), such as ulcerative colitis, Crohn's disease, ileitis, Celiac disease, nontropical Sprue, enteropathy associated 20 with seronegative arthropathies, microscopic or collagenous colitis, eosinophilic gastroenteritis, or pouchitis resulting after proctocolectomy, and ileoanal anastomosis. Pancreatitis and insulin-dependent diabetes mellitus are other diseases which can be treated using the present method. It has been reported that MAdCAM-1 is 25 expressed by some vessels in the exocrine pancreas from NOD (nonobese diabetic) mice, as well as from BALB/c and SJL mice. Expression of MAdCAM-1 was reportedly induced on endothelium in inflamed islets of the pancreas of the NOD mouse, and MAdCAM- 1 was the predominant addressin expressed by NOD islet endothelium at early stages of insulitis (Hanninen, A., et al., J. Clin. Invest., 92: 30 2509-2515 (1993)). Further, accumulation of lymphocytes expressing a4p7 within islets was observed, and MAdCAM-1 was implicated in the binding of lymphoma -22 cells via a4P7 to vessels from inflamed islets (Hanninen. A., et al., J. Clin. Invest., 92: 2509-2515 (1993)). Examples of inflammatory diseases associated with mucosal tissues which can be treated according to the present method include mastitis (mammary gland), 5 cholecystitis, cholangitis or pericholangitis (bile duct and surrounding tissue of the liver), chronic bronchitis, chronic sinusitis, asthma, and graft versus host disease (e.g., in the gastrointestinal tract). As seen in Crohn's disease, inflammation often extends beyond the mucosal surface, accordingly chronic inflammatory diseases of the lung which result in interstitial fibrosis, such as hypersensitivity pneumonitis, 10 collagen diseases, sarcoidosis, and other idiopathic conditions can be amenable to treatment. Treatment can be curative, induce remission or quiescence or prevent relapse or recurrence of active disease. According to the method, treatment can be episodic or chronic (e.g., chronic treatment of active disease, to maintain quiescent disease, to 15 induce quiescence and maintain quiescence), for example. In a particularly preferred embodiment, a human or humanized immunoglobulin having binding specificity for a4@7 integrin is administered to a human having inflammatory bowel disease, such as ulcerative colitis or Crohn's disease. The immunoglobulin can be administered to treat active disease and/or to 20 maintain quiescence (i.e., inhibit relapse or recurrence). In a particular embodiment, the human or humanized immunoglobulin can be administered to maintain quiescence of inflammatory bowel disease which has been induced by treatment with one or more other agents (e.g., steroids (prednisone, prednisolone, adrenocorticotrophic hormone (ACTH)), cyclosporin A, FK506, antibody having 25 binding specificity for TNFcx (infliximab, CDP571), azathioprene, 6-mercaptopurine, 5-aminosalicylic acid (5-ASA) or compounds containing 5-ASA (e.g., sulfsalazine, olsalazine, balsalazide), antibiotics (e.g., metronidazole), interleukins (IL-10, IL-1 1), nicotine, heparin, thalidomide, lidocane) or surgery (e.g., intestinal resection). 30 The human immunoglobulin or antigen-binding fragment thereof, or humanized immunoglobulin or antigen-binding fragment thereof is administered in -23 an effective amount. For therapy, an effective amount is an amount sufficient to achieve the desired therapeutic (including prophylactic) effect (such as, an amount sufficient to reduce or prevent a4p7 integrin-mediated binding to a ligand thereof and/or signalling, thereby inhibiting leukocyte adhesion and infiltration and/or 5 associated cellular responses; an amount sufficient to induce remission or prevent relapse or recurrence of disease). The human immunoglobulin or antigen-binding fragment thereof, or humanized immunoglobulin or antigen-binding fragment thereof can be administered in a single dose or in an initial dose followed by one or more subsequent doses as described herein. The amount of immunoglobulin or 10 antigen-binding fragment administered in a particular dose as well as the interval between doses can depend on the characteristics of the individual, such as general health, age, sex, body weight and tolerance to drugs as well as the type and severity of disease. The skilled artisan will be able to determine appropriate dosages depending on these and other factors. 15 According to the method, the human or humanized immunoglobulin can be administered to an individual (e.g., a human) alone or in conjunction with another agent (i.e., one or more additional agents). A human or humanized immunoglobulin can be administered before, along with or subsequent to administration of the additional agent. In one embodiment, more than one human or humanized 20 immunoglobulin which inhibits the binding of a4p7 integrin to its ligands is administered. In another embodiment, an antibody (e.g, human antibody, humanized antibody), such as an anti-MAdCAM-1, anti-VCAM-1, or anti-ICAM-1 antibody, which inhibits the binding of leukocytes to an endothelial ligand is administered in addition to a human or humanized immunoglobulin which binds a4P7 integrin. In 25 yet another embodiment, an additional pharmacologically active ingredient (e.g., an antiinflammatory compound, such as 5-aminosalicylic acid (5-ASA) or compounds containing 5-ASA (e.g., sulfsalazine, olsalazine, balsalazide), another non-steroidal antiinflammatory compound, or a steroidal antiinflammatory compound (e.g., prednisone, prednisolone, adrenocorticotrophic hormone (ACTH)), 30 immunosuppressive agents (azathioprene, 6-mercaptopurine, cyclosporin A, FK506), immunomodulators (e.g., antibody having binding specificity for TNFa -24 (infliximab, CDP571), thalidomide, interleukins (e.g., recombinant human IL-10, recombinant human IL-11)), antibiotics (e.g., metronidazole), nicotine, heparin, lidocaine) can be administered in conjunction with a humanized immunoglobulin of the present invention. 5 A variety of routes of administration are possible, including, but not necessarily limited to, parenteral (e.g., intravenous, intraarterial, intramuscular, intrathecal, subcutaneous injection), oral (e.g., dietary), topical, inhalation (e.g., intrabronchial, intranasal or oral inhalation, intranasal drops), or rectal, depending on the disease or condition to be treated. Parenteral administration, particularly 10 intravenous injection and subcutaneous injection, is preferred. The human immunoglobulin or antigen-binding fragment thereof and/or the humanized immunoglobulin or antigen-binding fragment thereof can be administered to the individual as part of a pharmaceutical or physiological composition for the treatment of a disease associated with leukocyte infiltration of 15 mucosal tissues (e.g., inflammatory bowel disease (e.g., ulcerative colitis, Crohn's disease). Such a composition can comprise an immunoglobulin or antigen-binding fragment having binding specificity for a4p7 integrin as described herein, and a pharmaceutically or physiologically acceptable carrier. Pharmaceutical or physiological compositions for co-therapy can comprise an immunoglobulin or 20 antigen-binding fragment having binding specificity for cx407 integrin and one or more additional therapeutic agents. An immunoglobulin or antigen-binding fragment having binding specificity for a4P7 integrin function and an additional therapeutic agent can be components of separate compositions which can be mixed together prior to administration or administered separately. Formulation will vary 25 according to the route of administration selected (e.g., solution, emulsion, capsule). Suitable -carriers can contain inert ingredients which do not interact with the immunoglobulin or antigen-binding fragment and/or additional therapeutic agent. Standard pharmaceutical formulation techniques can be employed, such as those described in Remington's Pharmaceutical Sciences, Mack Publishing Company, 30 Easton, PA. Suitable carriers for parenteral administration include, for example, sterile water, physiological saline, bacteriostatic saline (saline containing about 0.9% -25 benzyl alcohol), phosphate-buffered saline, Hank's solution, Ringer's-lactate and the like. Methods for encapsulating compositions (such as in a coating of hard gelatin or cyclodextran) are known in the art (Baker, et al., "Controlled Release of Biological Active Agents", John Wiley and Sons, 1986). For inhalation, the agent 5 can be solubilized and loaded into a suitable dispenser for administration (e.g., an atomizer, nebulizer or pressurized aerosol dispenser). The present invention will now be illustrated by the following Examples, which are not intended to be limiting in any way. 10 EXAMPLES Introduction LDP-02 is a humanized IgG1 monoclonal antibody that binds a4p7 integrin, a cell surface glycoprotein present on the surface of most T and B lymphocytes. a407 mediates lymphocyte trafficking to gastrointestinal mucosa and gut-associated 15 lympioid tissue through adhesion interaction with the homing receptor MAdCAM 1. By blocking a4p7-MAdCAM-1 interactions, LDP-02 can inhibit the recruitment of leukocytes from the vasculature to the gastrointestinal mucosa, thus having a beneficial effect on the inflammatory activity in patients afflicted with inflammatory bowel disease (IBD) such as ulcerative colitis and Crohn's Disease. 20 This section presents information from the two LDP-02 clinical trials that have been completed. These trials include one completed Phase I study conducted in healthy subjects (Study L297-007) and one completed Phase Ib/Ila trials in patients with ulcerative colitis (UC)(Study L297-006). Table I describes each of the studies.
-26 Table I Study No. Dosing Regimen, Number of # Sites Study Status Study Design/ Population Dose, Route Subjects Country ._Enrolled 5 L297-007 Completed Phase I, randomized, double- Day 1 (single dose) Total= 19 blind, placebo-controlled, 0.15 mg/kg IV LDP-02= 14 UK ascending single dose study. 0.15 mg/kg SC Placebo= 5 0.5 mg/kg IV Healthy Male Subjects 1.5 mg/kg IV 18-50 years of age 2.5 mg/kg IV L297-006 Completed Phase Ib/Ila, randomized, Day 1 (single dose) Total= 29 5 double-blind, placebo- 0.15 mg/kg SC LDP-02= 21 10 Canada controlled, single rising 0.15 mg/kg IV Placebo= 8 dose, multicenter study. 0.5 mg/kg IV 2.0 mg/kg IV Patients with moderately placebo IV severe ulcerative colitis. Prior steroid use was limited (<20mg/day). Use of 5 ASAs was allowed. Example 1: Study L297-007 Study L297-007 entitled, "A Placebo-Controlled, Double-Blind, Rising Dose Study Investigating the Tolerability, Pharmacodynamics and Pharmacokinetics of LDP-02 Given by the Subcutaneous and Intravenous Routes in Healthy Male 15 Volunteers" has been completed and final results are presented in this section. Study Design Study L297-007 was a randomized, double-blind, placebo-controlled, ascending single-dose study in healthy male volunteers. Healthy male volunteers 18 to 50 years of age meeting all inclusion/exclusion criteria were enrolled in the study 20 sequentially by study group and, within each study group, were randomly assigned to receive LDP-02 or placebo (i.e., isotonic sodium citrate buffer). To minimize risk to subjects, safety and tolerability were reviewed at each dose level prior to escalating to the next dose level. The treatment groups and numbers of subjects planned for the study are shown in Table 2.
-27 Table 2 Study L297-007: Study Groups Group Route of LDP-02 Placebo Administration # subjects # subjects * Dose 1 IV 3 0.15 mg/kg 1 SC 3 0.15 mg/kg 1 2 IV 3 0.5 mg/kg 1 5 3 IV . 3 . 15 mg/kg 1 4 IV 3 2.5 mg/kg 1 *SC= subcutaneous administration; IV = intravenous administration On study Day 1, LDP-02 or placebo was administered either SC into the thigh (Group 1 SC dosing only) or via a 30 minute constant rate IV infusion (Groups 10 1-4). Safety assessments included recording of adverse events, physical examinations, vital signs, clinical laboratories (i.e., hematology, blood chemistries, and urinalysis), plasma cytokine levels, and 12-lead electrocardiograms (ECGs). In addition, since this was the first clinical trial of LDP-02, continuous cardiac monitoring was carried out pre-dose through 4 hours post-dose. Blood samples were 15 obtained to assess anti-antibody response to LDP-02, cytokine levels, serum LDP-02 concentration (pharmacolinetics), and saturation and binding site occupation of a4P7 receptors and lymphocyte subsets (pharmacodynamics). Study assessments were conducted at specified times through 36 days post-treatment. Following the results of the Day 36 pharmacokinetic and pharnacodynamic (immunological) 20 analyses, the protocol was amended to allow additional blood draws for subjects who received LDP-02. These blood draws were used to follow LDP-02 serum levels until they became non-quantifiable (i.e., below the limit of quantification [BLQ]) and to ensure that a4p7 saturation and memory cell populations had returned to baseline (pre-dose) levels. This amendment was particularly important in the higher 25 dose groups where the characteristics of terminal phase kinetics were not well established by Day 36.
-28 Study Results Pharmacokinetics The assay of LDP-02 in serum was performed using a validated cell-based assay. Standards and samples were incubated with a target cell line (HUT-78) which 5 expresses the a4P7 antigen. After washing, a fluorescently labeled polyclonal anti human IgG1 was added. Fluorescence intensity was measured by flow cytometry and compared with the fluorescence intensity of LDP-02 standards. The effective serum concentration of LDP-02 was then defined by comparison of the sample with a standard curve generated with known concentrations of LDP-02. 10 Blood samples for determination of LDP-02 serum concentration were collected pre-dose, 1, 1.5, 3, 8, 12 and 24 hours after dosing, and on Days 3, 5, 7, 8, 15, 22, and 36. When it became known that LDP-02 was still detectable at Day 36, blood draws for subjects who received LDP-02 continued until levels had fallen to below the limits of quantitation of the assay. Thirteen of the 14 subjects who 15 received LDP-02 returned for follow-up blood draws up to a maximum of 226 days post-dose. LDP-02 concentrations over time by individual patient and mean pharmacokinetic parameters by LDP-02 dose group are presented in the Appendix to Study L297-007. Mean LDP-02 serum concentrations over time are plotted out to 20 the last blood draw for all treatment groups in FIG. 6.
-29 Table 3 Study L297-007: Mean Pharmacokinetic Parameters of LDP-02 in Healthy Subjects' Pharmacokinetic Dose and Route of Administration of LDP-02 (number of subjects) Parameter 0.15 mg/kg SC 0.15 mg/kg 0.5 mg/kg 1.5 mg/kg 2.5 mg/kg (n=3) IV IV IV IV (n=3) (n=3) (n=3) (n=2) 5 C". (g/mL) 1.112 7.648 15.760 118.813 101.749 (0.519) (3.201) (7.476) (14.544) (5.117) (days) 6.01 0.13 0.5 0.13 0.05 (median & (4.01 - 6.01) (0.04 - 0.33) (0.06 - (0.06- (0.04 range) 0.5) 0.33) 0.06) Tu2 (days) 4.33 4.39 4.02 14.9 17.1 (2.23) (1.51) (0.71) (10.3) (8.91) 10 AUC, 10.4 19.5 83.6 .660 1651 (4g-day/mL) (4.40) (5.00) (18.3) (229) (229) (1/day) 0.1852 0.1731 0.1763 0.0994 0.0469 (0.0735) (0.0673) (0.0344) (0.1145) (0.0244) AUC 11.4 20.3 85.1 755 1747 (4g-day/mL) (5.80) (5.88) (18.2) (308) (95.8) 15 AUC Extrapolated 5.9 3.4 1.8 9.5 5.7 % (3.2) (1.4) (16.1) (8.0) CL* 15.3 7.75 6.06 2.31 1.43 (miLday/kg) (6.26) (1.93) (1.32) (11) (0.08) V.* 82.5 46.6 34.3 54.0 35.9 20 (mlJkg) (6.88) (10.1) (2.84) (51.4) (20.3)_ 'All values are mean +/- SD unless otherwise indicated. The SD appears in parenthesis. *Clearance and volume terms for the SC dose group are the apparent clearance (CL/F) and apparent volume (V/F). 25 Values were obtained for the mean single dose IV pharmacokinetic parameters for the 4 dose groups (C..., t,,2 and AUC). Follow-up samples (i.e., those taken beyond Day 36), where the focus was on safety, allowed some further characterization of the concentration-time profiles. The difference in the t,/, values between the 2 lower dose groups (0.15 and 0.5 mg/kg) and the higher dose groups 30 (1.5 and 2.5 mg/kg) of around 10 days could be explained in that the "true" terminal phase for the higher dose groups had not been characterized. The non compartmental pharmacokinetics of the lower doses of LDP-02 (0.15 and 0.5 mg/kg) were well characterized and non-linear pharmacokinetics became evident as the dose was increased up to 2.5 mg/kg.
-30 Assessment of the Pharmacodynamic Effect of LDP-02 Fluorescece-activated cell sorting (FACS) analysis was used to measure the presence of a4p7 sites on peripheral blood lymphocytes pre- and post-LDP-02 administration. To detect a4p7 that were recognized by antibody, biotin labeled 5 ACT-1, the murine homologue of LDP-02, was added to samples of patient blood and detected using PE-streptavidin. The standardized mean equivalent soluble fluorescence (MESF) is proportional to the number of detectable a407 sites. Serum a4P7 binding over time (MESF values and percentage of baseline at each post-dose time point) are presented by individual subject and by treatment 10 group in the Appendix to Study L297-007. As measured by FACS analysis, mean saturation of a4P7 integrin on lymphocytes over time (i.e., to Day 36) for each treatment is presented in FIG. 7. As seen in FIG. 7, there was no detection of free a407 binding sites on lymphocytes for at least two weeks following administration of all LDP-02 doses. Between about 15 day 7 and day 22, a4p7 signal started to return to baseline for the 0.15 mg/kg IV dose group and for the 0.15 mg/kg SC dose group. Between day 22 and day 36, x437 signal started to return to baseline for the 0.5 mg/kg IV dose group. At the higher doses of LDP-02 studied (1.5, and 2.5 mg/kg) loss of a4p7 signal persisted for longer than 36 days following single IV doses. For the 2.5 mg/kg dose group, 20 a4p7 binding saturation continued up to Day 70 (see, data in Appendix to Study L297-007). Follow-up blood sampling up to about Study Day 200 was done to confirm that free a437 binding sites on lymphocytes has returned to baseline (pre-dose) levels. The initial reappearance of free a407 sites appeared to occur when LDP-02 25 blood concentrations became non-detectable. Conclusions The administration of LDP-02 at IV doses of 0.15, 0.50, 1.50, and 2.5 mg/kg and a SC dose of 0.15 mg/kg to healthy male subjects was well-tolerated. Following administration of all LDP-02 doses there was no detection of free 30 a4p7 binding sites on lymphocytes for approximately two weeks post-dose.
-31 Saturation of a4p7 binding sites continued for up to approximately 2 weeks post dosing for the 0.15 mg/kg IV group and for up to approximately 3 weeks post-dosing for the 0.15 mg/kg SC and 0.5 mg/kg IV groups. Duration of effect persisted for a month or longer with the 1.5 mg/kg TV dose and continued to approximately Day 70 5 with 2.5 mg/kg LDP-02 IV. Follow-up samples obtained after Day 36 demonstrated that expression of free a407 binding sites had returned to baseline (pre-dose levels). No anti-idiotype antibodies were raised to LDP-02 indicating that it did not initiate a humoral immunogenic response. The non-compartmental pharmacokinetics of the lower doses of LDP-02 (0.15 and 0.5 mg/kg) became evident as the dose was 10 increased up to 2.5 mg/kg. APPENDIX TO STUDY L297-007 LDP-02 Serum Concentration Over Time by Subject by Treatment Group. Data from individual patients are presented in Tables 4-9.
-32 C- q- c7 00 C 0 -4' 0 0 p QC>c>C 0 0> 0 0 C:' 0 0 0o C; 6 t,: ooo %r; C 0 6 fnD0 v00 ~ 0 0 ~ o o o 00 rcr 000 O o 0 0 =00 r,~o 'I00 q V Ll 0000 C 0 0 o C>o 0 0 M~ VC)C 0 C(0O r'0 Q 0C 00 V~~ V~r~000 0000 m 00 -- 4 'r N 6 0 00004o00 g0en VU 00 0\0q\ r- V 0 jo oo oo 000 c) D ; C - t- co V-, 00 CO F -33 Table 5 0.15 mg/kg LDP-02 SC Subject # 5 Subject # 6 Subject # 8 Mean Time Time Time Time Time Time p )/mL (hr) (day) (hr) (day) (hr) (day) (n=3) 5 Pre-Dose Pre-Dose 0.01 Pre-Dose Pre-Dose 0.01 Pre-Dose Pre-Dose 0.01 0.01 1.0 0.042 0.01 1.0 0.042 0.01 1.0 0.042 0.01 0.01 1.5 0.063 0.01 1.5 0.063 0.01 1.5 0.063 0.01 0.01 3.0 0.125 0.01 3.0 0.125 0.01 3.0 0.125 0.01 0.01 8.0 0.333 0.06 8.0 0.333 0.09 8.0 0.333 0.09 0.08 10 12.0 0.500 0.11 12.0 0.500 0.12 12.0 0.500 0.10 0.11 24.0 1.000 0.12 24.0 1.000 0.30 24.0 1.000 0.55 0.32 72.0 3.000 0.23 72.0 3.000 0.81 72.0 3.000 0.91 0.65 120.0 5.000 0.54 120.0 5.000 0.93 120.0 5.000 1.13 0.86 168.0 7.000 0.71 168.0 7.000 0.88 168.0 7.000 1.70 1.10 15 192.0 8.000 0.62 192.0 8.000 0.81 192.0 8.000 1.05 0.83 360.0 15.000 0.28 360.0 15.000 0.08 360.0 15.000 0.53 0.30 528.0 22.000 0.02 528.0 22.000 0.03 528.0 22.000 0.26 0.11 864.0 36.000 0.04 864.0 36.000 0.04 864.0 36.000 0.01 0.03 3912.0 163.000 0.01 3912.0 163.000 0.01 3912.0 163.000 0.01 0.01 20 5088.0 212.000 0.01 5088.0 212.000 0.01 5088.0 212.000 0.01 0.01 Table 6 0.5 mg/kg LDP-02 IV Subject # 9 Subject # 10 Subject # 12 Mean Time Time Time Time Time Time Ig/mL I.g/mL ±g/mL pg/miL (hr) (day) (hr) (day) (hr) (day) (n=3) 25 Pre-Dose Pre-Dose 0.01 Pre-Dose Pre-Dose 0.01 Pre-Dose Pre-Dose 0.01 0.01 1.0 0.042 9.06 1.0 0.042 10.74 1.0 0.042 10.93 10.24 1.5 0.063 24.39 1.5 0.063 6.62 1.5 0.063 8.17 13.06 3.0 0.125 16.37 3.0 0.125 10.14 3.0 0.125 9.94 12.15 8.0 0.333 15.04 8.0 0.333 9.30 8.0 0.333 9.35 11.23 30 12.0 0.500 10.64 12.0 0.500 11.70 12.0 0.500 11.19 11.18 24.0 1.000 9.17 24.0 1.000 9.00 24.0 1.000 8.52 8.90 72.0 3.000 5.34 72.0 3.000 7.55 72.0 3.000 7.60 6.83 120.0 5.000 10.25 120.0 5.000 2.43 120.0 5.000 8.58 7.09 168.0 7.000 5.74 168.0 7.000 6.59 168.0 7.000 4.93 5.75 35 192.0 8.000 3.79 192.0 8.000 2.48 192.0 8.000 4.32 3.53 360.0 15.000 1.70 360.0 15.000 2.21 360.0 15.000 2.49 2.13 528.0 22.000 0.41 528.0 22.000 0.12 528.0 22.000 1.65 0.73 864.0 36.000 0.01 864.0 36.000 0.01 864.0 36.000 0.1 1 0.04 3576.0 149.00 0.01 3912.0 163.000 0.01 3576.0 149.000 0.01 0.01 5424.0 226.000 0.01 0.01 -34 Table 7 1.5 mg/kg LDP-02 TV Subject # 13 Subject # 15 Subject# 16 Mean Time Time Time Time Time Time p±pg/mL (hr) (day) (hr) (day) (hr) (day) (n=3) 5 Pre-Dose Pre-Dose 0.01 Pre-Dose Pre-Dose 0.01 Pre-Dose Pre-Dose 0.01 0.01 1.0 0.042 87.62 1.0 0.042 58.06 1.0 0.042 103.10 82.93 1.5 0.063 63.67 1.5 0.063 134.97 1.5 0.063 86.05 94.90 3.0 0.125 92.78 3.0 0.125 63.78 3.0 0.125 106.78 87.78 8.0 0.333 114.69 8.0 0.333 64.12 8.0 0.333 84.42 87.74 10 12.0 0.500 73.02 12.0 0.500 43.76 12.0 0.500 44.09 53.62 24.0 1.000 99.61 24.0 1.000 77.77 24.0 1.000 71.80 83.06 72.0 3.000 102.88 72.0 3.000 38.82 72.0 3.000 67.61 69.77 120.0 5.000 42.46 120.0 5.000 25.26 120.0 5.000 23.95 30.56 168.0 7.000 26.10 168.0 7.000 18.42 168.0 7.000 23.85 22.79 15 192.0 8.000 46.47 192.0 8.000 11.90 192.0 8.000 19.85 26.07 360.0 15.000 19.83 360.0 15.000 5.80 360.0 15.000 19.54 15.06 528.0 22.000 10.93 528.0 . 22.000 0.11 528.0 22.000 13.89 8.31 864.0 36.000 0.19 864.0 36.000 0.69 864.0 36.000 9.49 3.46 1968.0 82.000 0.48 - 1968.0 163.000 0.30 0.39 20 3264.0 136.000 0.01 3264.0 212.000 0.03 0.02 4272.0 178.000 0.01 3960.0 165.000 0.01 0.01 4824.0 201.000 0.01 0.01 -35 Table 8 2.5 mg/kg LDP-02 IV Subject# 18 Subject # 19 Mean Time (br) Time Time (hr) Time (day) pg/mL (day) (n=2) 5 Pre-Dose Pre-Dose 0.01 Pre-Dose Pre-Dose 0.01 0.01 1.0 0.042 105.37 1.0 0.042 84.06 94.72 1.5 0.063 71.27 1.5 0.063 98.13 84.70 3.0 0.125 73.49 3.0 0.125 81.59 77.54 8.0 0.333 84.00 8.0 0.333 80.17 82.09 10 12.0 0.500 103.81 12.0 0.500 85.53 94.67 24.0 1.000 68.79 24.0 1.000 85.52 77.15 72.0 3.000 63.30 72.0 3.000 69.49 66.40 120.0 5.000 53.33 120.0 5.000 59.11 56.22 168.0 7.000 50.72 168.0 7.000 54.63 52.67 15 192.0 8.000 43.47 192.0 8.000 67.32 55.40 360.0 15.000 22.82 360.0 15.000 23.85 23.34 528.0 22.000 22.45 528.0 22.000 21.92 22.19 864.0 36.000 17.42 864.0 36.000 20.63 19.03 1680.0 70.000 5.48 1656.0 69.000 4.63 5.06 20 3312.0 138.000 0.01 2976.0 124.000 0.08 0.04 3984.0 166.000 0.01 3648.0 152.000 0.01 0.0~1 1 __ 1 4536.0 189.000 0.01 0.01 -36 Table 9 placebo group Time Time Subject # Subject # Subject # Subject # Subject # (hr) (day) 1 7 11 14 17 Pre-Dose Pre-Dose Its Its Its Its Its 5 1.0 0.042 Its Its Its Its Its 1.5 0.063 Its Its Its Its Its 3.0 0.125 Its Its Its Its Its 8.0 0.333 Its Its Its Its Its 12.0 0.500 Its Its Its Its Its 10 24.0 1.000 Its Its Its its Its 72.0 3.000 Its Its Its Its Its 120.0 5.000 Its Its Its Its Its 168.0 7.000 Its Its Its Its Its 192.0 8.000 Its Its Its Its Its 15 360.0 15.000 Its Its Its Its Its 528.0 22.000 Its Its Its Its Its 864.0 36.000 Its Its Its Its Its Its = below the limit of detection -37 Study L297-007: Mean Pharmacokinetic Parameters by Treatment Group Data from individual patients are presented in Tables 10-14. Table 10 0.15 mg/kg LDP-02 IV Subject C. t. AUC, X. t% AUC AUC, V, CL (pg/ml) (days) (iig-day/ml) (1/day) (days) (pig-day/ml) (%) (mI/kg) (ml/day/kg) 5 2 10.667 0.33 16.4 0.2486 2.79 16.5 0.3 36.7 9.11 3 7.984 0.04 25.3 0.1196 5.79 27.1 6.7 46.3 5.53 4 4.292 0.13 16.9 0.1510 4.59 17.5 3.3 56.9 8.60 Mean 7.648 0.13* 19.5 0.1731 4.39 20.3 3.4 46.6 7.75 SD 3.201 5.00 0.0673 1.51 5.88 3.2 10.1 1.93 10 *Median value C. = maximum concentration t. = time to maximum concentration = a measure of elimination t, = terminal half-live 1 5 AUC, AUC = area under the curve using all time points AUC= AUC = area under curve extrapolated AUC ext (%) = % of area under curve attributed to extrapolation extrapolation V,= apparent volume of distribution CL = Clearance 20 Table 11 0.15 mg/kg LDP-02 SC Subject C. t. AUC, AUC AUC, V, CL ( ig/ml) (days) (pg-day/ml) (1/day) (days) (4g-day/ml) (%) (ml/kg) (ml/day/kg) 5 0.711 6.01 7.18 0.2298 3.02 7.32 2.0 89.1 20.5 6 0.927 4.01 8.71 0.2253 3.08 8.83 1.4 75.4 17.0 8 1.699 6.01 15.4 0.1003 6.91 18.0 14.3 82.9 8.32 25 Mean 1.112 6.01* 10.4 0.1852 4.33 11.4 5.9 82.5 15.3 SD 0.519 4.40 0.0735 2.23 5.80 7.3- 6.88 6.26 *Median value -38 Table 12 0.5 mg/kg LDP-02 IV Subject C. t. AUC, t. AUC AUCV V, CL (pig/ml) (days) (4g-day/nl) (1/day) (days) (pig-day/ml) (%) (ml/kg) (mil/day/kg) 9 24.388 0.06 82.2 0.1586 4.37 85.1 3.4 37.0 5.87 10 11.699 0.50 66.1' 0.2159 3.21 67.0 1.3 34.6 7.47 5 12 11.194 0.50 102.5 0.1543 4.49 103 0.8 31.4 4.84 Mean 15.760 0.50* 83.6 0.1763 4.02 85.1 1.8 34.3 6.06 SD 7.476 18.3 0.0344 0.71 18.2 1.4 2.84 1.32 *Median value Table 13 1.5 mg/kg LDP-02 IV 10 Subject C. t. AUC, t 1 AUG AUC, V, CL (pg/ml) (days) ( ±g-day/m1) (1/day) (days) (pg-day/ml) (%) (mi/kg) (ml/day/kg) 13 114.686 0.33 854 0.2316 2.99 855 0.1 7.58 1.75 15 134.975 0.06 408 0.0336 20.6 409 0.2 109 3.67 36 106.779 0.13 719 0.0331 20.9 1000 28.1 45.3 1.50 Mean 118.813 0.13* 660 0.0994 14.9 755 9.5 54.0 2.31 15 SD 14.544 229 0.1145 10.3 308 16.1 51.4 1.19 *Median value Table 14 2.5 mg/kg LDP-02 IV Subject C. t. AUG, t" AUC AUC, V. CL (p±g/ml) (days) (pg-day/ml) (1/day) (days) (g-day/ml) (%) (ml/kg) (ml/day/kg) 18 105.367 0.04 1489 0.0296 23.4 1680 11.3 50.2 1.49 20 19 98.131 0.06 1814 0.0642 10.8 1815 0.1 21.5 1.38 Mean 101.749 0.05* 1651 0.0469 17.1 1747 5.7 35.9 1.43 SD 5.117 229 0.0244 8.91 95.8 8.0 20.3 0.08 *Median value -39 L297-007: Serum ca4P7 Binding Over Time by Subject by Treatment Group, Data from individual patients are presented in Tables 15-20. For each subject the time of blood sampling, MESF of the sample and % of baseline (pre-dose) MESF is presented. 5 Table 15 0.15 mg/kg LDP-02 IV Subject #2 Subject # 3 Subject # 4 Mean Pre-Dose5689 100% Pre-Dose 5424 100% Pre-Dose 4177 100% 5097 100% 3 hr 605 11% 3 hr 591 11% 3 hr 588 14% 595 12% 24 hrs 589 10% 24 hrs 600 11% 24 hrs 631 15% 607 12% 10 Day 3 501 9% Day 3 496 9% Day 3 548 13% 515 10% Day 7 474 8% Day 7 473 9% Day 7 512 12% 487 10% Day 15 1819 32% Day 15 578 11% Day 15 599 14% 999 20% Day 22 2426 43% Day 22 558 10% Day 22 609 15% 1198 23% Day 36 3028 53% Day 36 3570 66% Day 36 3469 83% 3356 66% Day 163 6934 128% Day 163 6837 164% 3885 P35% Day 205 4675 86% Day 205 6755 162% 5715 412% 15 Table 16 0:15 mg/kg LDP-02 SC Subject # 5 Subject # 6 Subject # 8 Mean Pre-Dosej 6043 00% Pre-Dose 6779 100% Pre-Dose 5857 100% 6226 100% 3 hr 1797 30% 3hr 4727 70% 3 hr 1514 26% 2679 43% 20 2 637 11% 24 hrs 588 9% 24 hrs 616 11% 614 10% Day 3 529 9% Day 3 520 8% Day3 527 9% 525 8% Day 7 486 8% Day 7 474 7% Day7 485 8% 482 8% Day 15 598 10% Day 15 642 9% Day 15 635 11% 625 10% Day 22 759 13% Day 22 934 14% Day 22 579 10% 757 12% 25 Da 36 1455 24% Day 36 1452 21% Day 36 2799 48% 1902 31% Day 163 2743 45% Day 163 1989 29% Day 163 4621 . 79% 3118 50% Day 212 4201 70% Day 212 2601 38% Day 212 4832 1 82% 3878 62% Table 17 0.5 mg/kg LDP-02 IV Subject # 9 Subject # 10 Subect # 12 Mean 30 Pre-Dose 5519 100% Pre-Dose 5966 100% Pre-Dose 8550 100% 6678 00% 3 br 533 10% 3 hr 548 9% 3 hr 539 6% 540 8% 24 hrs 542 10% 24 hrs 554 9% 24 hrs 527 6% 541 8% Day 3 565 10% Day 3 574 10% Day3 539 6% 560 8% Day 7 544 10% Day 7 551 9% Day7 547 6% 547 8% 35 Dayl1 540 10% Day_15 525 9% Day5 520 6% 528 8% Day 22 555 10% Day22 572 10% Day 22 543 6% 557 8% Day 36 885 16% Day 36 1182 20% Day 36 643 8% 903 14% Day 149 4448 81% 5256 88% tDay 149 7810 91% 5838 87% -40 Table 18 1.5 mg/kg LDP-02 IV Subject # 13 Subject # 15 Subject # 16 Mean Pre-Dose 4966 100% Pre-Dose 15544 100% Pre-Dose 5622 100% 5378 100% 3 hr 518 10% 3 hr 539. 10% 3 br 545 10% 534 10% 5 24 hrs 482 10% 24 hrs 487 9% 24 hrs 520 9% 496 9% Day 3 511 10% Day 3 475 9% Day 3 514 9% 500 9% Day 7 549 11% Day 7 535 10% Day 7 569 10% 551 10% Day 15 472 9% Day 15 474 9% Day 15 491 9% 479 9% Day 22 603 12% Day 22 617 11% Day 22 576 10% 599 11% 10 Day 36 618 12% Day 36 866 16% Day 36 606 11% 697 13% Day 82 922 19% Day 80 832 15% 877 16% Day 134 1647 33% Day 134 1531 28% 1589 30% Day 176 2322 47% 3 43% Table-19 2.5 mg/kg LDP-02 IV 15 Subject # 18 Subject # 19 Mean Pre-Dose 5922 100% "Pre-Dose 5065 100% 5494 100% 3 hr 527 9% - 3 hr 527 10% 527 10% 24 hrs 568 10% 24 hrs 571 11% 569 10% Day 3 511 9% Day 3 521 10% 516 9% 20 Day 7 503 9% Day 7 513 10% 508 9% Day 15 530 9% Day 15 544 11% 537 10% Day 22 588 10% Day 22 595 12% 591 11% Day 36 550 9% Day 36 554 11% 552 10% Day 70 615 10% Day 69 566 11% 590 11% 25 Day 138 4572 77% Day 124 1103 22% 2837 52% Day 166 5603 95% Day 152 4094 81% 4849 88% -41 Table 20 placebo group Subject # 1 Subject # 7 Subject # 11 Subject # 14 Subject # 17 Pre- 5807 00% 5198 100% 8747 -100% 7017 100% 5982 100% Dose 3 hr 5630 97% 4305 83% 8454 97% 6208 88% 5520 92% 5 24 hrs 5672 115% 4347 84% 8033 92% 6699 95% 5410 90% Day 3 6078 105% 4008 77% 8701 99% 6141 88% 5488 92% Day 7 5617 97% 4047 78% 8668 99% 6327 90% 5194 87% Day 5797 100% 4758 92% 7516 86% 4851 69% 759 96% 15 10 Day 5164 89% 4318 83% 6924 79% 5246 75% 5922 99% 22 Day 6200 107% 4686 90% 7065 81% 7857 112% 5349 89% 36 -42 Example 2. Study L297-006 The study entitled, "A Single Dose Phase Ib/lIa, Placebo Controlled, Randomized, Double-Blind Study to Determine the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Effectiveness of LDP-02 in Patients 5 with Moderately Severe Ulcerative Colitis" was completed and certain final results are presented in this section. Study Rationale Results from the Phase I trial (Example 1. Study L297-007) in healthy volunteers showed LDP-02 at doses of 0.15 mg/kg SC and IV, 0.5 mg/kg IV, 1.5 10 mg/kg IV, and 2.5 mg/kg IV was safe and well-tolerated. In addition, doses of 0.15 mg/kg IV or SC and 0.5 mg/kg IV were shown to have a t, of approximately 100 to 130 hours and flow cytometry data showed that unbound a4P7 begins to reappear in the 0.15 mg/kg dosage groups approximately two weeks after dosing. Based upon these data, LDP-02 dosages of 0.15 mg/kg SC, 0.15 mg IV, 0.5 mg/kg IV, 15 and 2.0 mg/kg IV were selected for use in the initial study in patients with ulcerative colitis. This study was designed so that each dose of LDP-02 was determined to be safe and well-tolerated prior to escalation to the next dose level. Study Design The study was a randomized, double-blind, placebo-controlled, ascending 20 single-dose study in patients diagnosed with moderately-severe ulcerative colitis. Patients with a documented diagnosis of ulcerative colitis with a minimum disease extent of 25 cm from the anal verge -were potentially eligible for the study. Patients with severe ulcerative colitis as defined by Truelove-Witts criteria (Br Med J; 2:1042-1048 (1955)) were excluded. Ulcerative colitis patients who met all 25 inclusion/exclusion criteria were enrolled sequentially into four study groups and, within each study group, were randomly assigned to receive LDP-02 or placebo (i.e., 0.9% sodium chloride). Treatment groups and numbers of patients enroUed are shown in Table 21.
-43 Table 21: Study Groups Route of LDP-02 Placebo Group Administration* # patients Dose # patients 1 SC 5 0.15 mg/kg 2 2 IV 5 0.15 mg/kg 2 5 3 TV 5 0.5 mg/kg 2 4 IV 5 2.0 mg/kg 2 Study medication (LDP-02 or placebo) was administered on Day 1 either SC into the thigh or via a 30 minute IV infusion. Safety assessments included recording of adverse events, physical examinations, vital signs, clinical laboratories 10 (i.e., hematology, blood chemistries, and urinalysis), plasma cytokine levels, and ECGs. Blood was drawn at various time points.to measure LDP-02 serum concentrations and to assess the effectiveness of LDP-02 to saturate and block a407 binding receptors on peripheral blood lymphocytes. The effectiveness of LDP-02 to reduce inflammation in the colon was measured by clinical disease 15 observations, endoscopic appearance, histopathology, and immunohistochemistry. Study Results Patient Disposition and Demography Twenty-nine patients with moderately-severe ulcerative colitis were entered into the study and 28 completed the trial. One patient was found to be Clostridium 20 difficile toxin positive at screening, but due to a delay in reporting laboratory results the patient was admitted into the trial and received 2.0 mg/kg TV of LDP-02. Once the laboratory results were obtained, the patient was treated with antibiotics and replaced by another patient. There were no other patients discontinued from the study. As patients were recruited into the study over time, there was no attempt 25 to balance the treatment groups with regard to baseline ulcerative colitis history. As such, severity and duration of ulcerative colitis disease and prior medications -44 for ulcerative colitis varied from patient to patient and from treatment group to treatment group. These data are presented in Table 22. Table 22: Ulcerative Colitis History by Treatment Group Treatment Group Time Since Time Since # of Acute Weeks on Weeks on Onset of UC Diagnosis Exacerbations continuous continuous Symptoms of UC in past oral 5-ASA oral (yrs)' (yrs)' 12 months' in past 6 steroids months' in past 6 months' 5 0.15 mg/kg SC 5.32 4.6 3 24.0 .0 (n=5)- (4.8,6.4) (4.3,6.4) (1,12) (3,26) (0,6) 0.15 mg/kg IV 9.58 4.9 1 24.0 10 (n-5) (2.6,14.2) (2.1,14.0) (1,3) (6,26) (0,24) 0.5 mg/kg IV 10.8 9.0 1 26.0 0 10 (n=5) (0.4,11.8) (0.3,11.8) (1,2) (0,26) (0,15) 2.0 mg/kg IV 9.34 7.65 2 25.0 5 (n=6) (3.4,58.8) (3.2,19.4) (1,5) (0,26) (0,26) All LDP-02 5.99 4.9 2 26.0 0 (n=21) (0.4,58.8) (0.3,19.4) (1,12) (0,26) (0,26) 15 Placebo 5.27 4.85 1.5 24.0 16 (n=8) (0.4,11.0) (0.3,9.7) (1,4) (0,26) (0,26) Median values Disease Measurements Although this was primarily a dose-ranging safety and pharmacokinetics 20 study, various parameters were measured to assess effectiveness of treatment. Effectiveness assessments included recording changes from baseline using a modified Baron's (endoscopy) Scoring System, the Mayo Clinic Disease Activity Index Score, the Powell-Tuck Disease Activity Index Score, stool frequency, and the Inflammatory Bowel Disease Questionnaire. Changes from baseline to Day 30 25 for these parameters are shown in Table 23. For patients in which there was no Day 30 evaluation, the last post-baseline observation obtained was carried forward to Day 30.
-45 Table 23: Change from Baseline to Day 30 in Disease Parameters Change from baseline to Day 30' Treatment Group Endoscopic Mayo Powell-. Stool Total Severity Clinic Tuck Frequency 1BDQ Score Activity Activity Index Index 0.15 mg/kg 0 -3.0 -3.0 -1.0 14.0 5 SC (-2,0) (-9,0) (-6,-2) (-7,1) (14,72) (n=5) 0.15 mg/kg 0 -1.0 0 -0.4 8.0 IV (0,1) (-3,2) (-3,3) (-5,2) (-3,95) (n=5) 10 0.5 mg/kg IV -2.0 -10 -6.0 -5.3 37.0 (n=5) (-3,0) (-11,0) (-13,-2) (-6,0) - (14,80) 2.0 mg/kg IV -0.5 -2.0 -1.5 -3.2 -2.5 (n=6) (-2,1) (-6,3) (-5,-5) (-8,2) (-59,95) All LDP-02 0 -3.0 -3.0 -2.4 14.0 15 (n=21) (-3,1) (-11,3) (-13,5) (-8,2) (-59,95) Placebo -1.0 -5.0 -6.0 -3.2 53.5 (n=8) (-3,2) (-8,4) (-9,-4) (-12,2) (-30,82) 'Median values and range. For patients without a Day 30 evaluation the last post-baseline evaluation was carried forward to Day 30. 20 As seen from the results presented in Table 23, there was variability in response among the different treatment groups. The patients receiving 0.5 mg/kg IV appeared to have the best responses; the median endoscopic severity score was reduced by two grades and the Mayo Clinic score was reduced by 10 points with a decrease in stool frequency. Three of the five patients receiving 0.5 mg/kg IV had 25 a two point improvement in the modified Baron sigmoidoscopy score which is considered an endoscopic response; only one patient (compared with a total of five treated per group) in both the 2.0 mg/kg IV and 0.15 mg/kg SC groups had an endoscopic response. The placebo group also experienced an improvement in -46 sigrnoidoscopic score and Mayo Clinic score, although both were less in magnitude when compared to the 0.5 mg/kg IV group. Two of the eight patients experienced an endoscopic response. The number of patients with a complete remission, defined as a zero on the 5 modified Baron sigmoidoscopic score and on the Mayo Clinic score at Day 30, are reported in Table 24. Table 24: Patients in Complete Remission at Day 30 Measured at Day 30' Treatment 10 Group Number of Patients with Percentage of Patients in Complete Remission Complete Remission 0.15 mg/kg 0 0 SC (n=5) 0.15 mg/kg 0 0 15 IV (n=5) 0.5 mg/kg IV 2 40% (n=5) 2.0 mg/kg IV 0 0 20 (n=6) All LDP-02 2 9.5% (n=2 1) Placebo 0 0 (n=8) 25 'Zero on the modified Baron Score and the Mayo Clinic Score in Day 30 results -47 None of the patients in the placebo group experienced a complete remission while two patients among those receiving LDP-02 had complete remissions. The two patients both were in the same group; both patients received a single administration of 0.5 mg/kg of LDP-02. One of the patients was receiving 5 concurrent mesalamine therapy, while the other was receiving concurrent low dose corticosteroid (20 mg prednisone per day orally). Pharmacokinetics The assay of LDP-02 in serum was performed by Cytometry Associates, Inc. as previously described (Study L297-007). Blood samples were collected prior to 10 and immediately following the completion of infusion (Day 1) and on Days 2, 3, 5, 10, 14, 21, 30 and 60 to assess the pharmacokinetic profile of LDP-02. LDP-02 concentrations over time by individual patient and mean phannacokinetic parameters by LDP-02 dose are presented in the Appendix to study L296-006. 15 As seen in FIG. 8, serum levels of LDP-02 for the 0.15 mg/kg IV and SC groups fall to <1.0 ig/mL approximately 20 days post-dose. For the 2.0 ng/kg dose group, LDP-02 levels remain elevated out to approximately Day 60. Table 25 presents the key pharmacokinetic parameters by treatment group.
-48 Table 25: Pharmacokinetic Parameters of LDP-02 Dose and Route of Administration of LDP-02 (number of subjects with data) 2 Pharmacokinetic 0.15 mg/kg SC 0. 15 mg/kg 0. 5 mg/kg 2. 0 Parameter' (n=5) IV IV mg/kg IV (n=5) (ni=5) (n=4 ) C. (ig/mL) 1.44 3.602 10.544 32.933 (0.33) (0.958) (2.582) (3.360) 5 t. (days) 5 0.13 0.13 0.13 (median & range) (3-10) (0.13-0.13) (0.13-0.13) (0.13-2) t., (days) 15.63 18.91 10.62 15.0 (15.92) (20.97) (5.23) (5.36) AUCj, 25 27 91 515 (pLg-day/mL) (16) (11) (32) (93) 10 (1/day) - 0.1226 0.0879 0.0927 0.0542 (0.1064) (0.0757) (0.0775) (0.0298) AUC(INF) 31 34 100 553 (pg-day/ftL) (23) (18) (39) (116)
CL
4 9.21 7.75 6.06 2.31 (mLday/kg) (9.54) (1.93) (1.32) (1.19) 15 V.
4 (mL/kg) 95.08 101.05 77.63 76.64 (54.19) (62.87) (30.90) (20.03) 'All values are mean +/- SD unless otherwise indicated. The SD appears in parenthesis. 2 Two patients, one in the 0.15 mg/kg SC and one in the 0.5 mg/kg IV groups had evaluable data through Study Day 21 with measurement at later times which were not physiologically possible. 20 3 One patient in the 2.0 mg/kg IV dosing group was withdrawn at Study Day 10 and had a surgical intervention. The pharmacokinetic results for this patient are not included. "Clearance and volume terms for the SC dose group are the apparent clearance (CL/F) and apparent volume (V/F).
-49 There does appear to be linearity with dose for the maximum concentration of LDP-02 and the area under the curve measured after IV administration. The clearance and the terminal elimination half life appear to be independent of IV dose administered. The volume of distribution appears to decrease slightly with 5 increasing doses of IV LDP-02. Assessment of the Pharmacodynaiic Effect of LDP-02 FACS analysis to measure the presence of a4P7 sites on blood lymphocytes was previously described (Study L296-007). Serum a407 binding over time (i.e., MESF values and percentage of baseline at each post-dose time point) are 10 presented by individual patient and by treatment group in the Appendix to Study L297-006. Mean percent of baseline MESF over time for all treatments are presented in FIG. 9. As seen in FIG. 9, percent of baseline MTESF rapidly falls to approximately 10% after SC and IV administration of LDP-02 with duration of effect dependent 15 upon dose. Starting at about day 10, c4p7 signal started to return to baseline for the 0.15 mg/kg IV and SC dose groups. However, x4P7 signal started to return to baseline between day 30 and day 60 for the 0.5 mg/kg IV and 2.0 mg/kg dose groups. Conclusions 20 Administration of LDP-02 at doses of 0.15 mg/kg IV and SC, 0.5 mg/kg IV, and 2.0 mg/kg IV to patients with moderately-severe ulcerative colitis was well tolerated. The pharmacokinetic and pharmacodynamic data from patients with ulcerative colitis were consistent with those found in healthy volunteers. There 25 appeared to be linearity with dose for the maximum concentration of LDP-02 and area under the curve measured after IV administration. The clearance and the terminal elimination half life appeared to be independent of TV dose administration. The volume of distribution appeared to decrease slightly with increasing doses of IV LDP-02. The percent of baseline MESF declined to -10% rapidly after SC and 30 IV administration of LDP-02 with duration of effect dependent upon dose. For the -50 0.15 mg/kg TV and SC dose groups, percent of baseline MESF started returning to baseline approximately 10 days after dosing whereas this started to occur at -30 days and -60 days for the 0.5 mg/kg IV and 2.0 mg/kg dose groups, respectively. Appendix to Study L297-006 5 LDP-02 Serum Concentration Over Time by Subject by Treatment Group. Data obtained from individual subjects are presented in Tables 26-30. The data presented in Tables 26-30 are in gg/mL. Table 26 Group 1: 0.15 mg/kg LDP-02 SC Time Subject # Subject # Subject # Subject # Subject # 10 (day) 201101 301103 302105 304107 401104 Pre-Dose BQL BQL BQL BQL BQL 0.125 BQL 0.07 BQL BQL NS 2 - 0.61 0.91 0.94 1.01 1.29 3 0.90 1.10 1.29 1.49 1.65 15 5 0.76 1.48 NR 1.66 1.74 10 0.15 1.12 1.40 0.92 1.44 14 BQL 0.61 0.78 0.24 0.99 21 BQL BQL NS 0.11 0.65 30 BQL 0.33 0.84 0.26 0.12 20 60 BQL 0.23 0.37 0.30 BQL BQL= reported as non-detectable NS= no sample received from laboratory -51 Table 27 Group 2: 0.15 mgc/kg LDP-02 IV Time Subject # Subject # Subject # Subject# Subject # (Day) 101201 102202 305204 402203 403206 Pre-Dose BQL BQL BQL BQL BQL 5 0.125 4.14 4.88 3.35 2.34 3.30 2 NR 2.74 1.92 1.83 2.34 3 3.12 3.15 1.55 1.42 2.03 5 1.82 1.83 1.33 0.82 1.19 10 0.81 0.88 0.86 0.37 0.79 10 14 0.32 0.15 BQL 0.23 0.26 21 0.38 0.12 0.10 BQL BQL 30 0.38 BQL 0.40 BQL 0.05 60 0.24 BQL 0.36 BQL 0.14 NR= no sample result reported from laboratory 15 Table 28 Group 3: 0.5 mg/kg LDP-02 IV Time (day) Subject # Subject# Subject Subject # Subject # 206302 208303 309306 502304 503307 Pre-Dose BQL BQL BQL BQL BQL 0.125 14.06 12.33 7.90 8.67 9.76 2 10.01 8.51 5.73 5.84 8.26 20 3 6.56 6.45 4.96 4.67 7.27 5 4.15 5.52 3.59 2.94 5.61 10 3.17 4.46 2.81 3.11 4.21 14 2.51 0.14 2.46 1.14 3.01 21 BQL 0.17 0.14 BQL 2.04 25 30 BQL 0.48 BQL 0.06 1.29 60 0.41 1.73 0.10 0.28 BQL Table 29 Group 4: 2.0 mg/kg LDP-02 IV Time Subject # Subject # Subject # Subject # Subject # Subject # (day) 104403 210402 310415 404401 504405 506407 30 Pre-Dose BQL BQL BQL BQL BQL BQL 0.125 30.45 38.83 37.66 29.71 28.90 32.18 2 32.18 28.22 35.14 27.49 27.49 26.87 3 23.93 17.40 27.49 24.45 22.92 22.46 5 21.52 15.34 21.52 18.42 21.52 17.79 35 10 13:10 41.11 14.82 13.10 10.99 11.96 14 11.72 3.13 13.10 11.23 1.22 9.03 21 7.53 0.08 10.99 8.55 0.12 5.70 30 5.80 BQL 8.26 7.02 NR 4.19 60 1.71 0.41 2.24 1.95 NR 1 0.06 -52 Table 30 placebo group Time Subject Subject Subject Subject Subject Subject Subject # Subject (day) # # # # # # 209404 # 202102 303106 103205 306207 308305 501301 505406 Pre-Dose BQL BQL BQL BQL BQL BQL BQL BQL 5 0.125 BQL BQL BQL BQL BQL BQL BQL BQL 2 BQL BQL BQL BQL BQL BQL BQL .BQL 3 BQL BQL BQL BQL BQL BQL BQL BQL 5 BQL BQL BQL BQL BQL BQL BQL BQL 10 BQL BQL BQL BQL BQL BQL BQL BQL 10 14 BQL BQL BQL BQL BQL BQL BQL BQL 21 BQL BQL NR BQL BQL BQL BQL BQL 30 BQL BQL BQL BQL BQL BQL BQL BQL 60 BQL BQL BQL BQL BQL BQL IBQL BQL BQL below quantitation limit.
-53 Mean Pharmacokinetic Parameters by Treatment Group. Data obtained from individual subjects are presented in Tables 31-34. Table 31 Group 1: 0.15 mg/kg LDP-02 SC Subject C. tmu t. AUC 1 AUC CL V 2 (p g/mL) (days) (days) (pLg-day/m (1/day) ([g-day/mL) (mL/day/kg) (mL/kg) 5 201101 0.90 3 2.58 5.30 0.2692 5.86 25.61 95.15 301103 1.48 5 34.61 30.39 0.0200 41.87 3.58 178.87 302105 1.40 10 31.35 46.94 0.0221 63.68 2.36 106.55 304107 1.66 5 3.88 15.41 0.1788 16.02 9.36 52.37 401104 1.74 5 5.72 28.17 0.1212 29.16 5.14 42.45 10 Mean 1.436 5.6 15.628 25.242 0.1223 31.318 9.21 95.078 SD 0.329 2.607 15.921 15.813 0.1064 22.613 9.54 54.190 C = maximum concentration t = time to maximum concentration , = a measure of elimination 15 t 1 ,, = terminal half-live AUC, AUCI = area under the curve using all time points AUC =AUC = area under curve extrapolated AUC ext (%) = % of area under curve attributed to extrapolation extrapolation V, = apparent volume of distribution 20 CL = Clearance Table 32 Group 2: 0.15 mg/kg LDP-02 IV Subject C. t. t" AUCJI AUC CL V, (pg/mL) (days) (days) (pg-day/mL) (1/day) (pg-day/mL) (mL/day/kg) (mL/kg) 101201 4.14 0.13 54.69 39.64 0.0127 58.58 2.56 202.06 102202 4.88 0.13 3.62 25.15. 0.1914 25.78 5.82 30.39 25 305204 3.35 0.13 19.37 34.17 0.0358 44.23 3.39 94.77 402203 2.34 0.13 4.88 12.10 0.1420 13.72 10.94 77.03 403206 3.30 0.13 11.99 23.28 0.0578 25.70 5.84 100.99 Mean 3.602 0.13 18.91 26.868 0.0879 33.602 5.71 101.05 SD 0.9579 0 20.97 10.611 0.0757 17.718 3.27 62.87 -54 Table 33 Group 3: 0.5 mg/kg LDP-02 IV Subject C. t. t, AUC.n xz AUC CL S(pg/mL) (days) (days) (pg-day/mL) (I/day) (ig-day/nL) (mL/day/kg) (mUkg) 206302 14.06 0.13 17.21 139.26 0.0403 149.44 3.35 83.08 208303 12.33 0.13 3.02 74.99 0.2293 75.73 6.60 28.79 5 309306 7.90 0.13 9.22 67.49 0.0751 68.82 7.27 96.69 502304 8.67 0.13 10.52 65.34 0.0659 69.59 7.19 109.09 503307 9.76 0.13 13.11 109.80 0.0529 134.20 3.73 70.48 Mean 10.544 0.13 10.616 91.376 0.0927 99.556 5.628 77.626 SD 2.582 0 5.229 32.207 0.0775 39.048 1.928 30.90 10 -55 ~O r- 01% t1 0 C n 00 Ln00 - - 0 ~ -< - \ ~0 0 0i 0 kn G o Do o0 - 0 (n V3 enO W
-
(0 .0 04 00 C E 0 en0 M C) C -56 0) r- -o cn e C14en 0 o *o, co ~0000 0000 O\7% t~ C cn 0 r-C>r 00 00 V) m~ 00 0o ~rt- 0 0 N00 CL) L E 0 C) C o C:, CL) 0) N0 %0 c , V ) q V en r- 0- C .) - 0) 0 00 '3ro. 0 0) .rq 0\ 0 E4 CC C; m r ~ 0 -57 C) C7\ M C)* m 0 N 0% - 00 00 tn cn0 N r- [*N 00 r- 0 - ON en N N 0000 Ln Ch%1 -r% 00 0 0----0 t-- V 00 4 C) CN 0 0, - n0 u \- r4ONO N00 00 v, r 3- en ch C)) 0 m00 ) 0 4F o C.) N= w- M -0 r -)>C4v rn m * 00 CDm n co M r 00 - Nr O>0 0\ rn00 \10\0 q V t OC N~ fO N Cri C) - tO t4 r 'T mLn 0 oo 0 C-0 00 00 Ln - C N toN0 0-- N - w (I O r-' kf N 0m 141: ~O~~ N N- N N4 Mo Mo M D ccQ00 N- NO C ) 0000 N 010C ON (7 0>
CL
-58 o~~~ N~ NN '. C-00-4 o -- V-0 D(4 c n Co 0 \0' -_ IR '. 00 0N 0\- N C> 0" fn C>0 ir~On 10 -> 0' -Nor D mo>I " - - - C' 4 0 0t V d). o o 0 00 0 \ 0r I -v)o CD) (U V- ~ 0.0~ r-0 0O O. r .0, DO O J -. 'o. oo r-m . c 0u m .0 00 N 0% 0 0.0 NN o C>'0~ N r Do 0 'l -- T -n-'n o V) 3- - 00 Q' 0 0 in'. ).00N00 0)4 C> 00 CN%0 00 r, r- o r 2 0 - C C) w - - n - q -t ON -r InJ -~r C,) r-\o00 040 6 M Ln ~0 -~~~C Z )0 U~~0 -59 r- o rN N Vn C . V- I'D U) C=) -: O't r ~0 CN C1 \0 -'4rC>co0 r- O\ en ON C) r r, CNO W3D C= 0 00\>'nk n V
)
C) - a0 - ;! v ,T U 00 r- V) Nr V kn C> C-e kr 00 Mj r- 00 DO0- r .. , OC14 %0- r m IT - 1 r- V -I-
-
4 ON 00 t (- 0-IT - r33 00 N n 0 VI VN ONGIN . z 0 Ir!r 0 0 r-. -O o 00 o ~ t - _ _- - C.) -60 Table 40 Placebo group Time Days Subject # Subject 505406 Mean 1__ _ 209404 Mean Pre-Dose 11012 100% 7579 100% 10095 100% 0.125 11826 107% 9025 119% 10396 103% 5 3 10549 96% 8792 116% 9919 98% 5 11614 105% 6217 82% 8710 86% 10 8238 75% 7150 94% 9170 91% 14 8382 76% 4787 63% 7689 76% 21 7031 64% 7160 94% 9300 92% 10 30 6817 62% 8166 108% 8375 83% 60 While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without 15 departing from the scope of the invention encompassed by the appended claims.

Claims (30)

1. A method for treating a human having a disease associated with leukocyte infiltration of mucosal tissues, comprising administering to said human an effective amount of a human immunoglobulin or antigen-binding fragment thereof having binding 5 specificity for a4p7 integrin, wherein said immunoglobulin or antigen-binding fragment thereof specifically binds a4p7 but does not bind a4P, wherein said immunoglobulin or fragment is administered in an initial dose followed by one or more subsequent doses and the minimum interval between any two doses is a period of at least about I day, and wherein no more than about 8 mg immunoglobulin or antigen-binding fragment per kg 1a body weight are administered during a period of about one month.
2. The method of claim 1, wherein each of said doses independently comprise about 0.1 to about 8 mg immunoglobulin or fragment per kg body weight.
3. The method of claim 1, wherein each of said doses independently comprise about 0.15, about 0.5, about 1.0, about 1.5 or about 2.0 mg immunoglobulin or fragment is per kg body weight.
4. The method of claim 1, wherein the interval between doses is at least about 7 days, at least about 14 days, at least about 21 days, at least about 28 days, at least about 30 days, at least about 40 days or at least about 50 days.
5. The method of claim 1, wherein said each of said doses independently 20 comprise an amount of immunoglobulin or fragment which is sufficient to achieve a) about 60% or greater saturation of a4p7 integrin binding sites on circulating lymphocytes and/or b) about 60% or greater inhibition of a4p7 integrin expression on the cell surface of circulating lymphocytes, and wherein said saturation and/or inhibition is 25 maintained for a period of at least about 10 days following administration of said dose.
6. The method of claim 5, wherein each of said doses independently comprise an amount of immunoglobulin or fragment which is sufficient to achieve a) about 70% or greater saturation of a467 integrin binding sites on circulating lymphocytes and/or 30 b) about 70% or greater inhibition of a4p 7 integrin expression on the cell surface of circulating lymphocytes.
7. The method of claim 5, wherein each of said doses independently comprise an amount of immunoglobulin or fragment which is sufficient to achieve 62 a) about 80% or greater saturation of a4p7 integrin binding sites on circulating lymphocytes and/or b) about 80% or greater inhibition of a4p7 integrin expression on the cell surface of circulating lymphocytes. 5
8. The method of claim 5, wherein each of said doses independently comprise an amount of immunoglobulin or fragment which is sufficient to achieve and maintain said saturation and/or inhibition for a period of at least about 14 days, at least about 30 days, at least about 40 days, at least about 50 days or at least about 60 days following administration of said dose. 10
9. The method of claim 5, wherein each of said doses independently comprise about 0.1 to about 8 mg immunoglobulin or fragment per kg body weight.
10. The method of claim 5, wherein each of said doses independently comprise about 0.15, about 0.5, about 1.0, about 1.5 or about 2.0 mg immunoglobulin or fragment per kg body weight. 15
11. The method of claim 5, wherein the interval between doses is at least about 7 days, at least about 14 days, at least about 21 days, at least about 28 days, at least about 30 days, at least about 40 days or at least about 50 days.
12. The method of claim 1, wherein a humanized immunoglobulin is administered and each of said doses comprises an amount of immunoglobulin which is sufficient to 20 achieve and maintain a serum concentration of immunoglobulin of at least about 1 Ig/mL for a period of at least about 10 days following administration of said dose.
13. The method of claim 12, wherein each of said doses independently comprise an amount of immunoglobulin which is sufficient to achieve and maintain said serum concentration for a period of at least about 14 days, at least about 30 days, at least about 25 40 days, at least about 50 days or at least about 60 days following administration of said dose.
14. The method of claim 12, wherein each of said doses independently comprise about 0.1 to about 8 mg immunoglobulin per kg body weight.
15. The method of claim 12, wherein each of said doses independently comprise 30 about 0.15, about 0.5, about 1.0, about 1.5 or about 2.0 mg immunoglobulin or fragment per kg body weight.
16. The method of claim 12, wherein the interval between doses is at least about 7 days, at least about 14 days, at least about 21 days, at least about 28 days, at least about 30 days, at least about 40 days or at least about 50 days. 63
17. The method of claim 1, further comprising administering an effective amount of one or more additional therapeutic agents.
18. The method of claim 17, wherein said agents are selected from the group consisting of steroids, immunosuppressive agents, non-steroidal anti-inflammatory agents 5 and immunomodulators.
19. The method of claim 17, wherein said agents are selected from the group consisting of azathioprene, 6-mercaptopurine, sulfasalazine, 5-amino salicylic acid, prednisone and prednisolone.
20. The method of claim 1, wherein said disease associated with leukocyte io infiltration of mucosal tissues is selected from the group consisting of an inflammatory bowel disease, pancreatitis, insulin-dependent diabetes mellitus, mastitis, cholecystitis, cholangitis, pericholangitis, chronic bronchitis, chronic sinusitis, asthma and graft versus host disease.
21. The method of claim 1, wherein said disease associated with leukocyte is infiltration of mucosal tissues is an inflammatory bowel disease.
22. The method of claim 21, wherein said inflammatory bowel disease is ulcerative colitis.
23. The method of claim 21, wherein said inflammatory bowel disease is Crohn's disease. 20
24. A method for treating a human having inflammatory bowel disease, comprising administering to said human an effective amount of a human immunoglobulin or antigen-binding fragment thereof having binding specificity for a4$7 integrin, wherein said immunoglobulin or antigen-binding fragment thereof specifically binds a4p7 but does not bind a4pl, wherein said immunoglobulin or fragment is administered in an 25 initial dose followed by one or more subsequent doses and the minimum interval between any two doses is a period of at least about 1 day, and wherein no more than about 8 mg immunoglobulin or fragment per kg body weight are administered during a period of about one month.
25. The method of claim 24, wherein the inflammatory bowel disease is 30 ulcerative colitis.
26. The method of claim 24, wherein the inflammatory bowel disease is Crohn's disease.
27. A method for inhibiting relapse and/or recurrence of quiescent inflammatory bowel disease in a human, comprising administering to said human an effective amount 64 of a human immunoglobulin or antigen-binding fragment thereof having binding specificity for a4p7 integrin, wherein said immunoglobulin or antigen-binding fragment thereof specifically binds a4P7 but does not bind a4p I, wherein said immunoglobulin or fragment is administered in doses and the minimum interval between doses is a period of 5 at least about 7 days, and wherein no more than about 8 mg immunoglobulin or fragment per kg body weight are administered during a period of about 30 days.
28. The method of claim 27, wherein quiescence has been induced by medical or surgical therapy.
29. The method of claim 27, wherein said inflammatory bowel disease is 1o ulcerative colitis.
30. The method of claim 27, wherein said inflammatory bowel disease is Crohn's disease. Dated 14 November, 2011 15 Millennium Pharmaceuticals, Inc. Genentech Inc. Patent Attorneys for the Applicant/Nominated Person SPRUSON & FERGUSON
AU2009200156A 2000-04-14 2009-01-15 Method of administering an antibody Expired AU2009200156B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2009200156A AU2009200156B2 (en) 2000-04-14 2009-01-15 Method of administering an antibody

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US09550082 2000-04-14
US09748960 2000-12-27
AU2001251629 2001-04-13
AU2006220426A AU2006220426B2 (en) 2000-04-14 2006-09-21 Method of administering an antibody
AU2009200156A AU2009200156B2 (en) 2000-04-14 2009-01-15 Method of administering an antibody

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2006220426A Division AU2006220426B2 (en) 2000-04-14 2006-09-21 Method of administering an antibody

Publications (2)

Publication Number Publication Date
AU2009200156A1 AU2009200156A1 (en) 2009-02-12
AU2009200156B2 true AU2009200156B2 (en) 2011-12-08

Family

ID=37153768

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2006220426A Expired AU2006220426B2 (en) 2000-04-14 2006-09-21 Method of administering an antibody
AU2009200156A Expired AU2009200156B2 (en) 2000-04-14 2009-01-15 Method of administering an antibody

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU2006220426A Expired AU2006220426B2 (en) 2000-04-14 2006-09-21 Method of administering an antibody

Country Status (1)

Country Link
AU (2) AU2006220426B2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998006248A2 (en) * 1996-08-15 1998-02-19 Leukosite, Inc. HUMANIZED IMMUNOGLOBULIN REACTIVE WITH α4β7 INTEGRIN

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998006248A2 (en) * 1996-08-15 1998-02-19 Leukosite, Inc. HUMANIZED IMMUNOGLOBULIN REACTIVE WITH α4β7 INTEGRIN

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Gordon F. H. et al (1999) Gastroentrology, 116 (abstract G3151) *
Gordon F. H. et al (1999) Gastroentrology, 116 (abstract G3152) *

Also Published As

Publication number Publication date
AU2009200156A1 (en) 2009-02-12
AU2006220426B2 (en) 2008-10-30
AU2006220426A1 (en) 2006-10-12

Similar Documents

Publication Publication Date Title
US20200002423A1 (en) Method of administering an antibody
CA2597717C (en) Antibodies against cxcr4 and methods of use thereof
JP4857259B2 (en) Use of anti-α5β1 antibodies to inhibit cancer cell growth
US8017116B2 (en) Chimeric and humanized antibodies to α5β1 integrin that modulate angiogenesis
CA3168591A1 (en) Plasma kallikrein binding proteins
EP3858862A9 (en) Anti pd-l1 antibody and use thereof
KR102667332B1 (en) How to Treat or Prevent Graft Versus Host Disease
JP5773882B2 (en) Treatment for pemphigus containing anti-FasL ligand antibody
CN116327920A (en) Method for preventing graft versus host disease
AU2009200156B2 (en) Method of administering an antibody
RU2361614C2 (en) APPLICATION OF ANTIBODIES AGAINST α5β1 FOR INHIBITION OF CANCER CELLS PROLIFERATION
AU2019204295A1 (en) Plasma kallikrein binding proteins

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired