WO1995009652A1 - Treatment of autoimmune and inflammatory disorders - Google Patents

Treatment of autoimmune and inflammatory disorders Download PDF

Info

Publication number
WO1995009652A1
WO1995009652A1 PCT/GB1994/000462 GB9400462W WO9509652A1 WO 1995009652 A1 WO1995009652 A1 WO 1995009652A1 GB 9400462 W GB9400462 W GB 9400462W WO 9509652 A1 WO9509652 A1 WO 9509652A1
Authority
WO
WIPO (PCT)
Prior art keywords
tnf
antibody
agent
inflammatory
mammal
Prior art date
Application number
PCT/GB1994/000462
Other languages
French (fr)
Inventor
Marc Feldmann
Ravinder Nath Maini
Richard Owen Williams
Original Assignee
The Kennedy Institute For Rheumatology
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
Priority claimed from PCT/GB1993/002070 external-priority patent/WO1994008619A1/en
Priority to PCT/GB1994/000462 priority Critical patent/WO1995009652A1/en
Priority to JP7510657A priority patent/JPH09510952A/en
Priority to EP94908462A priority patent/EP0765171A1/en
Priority to AU61495/94A priority patent/AU6149594A/en
Priority to US08/617,737 priority patent/US20020068057A1/en
Application filed by The Kennedy Institute For Rheumatology filed Critical The Kennedy Institute For Rheumatology
Publication of WO1995009652A1 publication Critical patent/WO1995009652A1/en
Priority to US08/690,775 priority patent/US6270766B1/en
Priority to US09/093,450 priority patent/US6770279B1/en
Priority to US09/754,004 priority patent/US20020010180A1/en
Priority to US09/921,937 priority patent/US20020136723A1/en
Priority to US10/762,096 priority patent/US20040228863A1/en
Priority to US11/225,631 priority patent/US7846442B2/en
Priority to US12/583,851 priority patent/US20110123543A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders

Definitions

  • T-cells The nature of autoantigens responsible for autoimmune disorders is not known, nor is the action which triggers the autoimmune response.
  • CD4 is a non-polymorphic surface
  • CD4 receptors define distinct subsets of mature peripheral T cells.
  • CD4 T cells expressing helper or regulatory functions interact with B cells in immune responses, while T cells expressing the
  • CD8 surface antigen function as cytotoxic T cells and have regulatory effects on immune responses. Since T-cell receptors are the pathway through which stimuli augment or modulate T-cell responses, they present a potential target for immunological intervention.
  • CD4+ T cells with antigen presenting cells lies at the root of the immune response. Many aspects of the autoimmune response are essentially similar to that of normal immune responses. Thus CD4+ autoantigen reactive T cells are restimulated by APC expressing class II with autoantigen peptides in the binding groove. In certain human diseases the evidence that this occurs has been provided: in
  • Graves' disease of the thyroid, in vivo activated T cells are present in the glands that are removed for refractory disease, and many of these cells after cloning can be shown to recognize autologous thyrocytes (as APC) not extrinsically supplied with any antigen, or APC supplied with the thyroid specific antigens thyroid peroxidase or thyroglobulin (Londei, M. et al.. ,cience 228: 85-89
  • CD4+ T cells from the blood have been cloned, including CD4+ T cells recognizing the acetylcholine receptor in myasthenia gravis (Hohlfeld, R. et al. , Nature 310: 224-246 (1984)); myelin basic protein in multiple sclerosis (Hafler, D.A. et al.. J. Immunol. 139: 68-72 (1987)); or islet cell membranes in insulin dependent diabetes mellitus (De Berardinis, P. et al.,
  • TNF ⁇ tumor necrosis factor- ⁇
  • cachectin also termed cachectin
  • TNF ⁇ is a protein secreted primarily by monocytes and macrophages in response to endotoxin or other stimuli as a soluble homotrimer of 17 kD protein Subunits (Smith, R.A. et al., J. Biol. Chem. 262: 6951-6954 (1987)).
  • a membrane-bound 26 kD precursor form of TNF has also been described (Kriegler, M. et al., Cell 53: 45-53 (1988).
  • TNF ⁇ is not limited to cells of the monocyte/macrophage family: TNF is also produced by CD4+ and CD8+ peripheral blood T lymphocytes, and by various cultured T and B cell lines (Cuturi, M.C. et al., J. EXP. Med. 165: (1581 (1987);
  • the current invention pertains to the discovery that combination therapy, involving the use of a CD4+ T cell inhibiting agent in conjunction with a TNF antagonist, produces markedly superior results than the use of each agent alone in the treatment of autoimmune or inflammatory disease, particularly in rheumatoid arthritis.
  • CD4+ T cell inhibiting agents include agents which block,
  • CD4+ T cells diminish, inhibit, or interfere with the activation of CD4+ T cells or the interaction of CD4+ T cells with antigen presenting cells (APC), such as antibodies to T cells or to their receptors; antibodies to APC or to their receptors; and other appropriate peptides or small
  • TNF antagonists include agents which block, diminish, inhibit, or interfere with TNF activity, TNF receptors, or TNF synthesis, such as anti-TNF antibodies; soluble TNF receptors; and other appropriate peptides or small molecules.
  • anti-CD4 antibodies are administered in conjunction (either
  • anti-CD4 antibodies are administered in conjunction with soluble TNF receptor, such as a TNF receptor/TgG fusion protein.
  • soluble TNF receptor such as a TNF receptor/TgG fusion protein.
  • cyclosporin is administered in conjunction with anti-TNF antibody.
  • the combination therapy can utilize any CD4+ T cell inhibiting agent in conjunction with any TNF
  • Combination therapy can also utilize inflammatory
  • the CD4+ T cell inhibiting agent and TNF antagonist can be administered together with a pharmaceutically acceptable vehicle; administration can be in the form of a Bingle dose, or a series of doses separated by intervals of days or weeks.
  • the benefits of combination therapy with CD4+ T cell inhibiting agents and TNF antagonists include improved results in comparison with the effects of treatment with each therapeutic modality separately.
  • lower dosages can be used to provide the same reduction of the immune and inflammatory response, thus increasing the therapeutic window between a therapeutic and a toxic effect.
  • Lower doses may also result in lower financial costs to the patient, and potentially fewer side effects.
  • Figure 1 contains a set of graphs, individually labelled as Fig. 1A and Fig. IB, from an experiment which illustrates the suppression of arthritis as assessed by clinical score (Fig. 1A) and pawswelling measurements (Fig. 1B) after the administration of 50 ⁇ g anti-TNF
  • Figure 2 contains a set of graphs, individually labelled as Fig. 2A, Fig. 2B, Fig. 2C, and Fig. 2D, from a second experiment which illustrates the potentiation of anti-CD4 with low dose (50 ⁇ g) anti-TNF or high dose (300 ⁇ g) anti-TNF on clinical score and pawswelling
  • Fig. 2A clinical score with low-dose anti-TNF
  • Fig. 2B clinical score with high-dose anti-TNF
  • Fig. 2C pawswelling with low-dose anti-TNF
  • Fig. 2D pawswelling with high-dose anti-TNF.
  • Figure 3 is a graph illustrating the suppression of arthritis as assessed by pawswelling measurements after the administration of 250 ⁇ g cyclosporin A, 50 ⁇ g anti-TNF antibody, and a combination of 250 ⁇ g cyclosporin A and 50 ⁇ g anti-TNF antibody to DBA/1 mice.
  • the present invention concerns the treatment of autoimmune or inflammatory diseases, such as rheumatoid arthritis, through the administration of a CD4+ T cell inhibiting agent in conjunction with a TNF antagonist.
  • the invention also encompasses the use of multiple CD4+ T cell inhibiting agents in conjunction with multiple TNF
  • CD4+ T cell inhibiting agent refers to an agent which blocks, diminishes, inhibits, or interferes with the activation of CD4+ T cells or the interaction of CD4+ T cells with antigen presenting cells (APC).
  • CD4+ T cell inhibiting agents include antibodies to T cells or to their receptors, such as anti-CD4, anti-CD28, anti-CD52 (e.g., CAMPATH-1H) and anti-IL-2R; antibodies to APC or to their receptors, such as anti-class II, anti-ICAM-1, anti-LFA-3, and anti-LFA-1; peptides and small molecules blocking the T cell/APC interaction, including those which block the HLA class II groove, or block signal transduction in T-cell activation, such as cyclosporins, particularly cyclosporin A, or FK-506; and antibodies to B cells including CD5+ B cells, such as CD19, 20, 21, 23 and BB/7 or Bl, ligands for CD28, B cells including CD5+ B cells are considered to be an important type of APC in disease processes (Plater-Zyberk, C. et al., Ann. N.Y. Acad. Sci. 651: 540-555 (1992)), and thus anti-B cell
  • TNF antagonist refers to an agent which blocks, diminishes, inhibits, or interferes with TNF activity, TNF synthesis, or TNF receptors, such as anti-TNF antibody; soluble TNF receptor (monomeric receptor and/or fusion proteins comprising the receptor, such as receptor/IgG fusion proteins, etc.); and other appropriate peptides or small molecules, such as
  • pentoxyfilline or other phosphodiesterase inhibitors and thalidomide.
  • Inflammatory mediators other than TNF antagonists can also be used instead of or in addition to TNF antagonists in the current invention.
  • In rheumatoid joint cell cultures Brennan et al. (Lancet 11, 244-247 (1989)) have shown that blocking TNF results in down-regulation of IL-1 production, and down-regulation of the pro-inflammatory cytokine GM-CSF (Haworth et al., E.J.I. 21:2575-2579
  • cytokine "networks'* or “hierarchies” also operate in vivo; rheumatoid arthritis patients treated with anti-TNF antibody reduced their serum IL-6 levels, as well as levels of IL-6 dependent acute phase proteins such as C reactive protein, in the weeks following treatment
  • Representative inflammatory mediators include agents which block, diminish, inhibit, or interfere with IL-1 activity, synthesis, or receptor signalling, such as anti-IL-1 antibody, soluble IL-1R, IL-1 receptor antagonist, or other appropriate peptides and small molecules; agents which block, diminish, inhibit, or interfere with IL-6 activity, synthesis, or receptor signalling, such as anti-IL-6 antibody, anti-gp 130, or other appropriate peptides and small molecules; modalities which block, diminish, inhibit, or interfere with the activity, synthesis, or receptor signalling of other inflammatory mediators, such as GM-CSF and members of the ch ⁇ mokin ⁇ (IL-8) family; and cytokine ⁇ with anti-inflammatory properties, such as IL-4, IL-10, and TGF/ ⁇ .
  • other anti-inflammatory agents such as the anti-rheumatic agent methotrexate, can be administered in conjunction with the CD4+ T cell inhibiting agent and/or the TNF antagonist.
  • anti-CD4 antibody is used in conjunction with anti-TNF antibody.
  • the term antibody is intended to encompass both polyclonal and monoclonal antibodies.
  • the term antibody is also intended to encompass mixtures of more than one antibody reactive with CD4 or with TNF (e.g., a cocktail of antibodies
  • antibody reactive with CD4 or with TNF.
  • the term antibody is further intended to encompass whole antibodies, biologically functional fragments thereof, bifunctional antibodies, and chimeric antibodies comprising portions from more than one species.
  • Biologically functional antibody fragments which can be used are those fragments sufficient for binding of the antibody fragment to CD4 or to TNF.
  • the chimeric antibodies can comprise portions derived from two different species (e.g., human constant region and murine variable or binding region).
  • the portions derived from two different species can be joined together chemically by conventional techniques or can be prepared as single contiguous proteins using genetic engineering techniques.
  • DNA encoding the proteins of both the light chain and heavy chain portions of the chimeric antibody can be expressed as contiguous proteins.
  • Monoclonal antibodies reactive with CD4 or with TNF can be produced using somatic cell hybridization
  • CD4 or of TNF can be used as the immunogen.
  • An animal is vaccinated with the immunogen to obtain anti-CD4 or anti-TNF antibody-producing spleen cells.
  • the species of animal immunized will vary depending on the species of monoclonal antibody desired.
  • the antibody producing cell is fused with an immortalizing cell (e.g., myeloma cell) to create a hybridoma capable of secreting anti-CD4 or anti-TNF antibodies.
  • the unfused residual antibody-producing cells and immortalizing cells are eliminated.
  • Hybridomas producing desired antibodies are selected using
  • Polyclonal antibodies can be prepared by immunizing an animal with a crude or purified protein or peptide comprising at least a portion of CD4 or of TNF. The animal is maintained under conditions whereby antibodies reactive with either CD4 or TNF are produced. Blood is collected from the animal upon reaching a desired titre of antibodies. The serum containing the polyclonal
  • antibodies (antisera) is separated from the other blood components.
  • the polyclonal antibody-containing serum can optionally be further separated into fractions of
  • antibodies e.g., IgG, IgM.
  • CD4 + T cell inhibiting agent and TNF antagonist can be administered by various routes , including
  • agents e.g., capsule, tablet, solution, emulsion
  • the form in which the agents are administered will depend at least in part on the route by which it is administered.
  • a therapeutically effective amount of the combination of anti-CD4 agent and anti-TNF agent is that amount necessary to significantly reduce or eliminate symptoms associated with a particular autoimmune or inflammatory disorder.
  • the therapeutically effective amount will be determined on an individual basis and will be based, at least in part, on consideration of particular agents used, the individual ' s size, the severity of symptoms to be treated, the result sought, etc.
  • the preferred therapeutically effective amount of anti-CD4 antibody administer ec in conjunction with anti-TNF antibody is in the range of 0.1 - 10 mg/kg/dose of each antibody.
  • the therapeutically effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
  • a maintenance amount of anti-CD4 agent, of anti-TNF agent, or of a combination of anti-CD4 agent and anti-TNF agent can be administered.
  • a maintenance amount is the amount of anti-CD4 agent, anti-TNF agent, or combination of anti-CD4 agent and anti-TNF agent necessary to maintain the reduction or elimination of symptoms achieved by the therapeutically effective dose.
  • the maintenance amount can be administered in the form of a single dose, or a series or doses separated by intervals of days or weeks. Like the therapeutically effective amount, the maintenance amount will be determined on an individual basis.
  • the combination therapy of the current invention is thus useful for the treatment of many autoimmune or inflammatory diseases of humans and of animals.
  • diseases for which the therapy is appropriate include rheumatoid arthritis (RA) and juvenile chronic arthritis (JCA).
  • other diseases and conditions for which combination therapy is appropriate include rheumatoid arthritis (RA) and juvenile chronic arthritis (JCA).
  • spondyloarthropathies such as ankylosing spondylitis, psoriatic arthritis, or arthritis associated with
  • vasculitidee such as
  • fibrosing alveolitis and other fibrotic lung diseases uveitis; multiple sclerosis; myasthenia gravis; hemolytic anemia; ⁇ cleroderma; graft versus host disease; allergy; and transplantation of kidneys, liver, heart, lungs, bone marrow, skin, or of other organs.
  • EXAMPLE 1 Treatment of Induced Arthritis in a Murine Model using Anti-CD4 Antibody and Anti-TNF Antibody
  • RA rheumatoid arthritis
  • anti-TNF antibody has beneficial effects (Williams, R.O. et al., PNAS 89:9784-9788 (1992); Elliott, M. J. et al., Arthritis & Rheumatism 36:1681-90 1992), and anti-CD4 antibody has minimal effect (Williams, R.O. fit al.. PNAS (in press) (1994); and Horneff, G. et al., Arthritis & Rheumatism 1991:34-129 (1992)).
  • the animal model serves as a good approximation to human disease.
  • mice Male DBA/1 mice were immunized intradermally at 8-12 weeks of age with 100 ⁇ g of bovine type II collagen emulsified in complete Freund's adjuvant (Difco).
  • mice were injected i.p. with anti-CD4; anti-TNF; anti-CD4 and anti-TNF; or isotype controls. Arthritis was monitored for clinical score and paw-swelling for 10 days. Antibody treatment was
  • mice Male DBA/1 mice were immunized intradermally at 8-12 weeks of age with 100 ⁇ g type II collagen emulsified in Freund's complete adjuvant (Difco Laboratories, East).
  • anti-TNF/anti-CD4 treatment also referred to herein as anti-CD4/TNF treatment
  • anti-CD4/TNF treatment produced a significant reduction in paw-swelling relative to anti-CD4 alone, and anti-TNF alone (P ⁇ 0.05).
  • mice After 10 days, the mice were sacrificed; the first limb that had shown clinical evidence of arthritis was removed from each mouse, formalin-fixed, decalcified, and wax-embedded before sectioning and staining with
  • proximal interphalangeal (PIP) joint of the middle digit was studied in a blind fashion for the presence or absence of erosions in either cartilage or bone (defined as demarcated defects in cartilage or bone filled with inflammatory tissue). The comparisons were made only between the same joints, and the arthritis was of
  • CD4+ T cells The possible persistence of CD4+ T cells in the joint despite virtual elimination of peripheral CD4+ T cells was next investigated by immunohistochemical analysis of sections from treated arthritic mice. Wax-embedded sections were de-waxed, trypsin digested, then incubated with anti-CD4 mAb (YTS 191.1.2/YTA 3.1.2). To confirm the T cell identity of the CD4+ T cells, sequential sections were stained with anti-Thy-1 mAb (YTS 154.7) (Cobbold, S.P. et al.. Nature 312:548-551 (1984)). Control sections were incubated with HRPNi ⁇ /l2a.
  • Detection of bound antibody was by alkaline phosphata ⁇ e/rat anti-alkaline phosphatase complex (APAAP; Dako, High Wycombe, UK) and fast red substrate as described (Deleuran, B.W. et al.. Arthritis t. Rheumatism 34:1125-1132 (1991)).
  • Small numbers of CD4+ T cells were detected in the joints, not only of mice given control mAb, but also of those treated with anti-CD4 (data not shown). Furthermore, within the small number of mice that were studied (four per treatment group), it was not possible to detect significantly reduced numbers of CD4+ T cells in the groups given anti-CD4 alone or anti-CD4 plus anti-TNF (data not shown).
  • Anti-CD4 treatment did not, therefore, eliminate CD4+ T cells from the joint.
  • Anti-collagen IgG Levels
  • Serum anti-collagen IgG levels were measured by enzyme-linked immunosorbent assay (ELISA). Microtitre plates were coated with bovine type II collagen (2 ⁇ g/ml), blocked, then incubated with test sera in serial dilution steps. Detection of bound IgG was by incubation with alkaline phosphata ⁇ e-conjugated goat anti-mouse IgG, followed by substrate (dinitrophenol phosphate). Optical densities were read at 405 nm. A reference sample, consisting of affinity-purified mouse anti-type II
  • IgM anti-TN4-19.12 levels on day 10 were compared. At this time, an IgG anti-TN3-19.12 response was not detected.
  • Microtitre plates were coated with TN3-19.12 (5 ⁇ g/ml), blocked, then incubated with serially diluted test sera. Bound IgM was detected by goat anti-mouse IgM-alkaline phosphatase conjugate, followed by substrate. The results demonstrated that anti-CD4 was highly effective in preventing the
  • mice Male DBA/1 mice were immunized intradermally with 100 ⁇ g of bovine type II collagen emulsified in complete Freund's adjuvant (Difco Laboratories, East Molsey, UK). The mean day of onset of arthritis was approximately one month after immunization. After the onset of clinically evident arthritis (erythema and/or swelling), mice were injected intraperitoneally with therapeutic agents.
  • mice were sacrificed and joints were processed for histology. Sera were collected for analysis on day 10.
  • Therapeutic agents were administered on day 1 (onset), day 4 and day 7. The therapeutic agents included TNF
  • mice were subjected to treatment with TNF receptor/IgG protein (2 ⁇ g) (18 mice), TNF receptor/IgG protein (20 ⁇ g) (18 mice), TNF receptor/IgG protein
  • mice 100 ⁇ g (12 mice), anti-TNF monoclonal antibody (mAb) (300 ⁇ g) (17 mice), methylprednisolone acetate (6 mice), an irrelevant human IgGl monoclonal antibody (mAb) (6 mice), or saline (control).
  • the TNF receptor/IgG fusion protein herein referred to as p55-sf2, (Butler et al., Cytokine (in press): (1994)), was provided by Centocor, Inc., Malvern PA; it is dimeric and consists of the human pS5 TNF receptor (extracellular domains) fused to a partial J sequence followed by the whole of the constant region of the human IgG1 heavy chain, itself associated with the constant region of a kappa light chain.
  • the anti-TNF antibody was TN3-19.12, a neutralizing hamster IgG1 anti-TNF ⁇ /,3 monoclonal antibody (Sheehan, K. C. et al., J. Immunology 142:3884-3893 (1989)), and was provided by R. Schreiber, Washington University Medical School (St. Louis, MO, USA), in conjunction with Celltech (Slough, UK). Neutralizing titres were defined as the
  • TNF ⁇ neutralizing agent concentration of TNF ⁇ neutralizing agent required to cause 50% inhibition of killing of WEHI 164 cells by trimeric recombinant murine TNF ⁇ ; the neutralizing titre of p55-sf2 was 0.6 ng/ml, compared with 62.0 ng/ml for anti-TNF mAb (TN3-19.12), using 60 pg/ml mouse TNF ⁇ .
  • Treatment with p55-sf2 resulted in a dose-dependent reduction in paw-swelling over the treatment period, with the doses of 20 ⁇ g and 100 ⁇ g giving statistically
  • mice given an irrelevant human IgGl mAb as a control did not show any deviation from the saline-treated group (data not shown), indicating that the therapeutic effects of p55-sf2 were attributable to the TNF receptor rather than the human IgGl constant region.
  • Similar reductions in paw-swelling were seen in mice given 300 ⁇ g of anti-TNF mAb as in those given 100 ⁇ g of p55-sf2, although anti-TNF mAb was
  • mice After 10 days, the mice were sacrificed; the first limb to show clinical evidence of arthritis was removed from each mouse, fixed, decalcified, wax-embedded, and sectioned and stained with haematoxylon and eosin.
  • Erosions were present in 92% and 100% of the PIP joints in the saline treated group and the control human IgG1 treated group, respectively. However, only 50%
  • Anti-collagen IgG levels on day 10 were measured by ELISA as described (Williams, R.O. et al., PNAS 89: 9784- 9788 (1992)). Microtitre plates were sensitized with type II collagen, then incubated with serially-diluted test sera. Bound IgG was detected using alkaline pho ⁇ phatase-conjugated goat anti-mouse IgG, followed by substrate (dinitrophenol phosphate). Optical densities were read at 405 nm. No differences between any of the treatment groups were detected (data not shown). this suggests that the therapeutic effect of p55-sf2 is not due to a
  • anti-CD4 monoclonal antibody mAb
  • mAb monoclonal antibody
  • a fourth group consisted of untreated control mice.
  • the cell-depleting anti-CD4 mAb (rat IgG2b) consisted of a 1:1 mixture of YTS 191.1.2 and YTA 3.1.2, provided by H.
  • the IgM/IgG responses to injected p55-sf2 were measured by ELISA at the end of the treatment period (day 10). Microtitre plates were coated with p55-sf2
  • mice is highly immunogenic in mice. This may account for the slightly greater efficacy of anti-TNF mAb in vivo described in Section B, above, despite the higher
  • Microtitre plates were coated with recombinant murine TNF- ⁇ (Genentech Inc., San Francisco, CA), blocked, then incubated with test sera. Goat anti-human IgG-alkaline phosphatase conjugate was then applied followed by
  • Quantitation was by reference to a sample of known concentration of p55-sf2.
  • mice given the fusion protein alone whereas in the mice given anti-CD4 mAb plus p55-sf2, the mean serum level of p55-sf2 was 12.3 ⁇ g/ml.
  • mice Male DBA/1 mice were immunized intradermally with 100 ⁇ g of bovine type II collagen emulsified in complete Freund's adjuvant (Difco Laboratories, East Molsey, UK). The mean day of onset of arthritis was approximately one month after immunization.
  • mice 11 mice each were subjected to treatment with one of the following therapies: 50 ⁇ g (2 mg/kg) L2 (the isotype control for anti-TNF antibody), intraperitoneally once every three days (days 1, 4 and 7); 250 ⁇ g (10 mg/kg) cyclosporin A intraperitoneally daily; 50 ⁇ g (2 mg/kg) anti-TNF mAb TN3-19.12, intraperitoneally once every three days (days 1, 4 and 7); or 250 ⁇ g cyclosporin A

Abstract

A method for treating autoimmune or inflammatory diseases, through the administration of a CD4+ T cell inhibiting agent, such as anti-CD4 antibody or cyclosporin A, in conjunction with or sequentially to a TNF antagonist, such as anti-TNF antibody or soluble TNF receptor, is disclosed. The method can be used to aid in therapy for humans and other mammals with a wide variety of autoimmune or inflammatory diseases.

Description

TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
Description
Background of the Invention
The nature of autoantigens responsible for autoimmune disorders is not known, nor is the action which triggers the autoimmune response. One popular theory involves the similarity of a viral protein to a self antigen, which results in autoreactlve T cells or B cells recognizing a self antiqen. Whereas B-lymphocytes produce antibodies, thymus-derived or "T-cells" are associated with cell-mediated immune functions. T-cells recognize antigens presented on the surface of cells and carry out their functions with these "antigen-presenting" cells.
Various markers have been used to define human T cell populations. CD4 is a non-polymorphic surface
glycoprotein receptor with partial sequence identity to immunoglobulins. CD4 receptors define distinct subsets of mature peripheral T cells. In general, CD4 T cells expressing helper or regulatory functions interact with B cells in immune responses, while T cells expressing the
CD8 surface antigen function as cytotoxic T cells and have regulatory effects on immune responses. Since T-cell receptors are the pathway through which stimuli augment or modulate T-cell responses, they present a potential target for immunological intervention.
Of the cellular interactions, that of CD4+ T cells with antigen presenting cells (APC) lies at the root of the immune response. Many aspects of the autoimmune response are essentially similar to that of normal immune responses. Thus CD4+ autoantigen reactive T cells are restimulated by APC expressing class II with autoantigen peptides in the binding groove. In certain human diseases the evidence that this occurs has been provided: in
Graves' disease of the thyroid, in vivo activated T cells are present in the glands that are removed for refractory disease, and many of these cells after cloning can be shown to recognize autologous thyrocytes (as APC) not extrinsically supplied with any antigen, or APC supplied with the thyroid specific antigens thyroid peroxidase or thyroglobulin (Londei, M. et al.. ,cience 228: 85-89
(1985); Dayan, CM. et al., Proc. Natl. Acad. Sci. USA 88: 7415-7419 (1991)). Similarly, in rheumatoid arthritis (RA), in vivo activated T cells recognizing collagen type II have been isolated from joints of an RA patient in three consecutive operations during the course of three years (Londei, M. et al., Proc. Natl. Acad. Sci. 86: 636-640 (1989)). In other human diseases displaying
autoimmune characteristics, CD4+ T cells from the blood have been cloned, including CD4+ T cells recognizing the acetylcholine receptor in myasthenia gravis (Hohlfeld, R. et al. , Nature 310: 224-246 (1984)); myelin basic protein in multiple sclerosis (Hafler, D.A. et al.. J. Immunol. 139: 68-72 (1987)); or islet cell membranes in insulin dependent diabetes mellitus (De Berardinis, P. et al.,
Lancet II: 823-824 (1988); Kontiainen, S. et al.,
Autoimmunity 8: 193-197 (1991)).
Factors other than CD4 also influence cellular immune response. The cytokine tumor necrosis factor-α (TNFα;
also termed cachectin) has multiple effects on
inflammation, tissue damage, immune response and cell trafficking into lesions, and thus plays a role in the pathogenesis of inflammatory joint diseases, including rheumatoid arthritis (Brennan, F.M. et a1.. Lancet 11, 244-247 (1989); Feldmann, M. et al., Ann. Rheumatic Dis.
51: 480-486 (1990)). TNFα is a protein secreted primarily by monocytes and macrophages in response to endotoxin or other stimuli as a soluble homotrimer of 17 kD protein Subunits (Smith, R.A. et al., J. Biol. Chem. 262: 6951-6954 (1987)). A membrane-bound 26 kD precursor form of TNF has also been described (Kriegler, M. et al., Cell 53: 45-53 (1988). The expression of the gene encoding TNFα is not limited to cells of the monocyte/macrophage family: TNF is also produced by CD4+ and CD8+ peripheral blood T lymphocytes, and by various cultured T and B cell lines (Cuturi, M.C. et al., J. EXP. Med. 165: (1581 (1987);
Sung, S.-S.J. et al., J. Exp. Med. 168: 1539 (1988);
Turner, M. et al., Eur. J. Immunol. 17: 1807-1814 (1987)). Recent evidence implicates TNF in the autoimmune
pathologies and graft versus host pathology (Piguet, P.-F. et al., J. EXP. Med. 166: 1280 (1987).
Because of the multiple factors involved in
autoimmune and inflammatory disorders, a great need exists for better therapies for autoimmune and inflammatory diseases.
Summary of the Invention
The current invention pertains to the discovery that combination therapy, involving the use of a CD4+ T cell inhibiting agent in conjunction with a TNF antagonist, produces markedly superior results than the use of each agent alone in the treatment of autoimmune or inflammatory disease, particularly in rheumatoid arthritis. CD4+ T cell inhibiting agents include agents which block,
diminish, inhibit, or interfere with the activation of CD4+ T cells or the interaction of CD4+ T cells with antigen presenting cells (APC), such as antibodies to T cells or to their receptors; antibodies to APC or to their receptors; and other appropriate peptides or small
molecules. TNF antagonists include agents which block, diminish, inhibit, or interfere with TNF activity, TNF receptors, or TNF synthesis, such as anti-TNF antibodies; soluble TNF receptors; and other appropriate peptides or small molecules.
In one embodiment of the current invention, anti-CD4 antibodies are administered in conjunction (either
simultaneously or sequentially) with anti-TNF antibodies. In another embodiment of the current invention, anti-CD4 antibodies are administered in conjunction with soluble TNF receptor, such as a TNF receptor/TgG fusion protein. In a third embodiment of the current invention,
cyclosporin is administered in conjunction with anti-TNF antibody. The combination therapy can utilize any CD4+ T cell inhibiting agent in conjunction with any TNF
antagonist, including multiple CD4+ T cell inhibiting agents in conjunction with multiple TNF antagonists.
Combination therapy can also utilize inflammatory
mediators other than TNF antagonists, in conjunction with CD4+ T cell inhibiting agents.
The CD4+ T cell inhibiting agent and TNF antagonist can be administered together with a pharmaceutically acceptable vehicle; administration can be in the form of a Bingle dose, or a series of doses separated by intervals of days or weeks.
The benefits of combination therapy with CD4+ T cell inhibiting agents and TNF antagonists include improved results in comparison with the effects of treatment with each therapeutic modality separately. In addition, lower dosages can be used to provide the same reduction of the immune and inflammatory response, thus increasing the therapeutic window between a therapeutic and a toxic effect. Lower doses may also result in lower financial costs to the patient, and potentially fewer side effects. Brief Description of the Figures
Figure 1 contains a set of graphs, individually labelled as Fig. 1A and Fig. IB, from an experiment which illustrates the suppression of arthritis as assessed by clinical score (Fig. 1A) and pawswelling measurements (Fig. 1B) after the administration of 50 μg anti-TNF
(hamster TN3.19.2) and 200 μg anti-CD4 to DBA/1 male mice. Open squares = control; diamonds = anti-CD4; triangles = anti-TNF; closed squares = anti-CD4/anti-TNF.
Figure 2 contains a set of graphs, individually labelled as Fig. 2A, Fig. 2B, Fig. 2C, and Fig. 2D, from a second experiment which illustrates the potentiation of anti-CD4 with low dose (50 μg) anti-TNF or high dose (300 μg) anti-TNF on clinical score and pawswelling
measurements. Fig. 2A: clinical score with low-dose anti-TNF; Fig. 2B: clinical score with high-dose anti-TNF; Fig. 2C: pawswelling with low-dose anti-TNF; Fig. 2D: pawswelling with high-dose anti-TNF. Open squares = control; diamonds = anti-CD4; triangles = anti-TNF; closed squares = anti-CD4/anti-TNF.
Figure 3 is a graph illustrating the suppression of arthritis as assessed by pawswelling measurements after the administration of 250 μg cyclosporin A, 50 μg anti-TNF antibody, and a combination of 250 μg cyclosporin A and 50 μg anti-TNF antibody to DBA/1 mice. Open squares = control; diamonds = cyclosporin A; triangles «= anti-TNF; closed squares = cyclosporin A/anti-TNF.
Detailed Description of the Invention
The present invention concerns the treatment of autoimmune or inflammatory diseases, such as rheumatoid arthritis, through the administration of a CD4+ T cell inhibiting agent in conjunction with a TNF antagonist. The invention also encompasses the use of multiple CD4+ T cell inhibiting agents in conjunction with multiple TNF
antagonists. The term "CD4+ T cell inhibiting agent", as used herein, refers to an agent which blocks, diminishes, inhibits, or interferes with the activation of CD4+ T cells or the interaction of CD4+ T cells with antigen presenting cells (APC). CD4+ T cell inhibiting agents include antibodies to T cells or to their receptors, such as anti-CD4, anti-CD28, anti-CD52 (e.g., CAMPATH-1H) and anti-IL-2R; antibodies to APC or to their receptors, such as anti-class II, anti-ICAM-1, anti-LFA-3, and anti-LFA-1; peptides and small molecules blocking the T cell/APC interaction, including those which block the HLA class II groove, or block signal transduction in T-cell activation, such as cyclosporins, particularly cyclosporin A, or FK-506; and antibodies to B cells including CD5+ B cells, such as CD19, 20, 21, 23 and BB/7 or Bl, ligands for CD28, B cells including CD5+ B cells are considered to be an important type of APC in disease processes (Plater-Zyberk, C. et al., Ann. N.Y. Acad. Sci. 651: 540-555 (1992)), and thus anti-B cell antibodies can be particularly useful in the current invention.
The term "TNF antagonist", as used herein, refers to an agent which blocks, diminishes, inhibits, or interferes with TNF activity, TNF synthesis, or TNF receptors, such as anti-TNF antibody; soluble TNF receptor (monomeric receptor and/or fusion proteins comprising the receptor, such as receptor/IgG fusion proteins, etc.); and other appropriate peptides or small molecules, such as
pentoxyfilline or other phosphodiesterase inhibitors, and thalidomide.
Inflammatory mediators other than TNF antagonists can also be used instead of or in addition to TNF antagonists in the current invention. In rheumatoid joint cell cultures, Brennan et al. (Lancet 11, 244-247 (1989)) have shown that blocking TNF results in down-regulation of IL-1 production, and down-regulation of the pro-inflammatory cytokine GM-CSF (Haworth et al., E.J.I. 21:2575-2579
(1991); Brennan≤£ al.. in preparation). Unpublished data indicates that anti-TNF also blocks IL-6 production.
These cytokine "networks'* or "hierarchies" also operate in vivo; rheumatoid arthritis patients treated with anti-TNF antibody reduced their serum IL-6 levels, as well as levels of IL-6 dependent acute phase proteins such as C reactive protein, in the weeks following treatment
(Elliott, M.J. et al., Arthritis & Rheumatism 36:1681-1690 (1993)). Since the pro-inflammatory mediators TNF, IL-1, GM-CSF, IL-6 and IL-8 are part of the same network or hierarchy, blocking any of these could have comparable effects and thus can be used as the inflammatory mediators of the current invention. Representative inflammatory mediators include agents which block, diminish, inhibit, or interfere with IL-1 activity, synthesis, or receptor signalling, such as anti-IL-1 antibody, soluble IL-1R, IL-1 receptor antagonist, or other appropriate peptides and small molecules; agents which block, diminish, inhibit, or interfere with IL-6 activity, synthesis, or receptor signalling, such as anti-IL-6 antibody, anti-gp 130, or other appropriate peptides and small molecules; modalities which block, diminish, inhibit, or interfere with the activity, synthesis, or receptor signalling of other inflammatory mediators, such as GM-CSF and members of the chβmokinβ (IL-8) family; and cytokineε with anti-inflammatory properties, such as IL-4, IL-10, and TGF/β. In addition, other anti-inflammatory agents, such as the anti-rheumatic agent methotrexate, can be administered in conjunction with the CD4+ T cell inhibiting agent and/or the TNF antagonist.
In one embodiment of the current invention, anti-CD4 antibody is used in conjunction with anti-TNF antibody. The term antibody is intended to encompass both polyclonal and monoclonal antibodies. The term antibody is also intended to encompass mixtures of more than one antibody reactive with CD4 or with TNF (e.g., a cocktail of
different types of monoclonal antibodies reactive with CD4 or with TNF). The term antibody is further intended to encompass whole antibodies, biologically functional fragments thereof, bifunctional antibodies, and chimeric antibodies comprising portions from more than one species. Biologically functional antibody fragments which can be used are those fragments sufficient for binding of the antibody fragment to CD4 or to TNF.
The chimeric antibodies can comprise portions derived from two different species (e.g., human constant region and murine variable or binding region). The portions derived from two different species can be joined together chemically by conventional techniques or can be prepared as single contiguous proteins using genetic engineering techniques. DNA encoding the proteins of both the light chain and heavy chain portions of the chimeric antibody can be expressed as contiguous proteins.
Monoclonal antibodies reactive with CD4 or with TNF can be produced using somatic cell hybridization
techniques (Kohler and Milstein, Nature 256: 495-497
(1975)) or other techniques. In a typical hybridization procedure, a crude or purified protein or peptide
comprising at least a portion of CD4 or of TNF can be used as the immunogen. An animal is vaccinated with the immunogen to obtain anti-CD4 or anti-TNF antibody-producing spleen cells. The species of animal immunized will vary depending on the species of monoclonal antibody desired. The antibody producing cell is fused with an immortalizing cell (e.g., myeloma cell) to create a hybridoma capable of secreting anti-CD4 or anti-TNF antibodies. The unfused residual antibody-producing cells and immortalizing cells are eliminated. Hybridomas producing desired antibodies are selected using
conventional techniques and the selected hybridomas are cloned and cultured.
Polyclonal antibodies can be prepared by immunizing an animal with a crude or purified protein or peptide comprising at least a portion of CD4 or of TNF. The animal is maintained under conditions whereby antibodies reactive with either CD4 or TNF are produced. Blood is collected from the animal upon reaching a desired titre of antibodies. The serum containing the polyclonal
antibodies (antisera) is separated from the other blood components. The polyclonal antibody-containing serum can optionally be further separated into fractions of
particular types of antibodies (e.g., IgG, IgM) .
Antibodies specific for CD4 have been used in
treatment of a wide range of both experimentally-induced and spontaneously-occurring autoimmune diseases. A more detailed description of anti-CD4 antibodies and their use in treatment of disease is contained in the following references, the teachings of which are hence incorporated by reference: U.S. Application NO. 07/867,100, filed June 25, 1992; Grayheb, J. et al., J. of Autoimmunity 2:627-642 (1989); Ranges, G.E. et al. J. Exp. Med. 162: 1105-1110 (1985); Horn, J.T. et al., Eur. J. Immunol. 18: 881-888 (1988); Wooley, P.H. et al., J. Immunol. 134: 2366-2374 (1985); Cooper, S.M. et al., J. Immunol. 141; 1958-1962 (1988); Van den Broek, M.F. et al., Eur. J. Immunol. 22: 57-61 (1992); Wofsy, D. et al.. J. Immunol. 134: 852-857 (1985); Wofsy, D. et al., J. Immunol. 136: 4554-4560
(1986); Ermak, T.J. et al., Laboratory Investigation 61: 447-456 (1989); Reiter, C. et al., 34:525-532 (1991);
Herzog, C. et al., J. Autoimmun. 2:627 (1989); Ouyang, Q. et al., Dig. Dis. Sci. 33:1528-1536 (1988); Herzog, C . et al., Lancet, p. 1461 (December 19, 1987); Emmrich, J. et al., Lancet 338:570-571 (August 31, 1991).
A more detailed description of anti-TNF antibodies and their use in treatment of disease is contained in the following references, the teachings of which are hence incorporated by reference: U.S. Application No.
07/943,852, filed September 11, 1992; Rubin et al., (EPO Patent Publication 0218868, April 22, 1987); Yone et al., (EPO Patent Publication 0288088, October 26, 1988); Liang, C.-M. et al., Biochem. Biophys. Res. Comm. 137:847-854 (1986) ; Meager , A. et al . , Hybridoma 6 : 305-311 ( 1987) ;
Fendly et al., Hybridoma 6:359-369 (1987); Bringman, T.S. et al., Hybridoma 6:489-507 (1987); Bringman T.S. et al., Hybridoτna 6:489-507 (1987); Hirai, M. et al., J. Immunol. Meth. 96:57-62 (1987); Moller, A. et al., Cytokine 2:162-169 (1990); Mathison, J.C. et al., J. Clin. Invest.
81:1925-1937 (1988); Beutler, B. et al., Science 229:869-871 (1985); Tracey, K.J. et al., Nature 330:662-664
(1987) Shimamoto, Y. et al., Immunol. Lett. 17:311-318 (1988) Silva, A.T. et al., J. Infect. Dis. 162: 421-427 (1990) Opal, S.M. et al., J. Infect. Dis. 161:1148-1152 (1990) Hinshaw, L.B. et al., Circ. Shock 30:279-292
(1990) Lancet 342:173-174 (1993); Williams, R.O. et al.,
Proc. Natl. Acad. Sci. USA 89:9784-9788 (1992) The CD4 + T cell inhibiting agent and TNF antagonist can be administered by various routes , including
subcutaneously, intravenously, intramuscularly, topically, orally, rectally, nasally, buccally, vaginally, by
inhalation spray, or via an implanted reservoir in dosage formulations containing conventional non-toxic
pharmaceutical ly-acceptable carriers, adjuvants and vehicles . The form in which the agents are administered (e . g. , capsule, tablet, solution, emulsion) will depend at least in part on the route by which it is administered.
A therapeutically effective amount of the combination of anti-CD4 agent and anti-TNF agent is that amount necessary to significantly reduce or eliminate symptoms associated with a particular autoimmune or inflammatory disorder. The therapeutically effective amount will be determined on an individual basis and will be based, at least in part, on consideration of particular agents used, the individual ' s size, the severity of symptoms to be treated, the result sought, etc. In one embodiment, for example, the preferred therapeutically effective amount of anti-CD4 antibody administer ec in conjunction with anti-TNF antibody is in the range of 0.1 - 10 mg/kg/dose of each antibody. Thus , the therapeutically effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
The therapeutically effective amount can be
administered in the form of a single dose, or a series of doses separated by intervals of days or weeks . Once the therapeutically effective amount has been administered, a maintenance amount of anti-CD4 agent, of anti-TNF agent, or of a combination of anti-CD4 agent and anti-TNF agent can be administered. A maintenance amount is the amount of anti-CD4 agent, anti-TNF agent, or combination of anti-CD4 agent and anti-TNF agent necessary to maintain the reduction or elimination of symptoms achieved by the therapeutically effective dose. The maintenance amount can be administered in the form of a single dose, or a series or doses separated by intervals of days or weeks. Like the therapeutically effective amount, the maintenance amount will be determined on an individual basis.
The combination therapy of the current invention is thus useful for the treatment of many autoimmune or inflammatory diseases of humans and of animals. In humans, diseases for which the therapy is appropriate include rheumatoid arthritis (RA) and juvenile chronic arthritis (JCA). other diseases and conditions for which combination therapy is appropriate include
spondyloarthropathies, such as ankylosing spondylitis, psoriatic arthritis, or arthritis associated with
inflammatory bowel disease; vasculitidee, such as
polyarteritis nodosa, Wegener's granulomatosis, giant cell arteritis, Henoch-Schoenlein purpura, and microscopic vasculitiε of the kidneys; Sjogren's syndrome; systemic lupus erythematosus; inflammatory bowel disease, including Crohn'ε disease and ulcerative colitis; chronic active hepatitis; primary biliary cirrhosis; cryptogenic
fibrosing alveolitis and other fibrotic lung diseases; uveitis; multiple sclerosis; myasthenia gravis; hemolytic anemia; εcleroderma; graft versus host disease; allergy; and transplantation of kidneys, liver, heart, lungs, bone marrow, skin, or of other organs.
The invention is further and more specif ically illustrated by the following Examples. EXAMPLE 1 Treatment of Induced Arthritis in a Murine Model using Anti-CD4 Antibody and Anti-TNF Antibody The murine model of collagen type II induced
arthritis has similarities to rheumatoid arthritis (RA) in its marked MHC class II predisposition, as well as in histology, immunohistology, and erosions of cartilage and bone. Furthermore, there is a good correlation of
therapeutic response with human rheumatoid arthritis. For example, in both diseases anti-TNF antibody has beneficial effects (Williams, R.O. et al., PNAS 89:9784-9788 (1992); Elliott, M. J. et al., Arthritis & Rheumatism 36:1681-90 1992), and anti-CD4 antibody has minimal effect (Williams, R.O. fit al.. PNAS (in press) (1994); and Horneff, G. et al., Arthritis & Rheumatism 1991:34-129 (1992)). Thus the animal model serves as a good approximation to human disease.
The model of rheumatoid arthritis used herein is described by Williams, R.O. et al., (PNAS, 89:9784-9788 (1992), i.e., the collagen type II induced arthritis in the DBA/1 mouse. Type II collagen was purified from bovine articular cartilage by limited pepsin
solubilization and salt fractionation as described by Miller (Biochemistry 11:4903-4909 (1972)). A. Study 1
Male DBA/1 mice were immunized intradermally at 8-12 weeks of age with 100 μg of bovine type II collagen emulsified in complete Freund's adjuvant (Difco
Laboratories, East Molsey, UK), and 21 days later with 100 μg of collagen intra-peritoneally (i.p.). Immediately after the onset of clinically evident arthritis (redness and/or swelling in one or more limbs), which was about 35 days after the initial injection, mice were injected i.p. with anti-CD4; anti-TNF; anti-CD4 and anti-TNF; or isotype controls. Arthritis was monitored for clinical score and paw-swelling for 10 days. Antibody treatment was
administered on day 1 (onset), day 4 and
day 7.
Clinical Score and Pawswelling
Two experiments were completed, assessing clinical score and pawswelling. In each, 200 μg of anti-CD4 were used per injection (rat YTS 191 and YTA 3.1) was used. Clinical score was assessed on the following scale: 0 = normal; 1 = slight swelling and/or erythema; 2 =
ronounced edematoma swelling; and 3 = joint rigidity. Each limb was graded, giving a maximum score of 12 per mouse. Pawswelling was monitored by measuring the
thicJcness of each affected hind paw with calipers. The results were expressed as the percentage increment in paw width relative to the paw width before the onset of arthritis.
In the first experiment, a single dose of 50 μg per injection of anti-TNF (hamster TN3.19.2) was administered to each of five mice per group. There was no significant effect of anti-CD4 or anti-TNF (TN3.19 given 3 times at 50 μg/mouse). Hence the benefit of combination therapy, in both clinical score and footpad swelling, is readily seen (see Figures 1A, IB) .
In the second experiment, either 50 μg or 300 μg of anti-TNF were administered to each of 7 mice per group. Both anti-CD4 and anti-TNF at low (50 μg) concentration had some effect, and benefit of combination therapy of these two concentrations was noted in pawswelling, not in clinical score. However, if anti-TNF was injected at 300 μg/mouse, the benefit of combination therapy with anti-CD4 was seen in both clinical score and more clearly in paw-swelling (see Figures 2A, 2B, 2C, 2D).
The results of the experiments indicate that there is a clear benefit to combination therapy with anti-TNF and anti-CD4 antibodies, as measured by clinical score and foot pad swelling.
B. Study 2
Male DBA/1 mice were immunized intradermally at 8-12 weeks of age with 100 μg type II collagen emulsified in Freund's complete adjuvant (Difco Laboratories, East
Molsey, UK). Day one of arthritis was considered to be the day that erythema and/or swelling was first observed in one or more limbs. Arthritis became clinically evident around 30 days after immunization with type II collagen. For each mouse, treatment was started on the first day that arthritis was observed and continued over a 10 day period, after which the mice were sacrificed and joints were processed for histology. Monoclonal antibody (mAb) treatment was administered on days 1, 4, and 7. For anti-TNF antibody, TN3-19.12, a neutralizing hamster IgG1 anti-TNFα/β monoclonal antibody (mAb), was used (Sheehan, K. C. et al., J. Immunology 142:3884-3893 (1989)). The isotype control was L2. The anti-TNF antibody and the isotype control were provided by R. Schreiber, Washington
University Medical School (St. Louis, MO, USA), in
conjunction with Celltech (Slough, UK). The cell-depleting anti-CD4 monoclonal antibody (rat IgG2b)
consisted of a 1:1 mixture of YTS 191.1.2 and YTA 3.1.2, provided by H. Waldmann (University of Cambridge, UK)
(Galfre, G. et al., Nature 277: 131-133 (1979); Cobbold, S.P. et al., Nature 312: 548-551 (1984); Qin, S. et al., European J. Immunology 17:1159-1165 (1987)).
Paw-Swelling
First, a sub-optimal dose of 50 μg of anti-TNF alone was compared with the same dose given together with 200 μg of anti-CD4. To verify the results, two separate but identical experiments were carried out (11-12 mice/group and 7-8 mice/group, respectively). Neither anti-CD4 alone nor sub-optimal anti-TNF alone were able to significantly reduce paw-swelling (data not shown). However, treatment with anti-TNF and anti-CD4 resulted in a consistently and statistically significant reduction in paw-swelling relative to the group given control mAb (P < 0.001).
Furthermore, in both experiments, combined anti-TNF/anti-CD4 treatment (also referred to herein as anti-CD4/TNF treatment) produced a significant reduction in paw-swelling relative to anti-CD4 alone, and anti-TNF alone (P < 0.05).
Next, an optimal dose of anti-TNF (300 μg) alone was compared in two separate but identical experiments (7-7 mice/group and 6-7 mice/group, respectively) with the same dose given in combination with anti-CD4. As before, the combined anti-TNF/anti-CD4 treatment resulted in a
significant reduction in paw-swelling compared to
treatment with the control mAb (P <0.005; data not shown). In the first experiment, paw-swelling was also
significantly reduced in the combined anti-CD4/anti-TNF treated group relative to the groups given anti-CD4 alone or anti-TNF alone (P < 0.05). Some reduction in paw-swelling was observed in mice given either anti-TNF alone or anti-CD4 alone although the differences were not significant, possibly because of the small group sizes (6 per group). In the second experiment, combined anti-CD4/anti-TNF gave significantly reduced paw-ewelling compared to anti-CD4 alone (P < 0.05) but not compared to anti-TNF alone since anti-TNF itself caused a significant reduction in paw-swelling, as expected from previous work (Williams, R.O. et al., PNAS 89: 9784-9788 (1992)). In the experiments, the reduction in paw-swelling
attributable to anti-TNF alone was 23% and 33%,
respectively. Thus, the reduction in paw-swelling
attributable to anti-TNF treatment was broadly comparable with our previously published findings in which treatment with TN3-119.12 (300 μg/mouse) resulted in a mean
reduction in paw-swelling over the treatment period of around 34% relative to controls (Williams, R.O. et al., PNAS 89 : 9784 -9788 ( 1992 ) ) . Li mb Involvement
In collagen-induced arthritis, as in RA.it is usual for additional limbs to become involved after the initial appearance of clinical disease and new limb involvement is an important indicator of the progression of the disease. To determine the effect of anti-CD4/anti-TNF treatment on new limb involvement, the number of limbs with clinically detectable arthritis at the end of the 10 day treatment period was compared with the number of arthritis limbs before treatment. In mice given the control mAb there was an increase in limb involvement over the 10 day period of approximately 50% The results from the two experiments were pooled, and are shown in Table 1.
Figure imgf000020_0001
There was some reduction in new limb involvement in the groups given anti-CD4 alone and sub-optimal anti-TNF alon although the differences were not significant. In the group given optimal anti-TNF the increase in limb involvement was less than 10% (P < 0.05). More striking, however, was the almost complete absence of new limb involvement in the groups given combined anti-CD4/anti-TNF. Thus, the increase in new limb involvement was only 3% in mice given anti-CD4 plus suboptimal anti-TNF
(P < 0.05) and 0% in mice given anti-CD4 plus optimal anti-TNF (P < 0.005).
Histology
After 10 days, the mice were sacrificed; the first limb that had shown clinical evidence of arthritis was removed from each mouse, formalin-fixed, decalcified, and wax-embedded before sectioning and staining with
haematcxylon and eosin. A sagittal section of the
proximal interphalangeal (PIP) joint of the middle digit was studied in a blind fashion for the presence or absence of erosions in either cartilage or bone (defined as demarcated defects in cartilage or bone filled with inflammatory tissue). The comparisons were made only between the same joints, and the arthritis was of
identical duration. Erosions were observed in almost 100% of the PIP joints from the control groups and in
approximately 70-80% of the joints given either anti-CD4 alone or sub-optimal anti-TNF alone. The results of the two experiments were pooled, and are shown in Table 2.
Figure imgf000022_0001
An optimal dose of anti-TNF alone significantly reduced pathology, as reported previously (Williams, R.O. et al., PNAS 89: 9784-9788 (1992)). Thus, in the mice given optimal anti-TNF alone the proportion of joints showing erosive changes was reduced to 54% (P < 0.001) whereas in the groups given anti-CD4 plus either sub-optimal or optimal anti-TNF,only 22% (P < 0.01) and 31%
(P > 0.01) of the joints, respectively, were eroded.
Thus, 300 μg of anti-TNF alone gave a degree of protection against joint erosion but combined anti-CD4/anti-TNF provided significantly greater protection. Depletion of CD4+ T Cells
The extent to which anti-cr4 treatment depleted peripheral CD4+ T cells was determined by flow cytometry. To enumerate the proportion of CD4+ lymphocytes in
disassociated spleen populations or peripheral blood, cells were incubated with phycoerythrin-conjugated anti-CD4 (Becton Dickinson, Oxford, UK), then analyzed by flow cytometry (FACScan, Becton Dickinson) with scatter gates set on the lymphocyte fraction. Anti-CD4 treatment resulted in 98% (± 1%) depletion of CD4+ T cells in the spleen and 96% (± 3%) depletion of CD4+ T cells in the blood.
Imraunohistochemistry
The possible persistence of CD4+ T cells in the joint despite virtual elimination of peripheral CD4+ T cells was next investigated by immunohistochemical analysis of sections from treated arthritic mice. Wax-embedded sections were de-waxed, trypsin digested, then incubated with anti-CD4 mAb (YTS 191.1.2/YTA 3.1.2). To confirm the T cell identity of the CD4+ T cells, sequential sections were stained with anti-Thy-1 mAb (YTS 154.7) (Cobbold, S.P. et al.. Nature 312:548-551 (1984)). Control sections were incubated with HRPNiι/l2a. Detection of bound antibody was by alkaline phosphataεe/rat anti-alkaline phosphatase complex (APAAP; Dako, High Wycombe, UK) and fast red substrate as described (Deleuran, B.W. et al.. Arthritis t. Rheumatism 34:1125-1132 (1991)). Small numbers of CD4+ T cells were detected in the joints, not only of mice given control mAb, but also of those treated with anti-CD4 (data not shown). Furthermore, within the small number of mice that were studied (four per treatment group), it was not possible to detect significantly reduced numbers of CD4+ T cells in the groups given anti-CD4 alone or anti-CD4 plus anti-TNF (data not shown).
Anti-CD4 treatment did not, therefore, eliminate CD4+ T cells from the joint. Anti-collagen IgG Levels
Serum anti-collagen IgG levels were measured by enzyme-linked immunosorbent assay (ELISA). Microtitre plates were coated with bovine type II collagen (2 μg/ml), blocked, then incubated with test sera in serial dilution steps. Detection of bound IgG was by incubation with alkaline phosphataεe-conjugated goat anti-mouse IgG, followed by substrate (dinitrophenol phosphate). Optical densities were read at 405 nm. A reference sample, consisting of affinity-purified mouse anti-type II
collagen antibody, was included on each plate. Results are shown in Table 3.
Figure imgf000025_0001
Serum levels of anti-type II collagen IgG were not
significantly altered within the 10 day treatment period by anti-CD4 alone, anti-TNF alone, or anti-CD4 plus anti-TNF.
Anti-Globulin Response
To find out whether anti-CD4 treatment prevented a neutralizing anti-globulin response against the anti-TNF mAb, IgM anti-TN4-19.12 levels on day 10, as measured by ELISA, were compared. At this time, an IgG anti-TN3-19.12 response was not detected. Microtitre plates were coated with TN3-19.12 (5 μg/ml), blocked, then incubated with serially diluted test sera. Bound IgM was detected by goat anti-mouse IgM-alkaline phosphatase conjugate, followed by substrate. The results demonstrated that anti-CD4 was highly effective in preventing the
development of an anti-TN3-19.12 antibody response (Table 4) . Next, to determine whether anti-CD4 treatment led to increased levels of circulating anti-TNF-α (by reducing the antibody response to the hamster anti-TNF), an ELISA was carried out in which recombinant murine TNF-α was used to detect free TN3-19.12 in the sera of mice on day 10 of the experiment. Microtitre plates were coated with recombinant murine TNF-α, blocked, then incubated with test sera. Goat anti-hamster IgG-alkaline phosphatase conjugate (adsorbed against murine IgG) was then applied, followed by substrate. Quantitation was by reference to a sample of known concentration of TN3-19.12. Results are shown in Table 4.
Figure imgf000026_0001
Levels of TN3-19.12 were slightly elevated in the groups given anti-CD4 plus anti-TNF compared to anti-TNF alone, although the differences were not significantly different. EXAMPLE 2. Treatment of Induced Arthritis in a Murine Mode) using TNF Receptor/IαG Fusion Protein with Anti- CP4 Antibody
The murine model of collagen type II induced
arthritis, described above, was used to investigate the efficacy of a human p55 TNF receptor/IgG fusion protein, in conjunction with anti-CD4 monoclonal antibody (mAb), for its ability to modulate the severity of joint disease in collagen-induced arthritis. First, a comparison was made between the efficacy of TNF receptor/IgG fusion protein treatment, anti-TNF mAb treatment, and high dose corticosteroid therapy. Subsequently, therapy with TNF receptor/IgG fusion protein in conjunction with anti-CD4 antibody was investigated. A. Experimental Procedure
Male DBA/1 mice were immunized intradermally with 100 μg of bovine type II collagen emulsified in complete Freund's adjuvant (Difco Laboratories, East Molsey, UK). The mean day of onset of arthritis was approximately one month after immunization. After the onset of clinically evident arthritis (erythema and/or swelling), mice were injected intraperitoneally with therapeutic agents.
Arthritis was monitored for clinical score and paw
swelling (measured with calipers) for 10 days, after which the mice were sacrificed and joints were processed for histology. Sera were collected for analysis on day 10. Therapeutic agents were administered on day 1 (onset), day 4 and day 7. The therapeutic agents included TNF
receptor/IgG fusion protein (p55-sf2), anti-TNF antibody, anti-CD4 antibody, and methylprednisolone acetate. B. Comparison of Treatment with TNF Receptor/IgG Fusion Protein, Anti-TNF Antibody, or Methylpredπisolone Acetate
Using the Experimental Procedure described above, groups of mice were subjected to treatment with TNF receptor/IgG protein (2 μg) (18 mice), TNF receptor/IgG protein (20 μg) (18 mice), TNF receptor/IgG protein
(100 μg) (12 mice), anti-TNF monoclonal antibody (mAb) (300 μg) (17 mice), methylprednisolone acetate (6 mice), an irrelevant human IgGl monoclonal antibody (mAb) (6 mice), or saline (control). The TNF receptor/IgG fusion protein, herein referred to as p55-sf2, (Butler et al., Cytokine (in press): (1994)), was provided by Centocor, Inc., Malvern PA; it is dimeric and consists of the human pS5 TNF receptor (extracellular domains) fused to a partial J sequence followed by the whole of the constant region of the human IgG1 heavy chain, itself associated with the constant region of a kappa light chain. The anti-TNF antibody was TN3-19.12, a neutralizing hamster IgG1 anti-TNFα/,3 monoclonal antibody (Sheehan, K. C. et al., J. Immunology 142:3884-3893 (1989)), and was provided by R. Schreiber, Washington University Medical School (St. Louis, MO, USA), in conjunction with Celltech (Slough, UK). Neutralizing titres were defined as the
concentration of TNFα neutralizing agent required to cause 50% inhibition of killing of WEHI 164 cells by trimeric recombinant murine TNFα; the neutralizing titre of p55-sf2 was 0.6 ng/ml, compared with 62.0 ng/ml for anti-TNF mAb (TN3-19.12), using 60 pg/ml mouse TNFα. The
corticosteroid, methyl-prednisolone acetate (Upjohn,
Crawley, UK) was administered by intraperitoneal injection as an aqueous suspension at a dosage level of 2 mg/kg body weight; using the protocol described above, this dosage is equivalent to 4.2 mg/kg/week, a dose which is higher than the typical dose used to treat refractory RA in humans (1-2 mg/kg/week).
Paw-Swelling
Treatment with p55-sf2 resulted in a dose-dependent reduction in paw-swelling over the treatment period, with the doses of 20 μg and 100 μg giving statistically
significant reductions in paw-swelling relative to mice given saline (P < 0.05). The group of mice given an irrelevant human IgGl mAb as a control did not show any deviation from the saline-treated group (data not shown), indicating that the therapeutic effects of p55-sf2 were attributable to the TNF receptor rather than the human IgGl constant region. Similar reductions in paw-swelling were seen in mice given 300 μg of anti-TNF mAb as in those given 100 μg of p55-sf2, although anti-TNF mAb was
marginally more effective than p55-sf2 at inhibiting paw-swelling. A reduction in paw-swelling was observed in the methylprednisolone acetate treated group that was
comparable in magnitude to the reductions given p55-sf2 at 100 μg or anti-TNF mAb at 300 μg.
Limb Involvement
The change in the number of arthritic limbs over the 10 day treatment period was examined. Results are shown in Table 5.
Figure imgf000030_0001
A strong trend towards reduced limb recruitment was seen in the groups of mice given p55-sf2, anti-TNF mAb or methylprednisolone acetate, but only in the anti-TNF mAb treated group did the reduction reach statistical significance (P < 0.05).
Histology
After 10 days, the mice were sacrificed; the first limb to show clinical evidence of arthritis was removed from each mouse, fixed, decalcified, wax-embedded, and sectioned and stained with haematoxylon and eosin.
Sagittal sections of the proximal lnterphalangeal (PIP) joint of the middle digit of each mouse were studied in a blind fashion and classified according to the presence or absence of erosions, as defined above. Comparisons were thus made between identical joints, and the arthritis was of equal duration. Results are shown in Table 6.
Figure imgf000031_0001
Erosions were present in 92% and 100% of the PIP joints in the saline treated group and the control human IgG1 treated group, respectively. However, only 50%
(P < 0.05) of joints from the mice treated with p55-sf2 (100 μg) and 41% (P < 0.01) of mice given anti-TNF mAb exhibited erosive changes. Some reductions in the proportion of eroded joints were observed in mice treated with 2 μg or 20 μg of p55-sf2, but these were not
statistically significant. Similarly, treatment with methylprednisolone acetate did not significantly reduce joint erosion. Anti-Collagen Antibody Levels
Anti-collagen IgG levels on day 10 were measured by ELISA as described (Williams, R.O. et al., PNAS 89: 9784- 9788 (1992)). Microtitre plates were sensitized with type II collagen, then incubated with serially-diluted test sera. Bound IgG was detected using alkaline phoεphatase-conjugated goat anti-mouse IgG, followed by substrate (dinitrophenol phosphate). Optical densities were read at 405 nm. No differences between any of the treatment groups were detected (data not shown). this suggests that the therapeutic effect of p55-sf2 is not due to a
generalized immunosuppressive effect. C. Effect of Treatment with p55-sf2 in Conjunction with Anti-CD4 Antibody
In view of the high titres of antibodies to p55-sf2 that were detected in mice treated with the fusion
protein, an experiment was carried out to determine whether concurrent administration of anti-CD4 monoclonal antibody (mAb) could enhance the therapeutic effects of p55-sf2. Using the Experimental Procedure described above, a comparison was made of three different treatment regimes: anti-CD4 mAb alone (200 μg), p55-sf2 alone (100 μg) or anti-CD4 mAb (200 μg) plus p55-sf2 (100 μg). A fourth group consisted of untreated control mice. The cell-depleting anti-CD4 mAb (rat IgG2b) consisted of a 1:1 mixture of YTS 191.1.2 and YTA 3.1.2, provided by H.
Waldmann (University of Cambridge, UK) (Galfre, G. et al., Nature 277: 131-133 (1979); Cobbold, S.P. et al., Nature 312: 548-551 (1984); Qin, S. et al., European J.
Immunology 17:1159-1165 (1987)). p55-Sf2 is described above. Paw-Swelling
Treatment with p55-sf2 alone resulted in a marked Inhibition of paw-swelling, but the synergistic inhibitory effect of anti-CD4 mAb in combination with p55-sf2 was remarkable. In contrast, anti-CD4 mAb treatment alone had very little effect on paw-swelling.
Limb Involvement
As before, the progressive involvement of additional limbs following the initial appearance of arthritis was studied. Results are shown in Table 7.
Figure imgf000033_0001
There was a mean increase in limb involvement of 71% in the control group, which was reduced to 56% in the group given anti-CD4 mAb alone, and only 19% in the group given p55-sf2. However, in the group given anti-CD4 mAb plus p55-sf2, the increase in limb involvement was 0%, a statistically significant difference. Histology
Histological analysis of PIP joints of treated mice was carried out as described above. Results are shown in Table 8.
Figure imgf000034_0001
The control group and the group given anti-CD4 mAb alone gave identical results, with 6/6 (100%) of PIP joints in both groups showing significant erosions. However, in the group given p55-sf2 alone, only 2/6 (33%) of PIP joints showed erosions. Only 1/6 (17%) of joints showed erosions in the group given anti-CD4 plus p55-sf2.
Antibody responses to pS5-sf2
The IgM/IgG responses to injected p55-sf2 were measured by ELISA at the end of the treatment period (day 10). Microtitre plates were coated with p55-sf2
(5 μg/ml), blocked, then incubated with serially diluted test sera. Negative controls consisted of sera from saline-treated mice. Bound IgM or IgG were detected by the appropriate goat anti-mouse Ig-alkaline phosphatase conjugate, followed by substrate. Results are shown in Table 9.
Figure imgf000035_0001
High titres of both IgM and IgG antibodies to p55-sf2 were detected in treated mice, with the highest titres being found in the mice given the 100 μg dose. These results indicate that p55-sf2, which is derived from human
proteins, is highly immunogenic in mice. This may account for the slightly greater efficacy of anti-TNF mAb in vivo described in Section B, above, despite the higher
neutralizing titre of the fusion protein in vitro.
Anti-CD4 mAb treatment was found to block almost
completely the formation of both IgM and IgG antibodies to pS5-sf2. Serum Levels of Free P55-sf2
Microtitre plates were coated with recombinant murine TNF-α (Genentech Inc., San Francisco, CA), blocked, then incubated with test sera. Goat anti-human IgG-alkaline phosphatase conjugate was then applied followed by
substrate. Quantitation was by reference to a sample of known concentration of p55-sf2.
The inhibition of the antibody response was
associated with pronounced differences in the circulating levels of p55-sf2 in treated mice. Thus, free p55-sf2 was undetectable in mice given the fusion protein alone, whereas in the mice given anti-CD4 mAb plus p55-sf2, the mean serum level of p55-sf2 was 12.3 μg/ml.
EXAMPLE 3 Treatment of Induced Arthritis in a Murine Model using Cyclosporin A and Anti-TNF Antibody
The murine model of collagen type II induced
arthritis, described above, was used to investigate the efficacy of the CD4+ T cell inhibiting agent cyclosporin A in conjunction with anti-TNF monoclonal antibody (mAb) , for the ability to modulate the severity of joint disease in collagen-induced arthritis. A comparison was made between the efficacy of treatment with cyclosporin A
(CsA), anti-TNF antibody, and combination of CsA and anti-TNF antibody. A. Experimental Procedure
Male DBA/1 mice were immunized intradermally with 100 μg of bovine type II collagen emulsified in complete Freund's adjuvant (Difco Laboratories, East Molsey, UK). The mean day of onset of arthritis was approximately one month after immunization. After the onset of clinically evident arthritis (erythema and/or swelling), groups of mice (11 mice each) were subjected to treatment with one of the following therapies: 50 μg (2 mg/kg) L2 (the isotype control for anti-TNF antibody), intraperitoneally once every three days (days 1, 4 and 7); 250 μg (10 mg/kg) cyclosporin A intraperitoneally daily; 50 μg (2 mg/kg) anti-TNF mAb TN3-19.12, intraperitoneally once every three days (days 1, 4 and 7); or 250 μg cyclosporin A
intraperitoneally daily in conjunction with 50 μg anti-TNF mAb intraperitoneally once every three days. Arthritis was monitored for paw swelling (measured with calipers) for 10 days, after which the mice were sacrificed and joints were processed for histology.
Paw-swellinh
Treatment with cyclosporin A in conjunction with anti-TNF mAb resulted in a reduction in paw-swelling over the treatment period, relative to mice treated with control antibody. Results are shown in Figure 3.
Limb Involvement
As before, the progressive involvement of additional limbs following the initial appearance of arthritis was studied. Results are shown in Table 10.
Figure imgf000038_0001
Treatment with cyclosporin A in conjunction with anti-TNF mAb resulted in statistically significant reductions in limb involvement in comparison to control monoclonal antibody (P = 0.03).
EQUIVALENTS
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to specific embodiments of the invention described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.

Claims

What is claimed is:
1. A method of treating autoimmune or inflammatory
disease in a mammal comprising administering to said mammal a therapeutically effective amount of a combination of a CD4+ T cell inhibiting agent and a TNF antagonist.
2. A method of Claim 1, wherein the CD4+ T cell
inhibiting agent is administered simultaneously with the TNF antagonist.
3. A method of Claim 1, wherein the CD4+ T cell
inhibiting agent is administered sequentially with the TNF antagonist.
4. A method of Claim 1, wherein the CD4+ T cell
inhibiting agent and the TNF antagonist are
administered by a route selected from the group consisting of: subcutaneouεly, intravenously, and intramuscularly.
5. A method of Claim 1, wherein the CD4+ T cell
inhibiting agent and the TNF antagonist are
administered in a pharmaceutically acceptable vehicle.
6. A method of Claim 1, wherein an anti-inflammatory agent is administered in conjunction with the CD4+ T cell inhibiting agent and the TNF antagonist.
7. A method of Claim 6, wherein the anti-inflammatory agent is an agent interfering with the activity or synthesis of TNF.
8. A method of Claim 6, wherein the anti-inflammatory agent is an agent interfering with the activity or synthesis of IL-1.
9. A method of Claim 6, wherein the anti-inflammatory agent is an agent interfering with the activity or synthesis of IL-6.
10. A method of Claim 6, wherein the anti-inflammatory agent is a cytokine with anti-inflammatory
properties.
11. A method of Claim 1, wherein the autoimmune disease is rheumatoid arthritis.
12. A method of Claim 1, wherein the CD4+ T cell
inhibiting agent is an antibody to T cells or to T cell receptors.
13. A method of Claim 1, wherein the CD4+ T cell
inhibiting agent is an antibody to an antigen presenting cell or to the receptors of an antibody presenting cell.
14. A method of Claim 1, wherein the CD4+ T cell
inhibiting agent is a peptide or small molecule which inhibits T cell interaction with antigen presenting cells.
15. A method of treating autoimmune or inflammatory disease in a mammal comprising administering to said mammal a therapeutically effective amount of a combination of a CD4+ T cell inhibiting agent and an inflammatory mediator.
16. A method of Claim 15, wherein the inflammatory
mediator is agent interfering with the activity or synthesis of TNF.
17. A method of Claim 15, wherein the inflammatory
mediator is an agent interfering with the activity or synthesis of IL-l.
18. A method of Claim 15, wherein the inflammatory
mediator is an agent interfering with the activity or synthesis of IL-6.
19. A method of Claim 15, wherein the inflammatory
mediator is a cytokine with anti-inflammatory
properties.
20. A method of treating autoimmune or inflammatory
disease in a mammal, comprising administering to said mammal a therapeutically effective amount of a combination of anti-CD4 antibody and anti-TNF
antibody.
21. A method of treating autoimmune or inflammatory
disease in a mammal, comprising administering to said mammal a therapeutically effective amount of a combination of anti-CD4 antibody and soluble TNF receptor.
22. A method of treating autoimmune or inflammatory disease in a mammal, comprising administering to said mammal a therapeutically effective amount of a combination of anti-CD4 antibody and TNF receptor/IgG fusion protein.
23. A method of treating autoimmune or inflammatory
disease in a mammal, comprising administering to said mammal a therapeutically effective amount of a combination of cyclosporin A and anti-TNF antibody.
PCT/GB1994/000462 1992-10-06 1994-03-10 Treatment of autoimmune and inflammatory disorders WO1995009652A1 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
PCT/GB1994/000462 WO1995009652A1 (en) 1993-10-06 1994-03-10 Treatment of autoimmune and inflammatory disorders
JP7510657A JPH09510952A (en) 1993-10-06 1994-03-10 Treatment of autoimmune and inflammatory diseases
EP94908462A EP0765171A1 (en) 1993-10-06 1994-03-10 Treatment of autoimmune and inflammatory disorders
AU61495/94A AU6149594A (en) 1993-10-06 1994-03-10 Treatment of autoimmune and inflammatory disorders
US08/617,737 US20020068057A1 (en) 1994-03-10 1994-03-10 Treatment of autoimmune and inflammatory disorders
US08/690,775 US6270766B1 (en) 1992-10-08 1996-08-01 Anti-TNF antibodies and methotrexate in the treatment of arthritis and crohn's disease
US09/093,450 US6770279B1 (en) 1992-10-08 1998-06-08 TNFα antagonists and cyclosporin in therapy of rheumatoid arthritis
US09/754,004 US20020010180A1 (en) 1992-10-08 2001-01-03 TNFalpha antagonists and methotrexate in the treatment of TNF-mediated disease
US09/921,937 US20020136723A1 (en) 1992-10-06 2001-08-03 Anti-TNF antibodies and methotrexate in the treatment of autoimmune disease
US10/762,096 US20040228863A1 (en) 1992-10-08 2004-01-20 Treatment of tumor necrosis factor-mediated diseases
US11/225,631 US7846442B2 (en) 1992-10-08 2005-09-12 Methods of treating rheumatoid arthritis with an anti-TNF-alpha antibodies and methotrexate
US12/583,851 US20110123543A1 (en) 1992-10-08 2009-08-26 TNFalpha antagonists and methotrexate in the treatment of TNF-mediated disease

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
PCT/GB1993/002070 WO1994008619A1 (en) 1992-10-08 1993-10-06 Treatment of autoimmune and inflammatory disorders
GBPCT/GB93/02070 1993-10-06
PCT/GB1994/000462 WO1995009652A1 (en) 1993-10-06 1994-03-10 Treatment of autoimmune and inflammatory disorders

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
US95824892A Continuation-In-Part 1992-10-08 1992-10-08
PCT/GB1993/002070 Continuation-In-Part WO1994008619A1 (en) 1992-10-06 1993-10-06 Treatment of autoimmune and inflammatory disorders
US08/403,785 Continuation-In-Part US5741488A (en) 1992-10-06 1993-10-06 Treatment of rheumatoid arthritis with anti-CD4 antibodies in conjunction with anti-TNF antibodies

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US08/403,785 Continuation-In-Part US5741488A (en) 1992-10-06 1993-10-06 Treatment of rheumatoid arthritis with anti-CD4 antibodies in conjunction with anti-TNF antibodies
US60741996A Continuation-In-Part 1992-10-06 1996-02-28

Publications (1)

Publication Number Publication Date
WO1995009652A1 true WO1995009652A1 (en) 1995-04-13

Family

ID=26302381

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1994/000462 WO1995009652A1 (en) 1992-10-06 1994-03-10 Treatment of autoimmune and inflammatory disorders

Country Status (2)

Country Link
JP (1) JPH09510952A (en)
WO (1) WO1995009652A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998005357A1 (en) * 1996-08-01 1998-02-12 The Kennedy Institute Of Rheumatology Anti-tnf antibodies and methotrexate in the treatment of autoimmune disease
EP0949865A1 (en) * 1996-12-10 1999-10-20 Thomas Jefferson University The use of cd4-binding small molecules to inhibit immune responses
EP1615664A1 (en) * 2004-02-18 2006-01-18 Medexgen Inc. Pharmaceutical composition for treatment of immunological disorders
US7217718B2 (en) 2002-11-21 2007-05-15 Genzyme Corporation Induction of immune tolerance
US7687530B2 (en) 2002-11-21 2010-03-30 Genzyme Corporation Inhibition of chronic tissue transplant rejection
US7820161B1 (en) * 1999-05-07 2010-10-26 Biogen Idec, Inc. Treatment of autoimmune diseases
US8440806B2 (en) 2003-03-21 2013-05-14 Biotest Ag Humanized anti-CD4 antibody with immunosuppressive properties
US8518999B2 (en) 2000-05-12 2013-08-27 Genzyme Corporation Modulators of TNF-αsignaling
WO2014164299A1 (en) * 2013-03-13 2014-10-09 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of autoimmune disorders
US8871723B2 (en) 2007-05-03 2014-10-28 Yeda Research And Development Co. Ltd. Glycan modified soluble receptors and binding protein and their use
US9044479B2 (en) 2010-06-16 2015-06-02 Bruce Chandler May Use of levocetirizine and montelukast in the treatment of influenza, common cold and inflammation
US9334325B2 (en) 2008-03-13 2016-05-10 Biotest Ag Method for treating psoriasis
US9512226B2 (en) 2008-03-13 2016-12-06 Biotest Ag Agent for treating disease
US9522148B2 (en) 2013-03-13 2016-12-20 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of traumatic injury
US9528088B2 (en) 2002-06-28 2016-12-27 Life Technologies Corporation Methods for eliminating at least a substantial portion of a clonal antigen-specific memory T cell subpopulation
US9550831B2 (en) 2008-03-13 2017-01-24 Biotest Ag Method for treating psoriasis
US9669025B2 (en) 2013-03-13 2017-06-06 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of vasculitis
US9925183B2 (en) 2014-09-15 2018-03-27 Inflammatory Response Research, Inc. Levocetirizine and montelukast in the treatment of inflammation mediated conditions
US9995733B2 (en) 2009-11-30 2018-06-12 Biotest Ag Agents for treating disease

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1237575B1 (en) * 1999-12-14 2008-08-06 Genentech, Inc. Tnf-alpha antagonist and lfa-1 antagonist for treating rheumatoid arthritis

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989008460A1 (en) * 1988-03-11 1989-09-21 Celltech Limited Antibodies for use in antilymphocyte antibody therapy

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989008460A1 (en) * 1988-03-11 1989-09-21 Celltech Limited Antibodies for use in antilymphocyte antibody therapy

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
D. STEINBRÜCHEL ET AL.: "Monoclonal antibody treatment (anti-CD4 and anti-interleukin-2 receptor) combined with cyclosporin A has a positive but not simple dose-dependent effect on rat renal allograft survival.", SCANDINAVIAN JOURNAL OF IMMUNOLOGY, vol. 34, no. 5, November 1991 (1991-11-01), OXFORD, GB, pages 627 - 633, XP008040841 *
E. RACADOT ET AL.: "Immunological follow-up of 17 patients with rheumatoid arthritis treated in vivo with an anti-T CD4+ monoclonal antibody (B-F5).", CLINICAL AND EXPERIMENTAL RHEUMATOLOGY, vol. 10, no. 4, July 1992 (1992-07-01), PISA, ITALY, pages 365 - 374, XP008022443 *
E. SCHACHT: "Gegenwärtige und zukünftige Therapiestrategien der rheumatoiden Arthritis.", ZEITSCHRIFT FOR RHEUMATOLOGIE, vol. 52, no. 6, November 1993 (1993-11-01), DARMSTADT, GERMANY, pages 365 - 382, XP009019298 *
F. BRENNAN ET AL.: "Inhibitory effect of TNFalpha antibodies on synovial cell interleukin-1 production in rheumatoid arthritis.", THE LANCET, vol. 2, no. 8657, 29 July 1989 (1989-07-29), LONDON, GB, pages 244 - 247, XP008040909 *
G. HORNEFF ET AL.: "Elevated levels of circulating tumor necrosis factor-alpha, interferon-gamma, and interleukin-2 in systemic reactions induced by anti-CD4 therapy in patients with rheumatoid arthritis.", CYTOKINE, vol. 3, no. 3, May 1991 (1991-05-01), BASEL, SWITZERLAND, pages 266 - 267, XP008040843 *
M. ELLIOTT ET AL.: "Treatment of rheumatoid arthritis with chimeric monoclonal antibodies to IFN-alpha: Safety, clinical efficacy and control of the acute phase response.", JOURNAL OF CELLULAR BIOCHEMISTRY, SUPPLEMENT, vol. 0, no. 17 B, 1993, NEW YORK NY, USA, pages 145, XP008040844 *
P. RALPH: "Clinical and preclinical studies presented at the Keystone symposium on arthritis, related diseases, and cytokines.", LYMPHOKINE AND CYTOKINE RESEARCH, vol. 12, no. 4, August 1993 (1993-08-01), NEW YORK NY, USA, pages 261 - 263, XP000891639 *
R. WILLIAMS ET AL.: "Synergy between anti-CD4 and anti-tumor necrosis factor in the amelioration of established collagen-induced arthritis.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE USA, vol. 91, no. 7, 29 March 1994 (1994-03-29), WASHINGTON DC, USA, pages 2762 - 2766, XP008040840 *
See also references of EP0765171A1 *

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6270766B1 (en) 1992-10-08 2001-08-07 The Kennedy Institute Of Rheumatology Anti-TNF antibodies and methotrexate in the treatment of arthritis and crohn's disease
US7846442B2 (en) 1992-10-08 2010-12-07 The Mathilda And Terence Kennedy Institute Of Rheumatology Trust Methods of treating rheumatoid arthritis with an anti-TNF-alpha antibodies and methotrexate
US8298537B2 (en) 1996-08-01 2012-10-30 The Mathilda And Terence Kennedy Institute Of Rheumatology Trust Concomitant treatment of rheumatoid arthritis with anti-TNF-α antibodies and methotrexate
WO1998005357A1 (en) * 1996-08-01 1998-02-12 The Kennedy Institute Of Rheumatology Anti-tnf antibodies and methotrexate in the treatment of autoimmune disease
US8383120B2 (en) 1996-08-01 2013-02-26 The Mathilda And Terence Kennedy Institute Of Rheumatology Trust Concomitant treatment of rheumatoid arthritis with anti-TNF-alpha antibodies and methotrexate
EP1593393A3 (en) * 1996-08-01 2007-12-12 The Kennedy Institute Of Rheumatology Anti-TNF antibodies and methotrexate in the treatment of autoimmune diseases
EP1941904A3 (en) * 1996-08-01 2009-09-09 The Kennedy Institute Of Rheumatology Anti-TNF antibodies and methotrexate in the treatment of autoimmune diseases
US7838489B2 (en) 1996-08-01 2010-11-23 The Mathilda And Terence Kennedy Institute Of Rheumatology Trust Methods of treating rheumatoid arthritis with p75 TNF-alpha receptor and methotrexate
EP0949865A4 (en) * 1996-12-10 2007-04-25 Univ Jefferson The use of cd4-binding small molecules to inhibit immune responses
EP0949865A1 (en) * 1996-12-10 1999-10-20 Thomas Jefferson University The use of cd4-binding small molecules to inhibit immune responses
US9993550B2 (en) 1999-05-07 2018-06-12 Genentech, Inc. Treatment of pemphigus
US20120201818A1 (en) * 1999-05-07 2012-08-09 Curd John G Treatment of multiple sclerosis
US7820161B1 (en) * 1999-05-07 2010-10-26 Biogen Idec, Inc. Treatment of autoimmune diseases
US8921547B2 (en) 2000-05-12 2014-12-30 Genzyme Corporation Modulators of TNF-α signaling
US8518999B2 (en) 2000-05-12 2013-08-27 Genzyme Corporation Modulators of TNF-αsignaling
US9579325B2 (en) 2000-05-12 2017-02-28 Genzyme Corporation Modulators of TNF-α signaling
US9528088B2 (en) 2002-06-28 2016-12-27 Life Technologies Corporation Methods for eliminating at least a substantial portion of a clonal antigen-specific memory T cell subpopulation
US7687530B2 (en) 2002-11-21 2010-03-30 Genzyme Corporation Inhibition of chronic tissue transplant rejection
US7678822B2 (en) 2002-11-21 2010-03-16 Genzyme Corporation Induction of immune tolerance
US7217718B2 (en) 2002-11-21 2007-05-15 Genzyme Corporation Induction of immune tolerance
US8440806B2 (en) 2003-03-21 2013-05-14 Biotest Ag Humanized anti-CD4 antibody with immunosuppressive properties
US8586715B2 (en) 2003-03-21 2013-11-19 Biotest Ag Humanized anti-CD4 antibody with immunosuppressive properties
US8673304B2 (en) 2003-03-21 2014-03-18 Biotest Ag Humanized anti-CD4 antibody with immunosuppressive properties
US8685651B2 (en) 2003-03-21 2014-04-01 Biotest Ag Method for screening for an anti-CD4 antibody suitable for use in immunotherapy
US9758581B2 (en) 2003-03-21 2017-09-12 Biotest Ag Humanized anti-CD4 antibody with immunosuppressive properties
EP1615664A1 (en) * 2004-02-18 2006-01-18 Medexgen Inc. Pharmaceutical composition for treatment of immunological disorders
EP1615664A4 (en) * 2004-02-18 2006-12-27 Medexgen Inc Pharmaceutical composition for treatment of immunological disorders
US8871723B2 (en) 2007-05-03 2014-10-28 Yeda Research And Development Co. Ltd. Glycan modified soluble receptors and binding protein and their use
US9334325B2 (en) 2008-03-13 2016-05-10 Biotest Ag Method for treating psoriasis
US9512226B2 (en) 2008-03-13 2016-12-06 Biotest Ag Agent for treating disease
US9550831B2 (en) 2008-03-13 2017-01-24 Biotest Ag Method for treating psoriasis
US9995733B2 (en) 2009-11-30 2018-06-12 Biotest Ag Agents for treating disease
US9044479B2 (en) 2010-06-16 2015-06-02 Bruce Chandler May Use of levocetirizine and montelukast in the treatment of influenza, common cold and inflammation
US10537568B2 (en) 2010-06-16 2020-01-21 IRR, Inc. Use of levocetirizine and montelukast to ameliorate inflammation following radiation exposure
US11103500B2 (en) 2013-03-13 2021-08-31 IRR, Inc. Use of levocetirizine and montelukast in the treatment of traumatic injury
US9669025B2 (en) 2013-03-13 2017-06-06 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of vasculitis
US9522148B2 (en) 2013-03-13 2016-12-20 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of traumatic injury
US11344545B2 (en) 2013-03-13 2022-05-31 IRR, Inc. Use of levocetirizine and montelukast in the treatment of autoimmune disorders
US9937166B2 (en) 2013-03-13 2018-04-10 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of traumatic injury
EP3300734A3 (en) * 2013-03-13 2018-04-18 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of autoimmune disorders
WO2014164299A1 (en) * 2013-03-13 2014-10-09 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of autoimmune disorders
US9669026B2 (en) 2013-03-13 2017-06-06 Inflammatory Response Research, Inc. Use of levocetirizine and montelukast in the treatment of autoimmune disorders
US10201537B2 (en) 2013-03-13 2019-02-12 IRR, Inc. Use of levocetirizine and montelukast in the treatment of autoimmune disorders
US10206919B2 (en) 2013-03-13 2019-02-19 IRR, Inc. Use of levocetirizine and montelukast in the treatment of vasculitis
US10195193B2 (en) 2014-09-15 2019-02-05 IRR, Inc. Levocetirizine and montelukast in the treatment of inflammation mediated conditions
US10792281B2 (en) 2014-09-15 2020-10-06 IRR, Inc. Levocetirizine and montelukast in the treatment of inflammation mediated conditions
US9925183B2 (en) 2014-09-15 2018-03-27 Inflammatory Response Research, Inc. Levocetirizine and montelukast in the treatment of inflammation mediated conditions
US11590125B2 (en) 2014-09-15 2023-02-28 IRR, Inc. Levocetirizine and montelukast in the treatment of inflammation mediated conditions

Also Published As

Publication number Publication date
JPH09510952A (en) 1997-11-04

Similar Documents

Publication Publication Date Title
US5741488A (en) Treatment of rheumatoid arthritis with anti-CD4 antibodies in conjunction with anti-TNF antibodies
WO1995009652A1 (en) Treatment of autoimmune and inflammatory disorders
CN111068062B (en) Methods of treating interleukin-6 related diseases
ES2299241T3 (en) PREVENTIVES OR REMEDIES FOR INFLAMMATORY INTESTINAL DISEASES CONTAINING ANTAGONIST ANTIBODIES OF THE IL-6 RECEIVER.
EP0936923B1 (en) SUPPRESSION OF TNFalpha AND IL-12 IN THERAPY
US5958409A (en) Method for treating multiple sclerosis
RU2607022C2 (en) Methods and compositions for treating lupus
WO1998042377A1 (en) Preventives or remedies for sensitized t cell-related diseases containing il-6 antagonists as the active ingredient
HU230197B1 (en) Remedies for infant chronic arthritis-relating diseases
WO2007130642A2 (en) Modulation of nkg2d
JP2763197B2 (en) Monoclonal antibodies to induce tolerance
JP2001525816A (en) Inhibition of tumor necrosis factor alpha and vascular endothelial growth factor in therapy
CN111971307A (en) anti-CD 40 antibodies for use in the prevention of transplant rejection
US20020068057A1 (en) Treatment of autoimmune and inflammatory disorders
US6770279B1 (en) TNFα antagonists and cyclosporin in therapy of rheumatoid arthritis
EP0667908B1 (en) Soluble t-cell receptor alpha chain and derivatives used as prophylactic and therapeutic agents for autoimmune diseases
Jenkins et al. Biological modifier therapy for the treatment of rheumatoid arthritis
EP2373339B1 (en) A b cell depleting agent for the treatment of atherosclerosis
EP0765171A1 (en) Treatment of autoimmune and inflammatory disorders
JP3973360B2 (en) CD25 binding molecule for use in the treatment of rheumatoid arthritis or skin diseases

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BB BG BR BY CA CH CN CZ DE DK ES FI GB GE HU JP KG KP KR KZ LK LU LV MD MG MN MW NL NO NZ PL PT RO RU SD SE SI SK TJ TT UA US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

ENP Entry into the national phase

Ref country code: US

Ref document number: 1995 403785

Date of ref document: 19950503

Kind code of ref document: A

Format of ref document f/p: F

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref country code: US

Ref document number: 1996 607419

Date of ref document: 19960228

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1994908462

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 08617737

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1994908462

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: CA