Treatment for inflammatory bowel disease

-

A method for the treatment of inflammatory bowel disease (IBD) is disclosed. The method comprises administration of an antibody, polypeptide or other molecule recognizing VLA-4, a surface molecule expressed on most types of white blood cells and involved in leukocyte adhesion to endothelium and other tissus in the gut.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

This application is a continuation application of U.S. patent application Ser. No. 10/252,978, filed on Sep. 23, 2002, which is a continuation application of U.S. patent application Ser. No. 09/157,452, filed on Sep. 21, 1998, now U.S. Pat. No. 6,482,409, which is a continuation application of U.S. patent application Ser. No. 08/950,660, filed on Oct. 15, 1997, now U.S. Pat. No. 5,932,214, which is a file wrapper continuation application of U.S. patent application Ser. No. 08/456,124, filed on May 31, 1995, now abandoned, which is a continuation-in-part application U.S. patent application Ser. No. 08/373,857, filed on Jan. 18, 1995, now abandoned, which is a continuation-in-part application of U.S. patent application Ser. No. 08/284,603, filed on Aug. 11, 1994, now abandoned, and of International Patent Application No. PCT/US93/00924, filed on Feb. 2, 1993, which is a continuation-in-part application of U.S. patent application Ser. No. 07/835,139, filed Feb. 12, 1992, now abandoned. The contents of the prior applications are hereby incorporated by reference in their entirety for all purposes.

FIELD OF THE INVENTION

The present invention relates to a treatment for inflammatory bowel disease (IBD). More particularly, this invention relates to the use of antibodies recognizing the integrin VLA-4 (very late antigen-4) in the treatment of IBD.

BACKGROUND OF THE INVENTION

Inflammatory bowel disease, or IBD, is a collective term encompassing ulcerative colitis and Crohn's disease (ileitis), which are chronic inflammatory disorders of the gastrointestinal tract. Ulcerative colitis is confined to the large intestine (colon) and rectum, and involves only the inner lining of the intestinal wall. Crohn's disease may affect any section of the gastrointestinal tract (i.e., mouth, esophagus, stomach, small intestine, large intestine, rectum and anus) and may involve all layers of the intestinal wall. Both diseases are characterized by abdominal pain and cramping, diarrhea, rectal bleeding and fever. The symptoms of these diseases are usually progressive, and sufferers typically experience periods of remission followed by severe flareups.

IBD affects an estimated two million people in the United States alone. Although IBD is not considered a fatal illness, prolonged disease can lead to severe malnutrition affecting growth or to the formation of abscesses or intestinal scar tissue, leading in turn to infection or bowel obstruction.

IBD has no cure, and the exact causes of IBD are not yet understood. Conventional treatments for IBD have involved anti-inflammatory drugs, immunosuppressive drugs and surgery. Sulfasalazine and related drugs having the bioactive 5-amino-salicylic acid (5-ASA) moiety are widely used to control moderate IBD symptoms and to maintain remission. Severe inflammation is often treated with powerful corticosteroids and sometimes ACTH or with immunosuppressants such as 6-mercaptopurine and azathioprine. The most common surgical treatments for severe chronic IBD are intestinal resections and, ultimately, colectomy, which is a complete cure only for ulcerative colitis.

Severe side effects are associated with the drugs commonly prescribed for IBD, including nausea, dizziness, changes in blood chemistry (including anemia and leukopenia), skin rashes and drug dependence; and the surgical treatments are radical procedures that often profoundly alter the everyday life of the patient. Accordingly, there is a great need for treatments for IBD that are effective yet less severe in their side effects and are less invasive of the IBD sufferer's body and quality of life.

The search for the causes of IBD and more effective treatments has led several investigators to study diseased and normal tissue on a cellular level. This has led to observations of variations in the normal content of intestinal mucin (Podolsky, 1988 [1]) and to the observation of colonic glycoproteins that emerge only in diseased tissue (Podolsky and Fournier, 1988a [2], 1988b [3]). Researchers have observed that the cell adhesion molecule ICAM-1 is expressed at elevated levels in IBD tissue (Malizia et al., 1991 [4]). This molecule is thought to mediate leukocyte recruitment to sites of inflammation through adhesion to leukocyte surface ligands, i.e., LFA-1 (CD11a/CD18 complex) on all leukocytes and Mac-1 (CD11b/CD18) on phagocytes. (See, e.g., Springer, 1990 [5].) Because flareups of IBD are often accompanied by increased concentrations of neutrophils and lymphocytes in the intestinal submucosa, blocking of interactions between endothelial cell receptors (such as ICAM-1) and their leukocyte ligands (such as LFA-1, Mac-1) has been proposed as a treatment for IBD.

Another cell adhesion molecule, VCAM-1 (vascular cell adhesion molecule-1) is expressed on inflamed endothelium and has been shown to recognize the α4β1 integrin, VLA-4, expressed on the surface of all leukocytes except neutrophils (Springer, 1990 [5]). VCAM-1 also has been found to be expressed constitutively in noninflamed tissue, including Peyer's patch follicular dendritic cells (Freedman et al., 1990 [6]; Rice et al., 1991 [7]). Additionally, besides mediating cell adhesion events, VCAM-1 also has recently been determined to play a costimulatory role, through VLA-4, in T cell activation (Burkly et al., 1991 [8]; Damle and Arrufo, 1991 [9]; van Seventer et al., 1991 [10]). Accordingly, further study of VCAM-1 has been taken up to investigate whether it plays a role as a regulator of the immune response as well as a mediator of adhesion in vivo.

It has now been surprisingly discovered that administering anti-VLA-4 antibody significantly reduces acute inflammation in a primate model for IBD. Cotton top tamarins suffering from a spontaneous intestinal inflammation comparable to ulcerative colitis in humans that were treated with an anti-VLA-4 antibody (HP1/2) showed significant reduction in inflammation of biopsied intestinal tissue.

SUMMARY OF THE INVENTION

Accordingly, the present invention provides novel methods for the treatment of IBD and further provides new pharmaceutical compositions useful in the treatment of IBD. In particular, the present invention provides a method comprising the step of administering to an IBD sufferer a VLA-4 blocking agent, e.g., an anti-VLA-4 antibody, such as antibody HP1/2 Also contemplated is the use of analogous antibodies, antibody fragments, soluble proteins and small molecules that mimic the action of anti-VLA-4 antibodies in the treatment of IBD.

DETAILED DESCRIPTION OF THE DRAWINGS

FIG. 1 is a schematic depicting structure of VCAM 2D-IgG fusion protein described in Example V. VCAM 2D-IgG is a soluble form of the ligand for VLA4 (VCAM1) and consists of the two N-terminal domains of VCAM 1 fused to the human IgG1 heavy chain constant region sequences (Hinges, CH2 and CH3).

FIG. 2 is graph depicting VCAM-IgG inhibitory dose response after single intraperitoneal doses in mouse contact hypersensitivity. VCAM-IgG tested over a dose range of 1.25 to 20 mg/kg (25-400 μg/mouse) maximally inhibited the mouse ear swelling response about 4/5 as well as the PS/2 antibody. Maximal inhibition occurred at the 10 mg/kg dose. The ED50 was about 2.5 mg/kg, i.p. (50 μg/mouse).

FIG. 3 is a graph depicting inhibition of the mouse ear swelling response by VCAM-IgG in mouse contact hypersensitivity. In a confirmation study, VCAM-IgG tested at 20 mg/kg i.p. (400 μg/mouse) maximally inhibited the mouse ear swelling response as effectively as the PS/2 8 mg/kg i.v. antibody dose.

FIG. 4 is a graph depicting the effect of VCAM 2D-IgG fusion protein and controls on prevention of diabetes after adoptive transfer of spleen cells; the frequency of recipients which became diabetic and day of disease onset are shown for transfer of 2×107 splenocytes from diabetic (D) NOD donors with an irrelevant rat LFA-3Ig fusion protein treatment (closed squares), and with VCAM 2D-IgG treatment (open circles) or of recipients which received PBS alone without cells transferred (closed triangles); the splenocytes were transferred with VCAM 2D-IgG or rat LFA-3Ig, and then VCAM 2D-IgG or rat LFA-3Ig was injected every other day through day 17 post-transfer (n=5 for all groups).

FIG. 5 is a graph depicting the effect of VCAM-Ig (30 mg, aerosol given 30 min before antigen challenge) on airway hyperresponsiveness in dual responder sheep. This dose resulted in significant but partial inhibition of LPR, but no effect on AHR.

FIG. 6 is a graph depicting the effect of VCAM-Ig (60 mg, aerosol given 30 min before antigen challenge) on airway hyperresponsiveness in dual responder sheep. This dose resulted in significant but partial inhibition of LPR, and inhibition of AHR.

FIG. 7 is a graph depicting the effect of VCAM-Ig (30 mgs, aerosol given 30 min. before antigen challenge and 8 h. after challenge) on airway hyperresponsiveness in dual responder sheep. This dose resulted in complete inhibition of LPR, but no inhibition of AHR.

FIG. 8 is a graph depicting the effect of VCAM-Ig (15 mgs, aerosol given 2, 8 and 24 h. after antigen challenge) on airway hyperresponsiveness in dual responder sheep. This dose resulted in a significant but partial inhibition of LPR, and inhibition of AHR.

FIG. 9 is a graph depicting the effect of VCAM-Ig (30 mgs, aerosol given 2, and 24 h. after antigen challenge) on airway hyperresponsiveness in dual responder sheep. This optimal dose resulted in complete inhibition of both LPR and AHR.

DETAILED DESCRIPTION OF THE INVENTION

The technology for producing monoclonal antibodies is well known. Briefly, an immortal cell line (typically myeloma cells) is fused to lymphocytes (typically splenocytes) from a mammal immunized with whole cells expressing a given antigen, e.g., VLA-4, and the culture supernatants of the resulting hybridoma cells are screened for antibodies against the antigen. (See, generally, Kohler and Milstein, 1975 [11].)

Immunization may be accomplished using standard procedures. The unit dose and immunization regimen depend on the species of mammal immunized, its immune status, the body weight of the mammal, etc. Typically, the immunized mammals are bled and the serum from each blood sample is assayed for particular antibodies using appropriate screening assays. For example, anti-VLA-4 antibodies may be identified by immunoprecipitation of 125I-labeled cell lysates from VLA-4-expressing cells. (See, Sanchez-Madrid et al., 1986 [13] and Hemler et al., 1987 [14].) Anti-VLA-4 antibodies may also be identified by flow cytometry, e.g., by measuring fluorescent staining of Ramos cells incubated with an antibody believed to recognize VLA-4 (see, Elices et al., 1990 [15]). The lymphocytes used in the production of hybridoma cells typically are isolated from immunized mammals whose sera have already tested positive for the presence of anti-VLA-4 antibodies using such screening assays.

Typically, the immortal cell line (e.g., a myeloma cell line) is derived from the same mammalian species as the lymphocytes. Preferred immortal cell lines are mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine (“HAT medium”). HAT-sensitive mouse myeloma cells may be fused to mouse splenocytes, e.g., using 1500 molecular weight polyethylene glycol (“PEG 1500”). Hybridoma cells resulting from the fusion are then selected using HAT medium, which kills unfused and unproductively fused myeloma cells (unfused splenocytes die after several days because they are not transformed). Hybridomas producing a desired antibody are detected by screening the hybridoma culture supernatants. For example, hybridomas prepared to produce anti-VLA-4 antibodies may be screened by testing the hybridoma culture supernatant for secreted antibodies having the ability to bind to a recombinant α4-subunit-expressing cell line, such as transfected K-562 cells (see, Elices et al., [15]).

To produce anti VLA-4-antibodies, hybridoma cells that test positive in such screening assays may be cultured in a nutrient medium under conditions and for a time sufficient to allow the hybridoma cells to secrete the monoclonal antibodies into the culture medium. Tissue culture techniques and culture media suitable for hybridoma cells are well known. The conditioned hybridoma culture supernatant may be collected and the anti-VLA-4 antibodies optionally further purified by well-known methods.

Alternatively, the desired antibody may be produced by injecting the hybridoma cells into the peritoneal cavity of a mouse primed with 2,6,10,14-tetramethylpentadecane (PRISTANE; Sigma Chemical Co., St. Louis Mo.). The hybridoma cells proliferate in the peritoneal cavity, secreting the antibody, which accumulates as ascites fluid. The antibody may be harvested by withdrawing the ascites fluid from the peritoneal cavity with a syringe.

Several anti-VLA-4 monoclonal antibodies have been previously described (see, e.g., Sanchez-Madrid et al., 1986 [12]; Hemler et al. (1987) [13]; Pulido et al. (1991) [14]). For the experiments herein, an anti-VLA-4 monoclonal antibody designated HP1/2 (obtained from Biogen, Inc., Cambridge, Mass.) was used. The variable regions of the heavy and light chains of the anti-VLA-4 antibody HP1/2 have been cloned, sequenced and expressed in combination with constant regions of human immunoglobulin heavy and light chains. Such a chimeric HP1/2 antibody is similar in specificity and potency to the murine HP1/2 antibody, and may be useful in methods of treatment according to the present invention. Similarly, humanized recombinant anti-VLA-4 antibodies may be useful in these methods. The HP1/2 VH DNA sequence and its translated amino acid sequences are set forth in SEQ ID NO: 1 and SEQ ID NO: 2, respectively. The HP1/2 VK DNA sequence and its translated amino acid sequence are set forth in SEQ ID NO: 3 and SEQ ID NO: 4, respectively.

Monoclonal antibodies such as HP1/2 and other anti-VLA-4 antibodies (e.g., Mab HP2/1, HP2/4, L25, P4C2) capable of recognizing the α chain of VLA-4 will be useful in the present invention. It is most preferred that the antibodies will recognize the B1 or B2 epitopes of the VLA-α4 chain (see, Pulido et al. (1991) [15]). While not wishing to be bound by one scientific theory, anti-VLA-4 antibodies used according to the method of the present invention may specifically inhibit, at least for an initial period, the migration of VLA-4-expressing leukocytes to inflamed sections of the gut. Or, the release of inflammatory mediators and cytokines by leukocytes already recruited to IBD tissue may be blocked by anti-VLA-4 antibodies that prevent some form of VCAM-1-mediated signal transduction, such as the T cell co-activation observed previously (e.g., Burkly et al. 1991 [8]). Monoclonal antibody HP1/2 has been shown to block leukocyte adhesion to VCAM-1-expressing cells but not to promote VLA-4-mediated T cell activation.

The method of the present invention comprises administering to a mammal suffering from inflammatory bowel disease a composition comprising a VLA-4 blocking agent, e.g., an anti-VLA-4 antibody. The examples below set forth the results observed in cotton top tamarins. The physiological and histochemical similarities between a spontaneous chronic diffuse colitis observed in the cotton top tamarin (CTT) and IBD humans has been documented (see, e.g., Podolsky et al., 1985a [16], Podolsky et al., 1985b [17]). Prior studies have also demonstrated parallel responses in CTTs to therapeutic compounds used in the management of the human IBD (see, e.g., Madara et al., 1985 [18]). Accordingly, the results reported herein will be relevant and applicable to, and the method claimed will be useful in any mammal, including humans, suffering from IBD.

The VLA-4 blocking agent, e.g., an anti-VLA-4 antibody, administered in accordance with the present invention may be administered prophylactically to a chronic IBD sufferer, to bring about or maintain remission of the disease; however, preferably the method of the present invention is used to treat acute flareups of the disease.

The VLA-4 blocking agent, e.g., an anti-VLA-4 antibody, can be administered in the form of a composition, e.g., a composition comprising an anti-VLA-4 antibody and a pharmaceutically acceptable carrier. Preferably, the composition will be in a form suitable for intravenous injection. For acute flareups of ulcerative colitis or Crohn's disease, dosages of antibodies from 0.05 mg/kg-patient/day to 5.0 mg/kg-patient/day (preferably from 0.5 mg/kg-patient/day to 2.0 mg/kg-patient/day) may be used, although higher or lower dosages may be indicated with consideration to the age, sensitivity, tolerance, and other characteristics of the patient, the acuteness of the flareup, the history and course of the disease, plasma level and half-life of the antibody employed and its affinity for its recognition site, and other similar factors routinely considered by an attending physician. For maintenance of remission from active disease, dosages from 0.05 mg/kg-patient/day to 5.0 mg/kg-patient/day (preferably from 0.5 mg/kg-patient/day to 2.0 mg/kg-patient/day) may be used, although higher or lower dosages may be indicated and employed with advantageous effects considering the age, sensitivity, tolerance, and other characteristics of the patient, the pattern of flareups, the history and course of the disease, the plasma level and half-life of the agent, e.g., an antibody, employed and its affinity for its recognition site, and other similar factors routinely considered by an attending physician. Dosages may be adjusted, for example, to provide a particular plasma level of an agent, e.g., an antibody, e.g., in the range of 5-30 μg/ml, more preferably 10- 15 μg/ml, for murine antibodies, and to maintain that level, e.g., for a period of time (e.g., 1 week) or until clinical results are achieved (e.g., flareup subsides). Chimeric and humanized antibodies, which would be expected to be cleared more slowly, will require lower dosages to maintain an effective plasma level. Also, antibodies or fragments having high affinity for VLA-4 will need to be administered less frequently or in lower doses than antibodies or antibody fragments of lesser affinity.

Suitable pharmaceutical carriers include, e.g., sterile saline, physiological buffer solutions and the like. The pharmaceutical compositions may additionally be formulated to control the release of the active ingredients or prolong their presence in the patient's system. Numerous suitable drug delivery systems are known for this purpose and include, e.g., hydrogels, hydroxmethylcellulose, microcapsules, liposomes, microemulsions, microspheres, and the like. Phosphate buffered saline (PBS) is a preferred carrier for injectible compositions.

It will also be recognized that for the purposes of the present invention, antibodies capable of binding to the α4 subunit of VLA-4 should be employed. It is preferred that monoclonal antibodies be used.

In addition to naturally produced antibodies, suitable recombinant antibodies capable of binding to VLA-4 may alternatively be used. Such recombinant antibodies include antibodies produced via recombinant DNA techniques, e.g., by transforming a host cell with a suitable expression vector containing DNA encoding the light and heavy immunoglobulin chains of the desired antibody, and recombinant chimeric antibodies, wherein some or all of the hinge and constant regions of the heavy and/or the light chain of the anti-VLA-4 antibody have been substituted with corresponding regions of an immunoglobulin light or heavy chain of a different species (i.e., preferably the same species as the IBD sufferer being treated, to minimize immune response to the administered antibody). (See, e.g., Jones et al., 1986 [19], Ward et al., 1989 [20], and U.S. Pat. No. 4,816,397 (Boss et al.) [21], all incorporated herein by reference.) Recombinant antibodies specifically contemplated herein include CDR-grafted antibodies or “humanized” antibodies, wherein the hypervariable regions of, e.g., murine antibodies are grafted onto framework regions of, e.g., a human antibody. (See, e.g., Riechmann et al., 1988 [22]; Man Sung Co et al., 1991 [23]; Brown, Jr., 1991 [24].)

Furthermore, VLA-4-binding fragments of anti-VLA-4 antibodies, such as Fab, Fab′, F(ab′)2, and F(v) fragments; heavy chain monomers or dimers; light chain monomers or dimers; and dimers consisting of one heavy chain and one light chain are also contemplated herein. Such antibody fragments may be produced by chemical methods, e.g., by cleaving an intact antibody with a protease, such as pepsin or papain, or via recombinant DNA techniques, e.g., by using host cells transformed with truncated heavy and/or light chain genes. Heavy and light chain monomers may similarly be produced by treating an intact antibody with a reducing agent such as dithiothreitol or β-mercaptoethanol or by using host cells transformed with DNA encoding either the desired heavy chain or light chain or both.

As an alternative to hybridoma technology, antibody fragments having the desired anti-VLA-4 specificities may be isolated by phage cloning methods. (See, e.g., Clackson et al., 1991 [25].)

Also, from the discussion herein it will be apparent that other VLA-4 blocking agents can be used in the methods described herein. For the purposes of the invention a VLA-4 blocking agent refers to an agent, e.g., a polypeptide or other molecule, which can inhibit or block VLA-4-mediated binding or which can otherwise modulate VLA-4 function, e.g., by inhibiting or blocking VLA-4-ligand mediated VLA-4 signal transduction and which is effective in the treatment of IBD, preferably in the same manner as are anti-VLA-4 antibodies.

A VLA-4 blocking agent is a molecule which has one or more of the following properties: (1) it coats, or binds to, a VLA-4 antigen on the surface of a VLA-4 bearing cell with sufficient specificity to inhibit a VLA-4-ligand/VLA-4 interaction, e.g., the VLA-4/VCAM-1interaction; (2) it coats, or binds to, a VLA-4 antigen on the surface of a VLA-4 bearing cell with sufficient specificity to modify, and preferably to inhibit, transduction of a VLA-4-mediated signal, e.g., VLA-4/VCAM-1-mediated signaling; (3) it coats, or binds to, a VLA-4-ligand, e.g., VCAM-1 or fibronectin, with sufficient specificity to inhibit the VLA-4/VLA-4-ligand interaction; (4) it coats, or binds to, a VLA-4-ligand, e.g., VCAM-1 or fibronectin, with sufficient specificity to modify, and preferably to inhibit, transduction of VLA-4-ligand mediated VLA-4 signaling, e.g., VCAM-1-mediated VLA-4 signaling. In preferred embodiments the VLA-4 blocking agent has one or both of properties 1 and 2. In other preferred embodiments the VLA-4 blocking agent has one or both of properties 3 and 4.

For purposes of the invention, any agent capable of binding to VLA-4 antigens on the surface of VLA-4 bearing cells and which effectively blocks or coats VLA-4 antigens, is considered to be an equivalent of the monoclonal antibody used in the examples herein.

As discussed herein, the blocking agents used in methods of the invention are not limited to antibodies or antibody derivatives, but may be other molecules, e.g., soluble forms of other proteins which bind VLA-4, e.g., the natural binding proteins for VLA-4. These binding agents include soluble VCAM-1 or VCAM-1 peptides, VCAM-1 fusion proteins, bifunctional VCAM-1/Ig fusion proteins, fibronectin, fibronectin having an alternatively spliced non-type III connecting segment, and fibronectin peptides containing the amino acid sequence EILDV (SEQ ID NO: 17) or a similar conservatively substituted amino acid sequence. These binding agents can act by competing with the cell-surface binding protein for VLA-4 or by otherwise altering VLA-4 function. For example, a soluble form of VCAM-1 (see, e.g., Osborn et al. 1989 [26]) or a fragment thereof may be administered to bind to VLA-4, and preferably compete for a VLA-4 binding site, thereby leading to effects similar to the administration of anti-VLA-4 antibodies. Soluble VCAM-1 fusion proteins can be used in the methods described herein. For example, VCAM-1, or a fragment thereof which is capable of binding to VLA-4 antigen on the surface of VLA-4 bearing cells, e.g., a fragment containing the two N-terminal domains of VCAM-1, can be fused to a second peptide, e.g., a peptide which increases the solubility or the in vivo life time of the VCAM-1 moiety. The second peptide can be a fragment of a soluble peptide, preferably a human peptide, more preferably a plasma protein, or a member of the immunoglobulin super family. In particularly preferred embodiments the second peptide is IgG or a portion or fragment thereof, e.g., the human IgG1 heavy chain constant region. A particularly preferred fusion protein is the VCAM 2D-IgG fusion.

Included in the invention as VLA-4 blocking agents are (at least) peptides (preferably peptides of less than 5 or 10 amino acid resides in length), peptide mimetics, carbohydrates, and small molecules, such as oligosaccharides, capable of blocking VLA-4 in any of the ways described herein, e.g., by binding VLA-4 antigens on the surface of VLA-4-bearing cells or by binding to VLA-4-ligands. Small molecules such as oligosaccharides that mimic the binding domain of a VLA-4 ligand and fit the receptor domain of VLA-4 may also be employed. (See, J. J. Devlin et al., 1990 [24], J. K. Scott and G. P. Smith, 1990 [25], and U.S. Pat. No. 4,833,092 (Geysen) [26], all incorporated herein by reference.) Examples of small molecules useful in the invention can be found in Adams et al. U.S. Ser. No. 08/376,372, filed Jan. 23, 1995, hereby incorporated by reference.

In preferred embodiments more than one VLA-4 blocking agent is administered to a patient, e.g., a VLA-4 blocking agent which binds to VLA-4 can be combined with a VLA-4 blocking agent which binds to VCAM-1.

The use of such VLA-4 blocking agents, e.g., VLA-4-binding polypeptides or molecules that effectively decrease inflammation in IBD tissue in treated subjects is contemplated herein as an alternative method for treatment of IBD.

It is also contemplated that anti-VLA-4 antibodies may be used in combination with other antibodies having a therapeutic effect on IBD. For instance, to the extent that the beneficial effects reported herein are due to the inhibition of leukocyte recruitment to endothelium, combinations of anti-VLA-4 antibodies with other antibodies that interfere with the adhesion between leukocyte antigens and endothelial cell receptor molecules may be advantageous. For example, in addition to the use of anti-VLA-4 antibodies in accordance with this invention, the use of anti-ELAM-1 antibodies, anti-VCAM-1 antibodies, anti-ICAM-1 antibodies, anti-CDX antibodies, anti-CD18 antibodies, and/or anti-LFA-1 antibodies may be advantageous.

When formulated in the appropriate vehicle, the pharmaceutical compositions contemplated herein may be administered by any suitable means such as orally, intraesophageally or intranasally, as well as subcutaneously, intramuscularly, intravenously, intra-arterially, or parenterally. Ordinarily intravenous (i.v.) or parenteral administration will be preferred to treat flareup conditions; oral administration in a timed release vehicle will be preferred to maintain remission.

Improvement for IBD patients as a result of the methods of this invention can be evaluated by any of a number of methods known to practitioners in this art. For example, improvement in observed symptomology such as the Truelove-Witts criteria (see, e.g., Lichtiger, et al., 1990 [30]) may be used, or specimens of colon tissue may be biopsied and characterized histologically (see, e.g., Madara et al., 1985 [18]).

In another aspect the invention features a chimeric molecule which includes: (1) a VLA-4 targeting moiety, e.g., a VCAM-1 moiety capable of binding to VLA-4 antigen on the surface of VLA-4 bearing cells; (2) optionally, a second peptide, e.g., one which increases solubility or in vivo life time of the VLA-4 targeting moiety, e.g., a member of the immunoglobulin super family or fragment or portion thereof, e.g., a portion or a fragment of IgG, e.g., the human IgG1 heavy chain constant region, e.g., CH2 and CH3 hinge regions; and (3) a toxin moiety. The VLA-4 targeting moiety can be any naturally occurring VLA-4 ligand or fragment thereof, e.g., a VCAM-1 peptide, fibronectin, fibronectin having an alternatively spliced non-type III connecting segment, and fibronectin peptides containing the amino acid sequence EILDV (SEQ ID NO: 17) or a similar conservatively substituted amino acid sequence. A preferred targeting moiety is a soluble VCAM-1 fragment, e.g., the N-terminal domains 1 and 2 of the VCAM-1 molecule. The toxin moiety can be any agent which kills or inactivates a cell when the toxin is targeted to the cell by the VLA-4 targeting moiety. Toxin moieties include: cytotoxic peptide moieties, e.g., Diphtheria toxin A, Pseudomonas Exotoxin, Ricin A, Abrin A, Schigella toxin, or Gelonin; radionucleotides; and chemotherapeutic agents.

The chimeric molecule can be used to treat a subject, e.g., a human, at risk for a disorder, e.g., IBD, characterized by the presence of cells bearing VLA-4, and preferably activated VLA-4.

The methods and compositions of the present invention will be further illuminated by reference to the following examples, which are presented by way of illustration and not of limitation.

EXAMPLE I

VCAM1 Expression in the Colon

Experiments were performed to determine whether active IBD involved the expression of endothelial cell surface proteins involved in leukocyte adhesion. Expression of VCAM-1 in colon tissue of IBD sufferers and normal or uninvolved colon tissue controls was evaluated. Human colonoscopic biopsy tissue samples were obtained, with informed consent, and prepared as frozen sections by mounting in OCT compound (TissueTek) and quick freezing in isopentane/liquid nitrogen. The human colon samples were from normal colon, active ulcerative colitis colon (UC-active), inactive ulcerative colitis colon (UC-inactive), uninvolved ulcerative colitis colon (UC-uninvolved), active Crohn's Disease colon (CD-active), and uninvolved Crohn's Disease colon (CD-uninvolved).

Frozen sections (˜4μ) were placed on gelatin-coated slides (1% gelatin, heated at 60° C. for 1-2 min., air dried, 1% formaldehyde at room temp., air dried), air dried 30 minutes, fixed in acetone for ten minutes at 4° C., washed three times in PBS and treated with 0.3% H202 in methanol (30 min., room temp.). The slides were then washed with PBS for 30 minutes, incubated with dilute normal human serum (1:100), and incubated with anti-VCAM-1 antibody 4B9 (1:100; obtained as a gift from Dr. John Harlan) for 60 minutes at room temperature. Control slides were incubated with an anti-bovine serum albumin (anti-BSA) antibody (Sigma Chemical Co., St. Louis Mo.). The samples were then washed with PBS for 10 minutes and incubated with a secondary biotinylated rabbit anti-mouse immunoglobulin (Dako Corp., Santa Barbara, Calif.) for 60 minutes at room temperature, then visualized using avidin-linked peroxidase (VECTASTAIN, Vector Labs, Burlingame Calif.).

The results of these tests are set forth in the following TABLE I:

TABLE I Endothelial Cell Staining In Human Tissue VCAM-1 Expression Tissue (n) n (%) Normal (11) 6 (54.4) UC active (23) 14 (60.9) UC inactive (8) 5 (62.5) UC uninvolved (10) 4 (40.0) CD active (9) 5 (55.5) CD uninvolved (12) 7 (58.3)

These data confirm the observations such as those reported by Freedman et al. [6] and Rice et al. [7] that VCAM-1 is expressed in both IBD-involved colon tissue and in normal colon tissue. In both CD and UC tissues, VCAM-1 was observed by immunocytochemistry in about 60% of samples.

EXAMPLE II

Anti-VLA-4 Antibody Recognition of CTT White Blood Cells

An anti-VLA-4 monoclonal antibody (HP1/2, obtained from Biogen, Inc., Cambridge Mass.) was tested to confirm that it recognized an epitope on CTT leukocytes.

Blood samples (3 ml) from CTTs were heparinized and the CTT peripheral blood mononuclear leukocytes (PBLs) were isolated using a Ficoll-Hypaque gradient (Pharmacia) according to the manufacturer's instructions for isolation of human PBLs. CTT PBLs were examined for their ability to bind to the murine anti-human VLA-4 monoclonal antibodies HP1/2 and HP2/1 by FACS analysis using a Becton Dickenson FACStar and standard techniques (see, e.g., Lobb et al., 1991a [31]). Both monoclonal antibodies bound to CTT PBLs, indicating that both human and CTT VLA4 have similar epitopes recognized by these two antibodies.

CTT PBLs were also observed to adhere to microtiter plates coated with immobilized recombinant soluble human VCAM-1 (Biogen, Inc.), which binding was blocked by HP1/2 and HP2/1. These results show that CTT PBLs bind to VCAM-1 in a VLA-4-dependent manner, and that HP1/2 and HP2/1 block the interaction of CTT VLA-4 with human VCAM-1. (Cf. Lobb et al., 1991b [32].)

EXAMPLE III

Cotton Top Tamarin Trials

A stock solution in sterile saline of the Anti-VLA-4 antibody, HP1/2 (IgG1), and a placebo control (saline only), were prepared for administration to ten cotton top tamarins (CTTs) exhibiting symptoms of spontaneous colitis (i.e., diarrhea, etc.; see, Madara et al. [18]). Five CTTs received HP1/2 and five received placebo, by intravenous injection. The CTTs receiving HP1/2 were injected with 1 mg HP1/2 per day (i.e., about 2 mg/kg/day, based on approximate half-kilogram weight of a CTT) for eight days (on Days 0, 1, 2, 3, 4, 5, 6, and 7 of the trial). Colon tissue samples obtained from the animals were biopsied every other day (on Days 0, 2, 4, 6, 8, and 10 of the trial).

Data from the biopsies were used to determine an acute inflammation index for each animal, giving a semi-quantitative analysis of the course of the colitis. (See, Madara et al. [18].) The inflammation indices before the trial began (Day 0) and at the end of the trial at Day 10 are set forth in Table II, below: (“Treated CTTs” received antibody HP1/2; “Control CTTs” received placebo)

TABLE II Day O Day 10 AII* AII Treated CTTs 1 2 0 2 1 0 3 1 0 4 2 0 5 2 1 MEAN 1.6 0.2 Control CTTs C1 2 0 C2 2 1 C3 1 1 C4 2 2 C5 2 2 MEAN 1.8 1.2
*AII = acute inflammation index

These results show that treatment with anti-VLA4 antibody resulted in a significant (p<0.01) decrease in acute inflammation index.

EXAMPLE IV

The trial described in Example III was repeated using 14 CTTs, 7 receiving HP1/2 and 7 receiving placebo. The change in acute inflammation index from Day 0 to Day 10 is set forth in Table III:

TABLE III Day 0 Day 10 AII AII Treated CTTs 6 2 0 7 2 0 8 2 0 9 2 0 10 2 0 11 2 1 12 2 2 MEAN 2.0 0.43 Control CTTs C6 2 2 C7 2 2 C8 1 1 C9 2 1  C10 2 1  C11 2 0  C12 1 0 MEAN 1.71 1.00

The foregoing results show a significant decrease in acute inflammation in the CTTs receiving HP1/2.

EXAMPLE V

VCAM 2D-IgG is a soluble form of the ligand for VLA4 (VCAM1) which consists of the two N-terminal domains of VCAM1 fused to the human IgG1 heavy chain constant region sequences (Hinges, CH2 and CH3). The VCAM 2D-IgG DNA sequence and its translated amino acid sequence are shown in SEQ ID NO: 5 and SEQ ID NO:12, respectively. In other systems, administration of this fusion peptide has been shown to have effects which are similar to the administration of anti-VLA-4 monoclonal antibody. FIG. 1 illustrates the fusion protein structure. The fusion protein was constructed by recombinant techniques as described below.

Isolation of cDNA of Human IgG1 Heavy Chain

Region and Construction of Plasmid pSAB144

In order to isolate a cDNA copy of the human IgG1 heavy chain region, RNA was prepared from COS7 cells which has been transiently transfected by the plasmid VCAM1-IgG1(also known as pSAB133). Construction of plasmid VCAM1-IgG1 is described in PCT patent application WO 90/13300. The RNA was reverse transcribed to generate cDNA using reverse transcriptase and random hexamers as the primers. After 30 min. at 42° C., the reverse transcriptase reaction was terminated by incubation of the reaction at 95° C. for 5 min. The cDNA was then amplified by PCR (Polymerase Chain Reaction, see, e.g., Sambrook et al., Molecular Cloning, Vol. 3, pp. 14.1-14.35 (Cold Spring Harbor; 1989) [34]) using the following kinased primers: 370-31 (SEQ ID NO: 6 and SEQ ID NO:13):

5′TCGTC GAC AAA ACT CAC ACA TGC C        Asp Lys Thr His Thr Cys

which contains a SalI site, and 370-32 (SEQ ID NO: 7):

5′ GTAAATGAGT GCGGCGGCCG CCAA,

which encodes the carboxy terminal lysine of the IgG1 heavy chain constant region, followed by a NotI site.

The PCR amplified cDNA was purified by agarose gel electrophoresis and glass bead elution for cloning in plasmid pNN03. Plasmid pNN03 was constructed by removing the synthetic polylinker sequence from the commercially available plasmid pUC8 (Pharmacia, Piscataway, N.J.) by restriction endonuclease digestion and replacing the synthetic polylinker sequence with the following novel synthetic sequence (SEQ ID NO: 8):

GCGGCCGCGG TCCAACCACC AATCTCAAAG CTTGGTACCC GGGAATTCAG ATCTGCAGCA TGCTCGAGCT CTAGATATCG ATTCCATGGA TCCTCACATC CCAATCCGCG GCCGC.

The purified PCR amplified cDNA fragment was ligated to pNN03 which had been cleaved with EcoRV, dephosphorylated, and purified by low melt agarose gel electrophoresis. The ligation reaction was used to transform E. coli JA221 and the resulting colonies were screened for a plasmid containing an insert of approximately 700 bp. The identity of the correct insert was confirmed by DNA sequence analysis, and the plasmid was designated pSAB144.

Construction of Plasmid pSAB142

The plasmid pSAB142 was constructed as follows. cDNA prepared from COS cells transfected with pSAB133 (as described in the previous section) was subjected to PCR amplification using oligonucleotides 370-01 and 370-29. Oligonucleotide 370-01 includes a NotI site and the nucleotides corresponding to amino acids 1 through 7 of the VCAM-1 signal sequence (SEQ ID NO:9 and SEQ ID NO:14):

5′ GAGCTCGAGG CGGCCGCACC ATG CCT GGG AAG ATG GTC                          Met Pro Gly Lys Met Val GTG Val

Oligonucleotide 370-29 corresponds to the VCAM-1 amino acids 214-219 and includes a SalI site (SEQ ID NO: 10):

5′AA GTC GAC TTG CAA TTC TTT TAC

The amplified DNA fragment was ligated to the vector fragment of pNN03, cleaved by EcoRV.
Construction of pSAB 132

pJOD-S (Barsoum, J., DNA and Cell Biol., 9, pp.293-300 (1990) [35]) was modified to insert a unique NotI site downstream from the adenovirus major late promoter so that NotI fragments could be inserted into the expression vector. pJOD-S was linearized by NotI cleavage of the plasmid DNA. The protruding 5′ termini were blunt-ended using Mung Bean nuclease, and the linearized DNA fragment was purified by low melting temperature agarose gel electrophoresis. The DNA fragment was religated using T4 DNA ligase. The ligated molecules were then transformed into E. coli JA221. Colonies were screened for the absence of a NotI site. The resulting vector was designated pJOD-S delta Notl. pJOD-8 delta Notl was linearized using SalI and the 5′ termini were dephosphorylated using calf alkaline phosphatase. The linearized DNA fragment was purified by low melting temperature agarose gel electrophoresis and ligated in the presence of phosphorylated oligonucleotide ACE175, which has the following sequence

(SEQ ID NO: 11): TCGACGCGGC CGCG

The ligation mixture was transformed into E. coli JA221, and colonies were screened for the presence of a plasmid having a NotI site. The desired plasmid was named pMDR901.

In order to delete the two SV40 enhancer repeats in the Sv40 promoter which controls transcription of the DHFR cDNA, pMDR901 and pJODΔe-tPA (Barsoum, DNA and Cell Biol., 9, pp. 293-300 (1990) [35]), both were cleaved with AatII and DraIII. The 2578 bp AatII-DraIII fragment from pMDR901 and the 5424 bp AatII-DraIII fragment from pJODΔe-tPA were isolated by low melting temperature agarose gel electrophoresis and ligated together. Following transformation into E. coli JA221, the resulting plasmid, pMDR902, was isolated. pSAB132 was then formed by eliminating the EcoRI-NotI fragment of pMDR902 containing the adenovirus major late promoter and replacing it with an 839 bp EcoRI-NotI fragment from plasmid pCMV-B (Clontech, Palo Alto, Calif.) containing the human cytomegalovirus immediate early promoter and enhancer.

Construction of pSAB146

pSAB144 was cleaved with SalI and NotI, and the 693 bp fragment isolated. pSAB142 was cleaved with NotI and SalI and the 664 bp fragment was isolated. The two fragments were ligated to pSAB132 which had been cleaved with NotI, and the 5′ termini dephosphorylated by calf alkaline phosphatase. The resulting plasmid, pSAB146, contained the DNA sequence encoding the VCAM-1 signal sequence, the amino terminal 219 amino acids of mature VCAM-1, ten amino acids of the hinge region of IgG1 and the CH2 and CH3 constant domains of IgG1.

Production of VCAM 2D-IgG from a Stably Transformed CHO Cell Line

A recombinant VCAM 2D-IgG expression vector was constructed as described below and transfected into CHO cells to produce a cell line continuously secreting VCAM 2D-IgG.

The 1.357 kb NotI fragment containing the VCAM 2D-IgG coding sequence of pSAB146 was purified by agarose gel electrophoresis. This fragment was ligated into the NotI cloning site of the expression vector pMDR901, which uses the adenovirus 2 major late promoter for heterologous gene expression and the selectable, amplifiable dihydrofolate reductase (dhfr) marker. The ligated DNA was used to transform E. coli DH5. Colonies containing the plasmid with the desired, correctly oriented insert were identified by the presence of 5853 and 3734 bp fragments upon digestion with HindIII; and 4301, 2555, 2293, and 438 bp fragments upon digestion with BglII. The resultant recombinant VCAM 2D-IgG expression vector was designated pEAG100. The identity of the correct insert was confirmed by DNA sequence analysis.

The recombinant expression plasmid pEAG100 was electroporated into dhfr-deficient CHO cells according to the published protocol of J. Barsoum (DNA Cell Biol 9: 293-300, 1990 [35]), with the following changes: 200 μg of PvuI-linearized pEAG100 plasmid and 200 μg of sonicated salmon sperm DNA were used in the electroporation protocol. In addition, cells were selected in alpha-complete medium supplemented with 200 nM methotrexate.

To determine expression levels of secreted VCAM 2D-IgG, clones were transferred to a flat bottom 96 well microtiter plate, grown to confluency and assayed by ELISA as described below.

Wells of Immulon 2 plates (Dynatech, Chantilly, Va.) were each coated with anti-VCAM MAb 4B9 (isolated and purified on Protein A Sepharose as described by Carlos et al, 1990 [56]) with 100 μl of anti-VCAM 4B9 MAb diluted to 10 μg/ml in 0.05 M sodium carbonate/bicarbonate buffer, pH 9.6, covered with Parafilm, and incubated overnight at 4° C. The next day, the plate contents were dumped out and blocked with 200 μl/well of a block buffer (5% fetal calf serum in 1×PBS), which had been filtered through a 2 filter. The buffer was removed after a 1 hour incubation at room temperature and the plates were washed twice with a solution of 0.05% Tween-20 in 1×PBS. Conditioned medium was added at various dilutions. As a positive control, an anti-mouse Ig was also included. Block buffer and LFA-3TIP constituted as negative controls. The samples and controls were incubated at room temperature for 2 hours.

The plates were then washed twice with a solution of 0.05% Tween-20 in 1×PBS. Each well, except for the positive control well, was then filled with 50 μl of a 1:2000 dilution of HRP-Donkey anti-human IgG (H+L) (Jackson Immune Research Laboratories, Inc.; West Grove, Pa.) in block buffer. The positive control well was filled with 50 μl of a 1:2000 dilution of HRP-Goat anti-mouse IgG (H+L)(Jackson Immune Research Laboratories, Inc.; West Grove, Pa.) in block buffer. The plates were then incubated for 1 hour at room temperature.

The HRP conjugated Ab solutions were removed, and the wells were washed twice with 0.05% Tween-20 in 1×PBS. Then, 100 μl of HRP-substrate buffer was added to each well at room temperature. HRP-substrate buffer was prepared as follows: 0.5 ml of 42 mM 3,3′, 5,5′-tetramethylbenzidine (TMB), (ICN Immunobiologicals, Lisle, S.C., Catalogue No. 980501) in DMSO (Aldrich) was slowly added to 50 ml of substrate buffer (0.1 M sodium acetate/citric acid, pH 4.9); followed by addition of 7.5 μl of 30% hydrogen peroxide (Sigma, Catalogue No. H-1009).

The development of a blue color in each well was monitored at 650 nm on a microtiter plate reader. After 7-10 minutes, the development was stopped by the addition of 100 μl of 2N Sulfuric acid. The resulting yellow color was read at 450 nm on a microtiter plate reader. A negative control well was used to blank the machine.

Purification of VCAM 2D-IgG

CHO cells expressing VCAM 2D-IgG were grown in roller bottles on collagen beads. Conditioned medium (5 Liters) was concentrated to 500 ml using an Amicon S1Y10 spiral ultrafiltration cartridge (Amicon, Danvers, Mass.). The concentrate was diluted with 1 liter of Pierce Protein A binding buffer (Pierce, Rockford, Ill.) and gravity loaded onto a 10 ml Protein A column (Sepharose 4 Fast Flow, Pharmacia, Piscataway, N.J.). The column was washed 9 times with 10 ml of Protein A binding buffer and then 7 times with 10 ml of PBS. VCAM 2D-IgG was eluted with twelve-5 ml steps containing 25 mM H3PO4 pH 2.8, 100 mM NaCl. The eluted samples were neutralized by adding 0.5 M Na2HPO4 pH 8.6 to 25 mM. Fractions were analyzed for absorbance at 280 nm and by SDS-PAGE. The three peaks fractions of highest purity were pooled, filtered, aliquoted and stored at −70° C. By SDS-PAGE, the product was greater than 95% pure. The material contained less than 1 endotoxin unit per mg of protein. In some instances, it was necessary to further purify the Protein A eluate product on Q-Sepharose FF (Pharmacia). The protein A eluate was diluted with 3 volumes of 25 mM Tris HCl pH 8.0 and loaded onto a Q-Sepharose FF column at 10 mg VCAM 2D-IgG per ml of resin. The VCAM 2D-IgG was then eluted from the Q-Sepharose with PBS.

Evaluation of VCAM 2D-IgG

The following examples (Example VI-VIII) have been included to provide evidence that the VCAM-Ig parallels the efficacy of anti-α4 monoclonal antibodies in vivo in three out of three models studied below. Two murine models, contact hypersensitivity model and NOD diabetic mouse model have been tested with the VCAM-Ig fusion protein, as well as the sheep asthma model. The sheep asthma model is quite distinct from the mouse models in that: 1) it is a different species; 2) a different organ (lung, versus skin or pancreas); 3) the leukocytes targeted are unknown (i.e. not clearly T cell dependent classical immune reaction); 4) aerosol versus i.v./i.p. administration; 5) mAbs used are different (anti-murine-α4 mAbs PS/2 and R1/2 in mice versus anti-human-α4 mAb HP1/2 in sheep). Therefore, using the correlation between VCAM-Ig and the anti-α4 mAbs provided by the examples below, strong evidence has been generated that the VCAM-Ig fusion protein would be effective in the treatment of IBD because the anti-human-α4 mAb HP1/2 has been proven effective in the cotton top tamarin trials described above.

EXAMPLE VI The Effect of VCAM-Ig on the Murine Contact Hypersensitivity Response

The experiments described below can be performed essentially as follows.

BALB/c mice (purchased from Taconic Farms, Germantown, N.Y.) were sensitized to 2,4-Dinitrofluorobenzene (DNFB)(Aldrich Chemical Co., Wis.) by the application of 25 μg of 0.5% DNFB in an acetone/olive oil vehicle (4:1 v/v) on day 0 and again on day 1 to the shaved abdominal skin (see, Current Protocols in Immunology, John Wiley and Sons, NY 1001, Section 4.2). Mice were sensitized to oxazalone by the application of 150 μl of 3.0% oxazalone in 100% ethanol to the shaved abdominal skin on day 0 (see, Current Protocols in Immunology, John Wiley and Sons, NY 1001, Section 4.2). For the day 0 sensitization procedure the mice were lightly anesthetized with avertin (0.010-0.015 ml/h body weight of 2.5% avertin stock, intraperitoneal). Immediately prior to challenge, baseline ear thickness measurements were recorded in units of 10−4 inches, for both ears of all of the mice, using a Mitutoyo engineer's micrometer (see, Current Protocols in Immunology, John Wiley and Sons, NY 1001, Section 4.2). All measurements were done in quadruplicate and summarized as mean thickness±SEM. The DNFB response was elicited on day 5 by skin-painting the dorsal surface of the left ear with 10 μl of 0.2% DNFB in 4:1 acetone/olive oil. The dorsal surface of the right (control) ear was painted with 10 μl of vehicle only and was a specificity control for the reaction. The oxazalone response was elicited on day 6 by painting the dorsal surface of the left ear with 20 μl of 1% oxazalone in ethanol. The right ear was painted with 20 μl of ethanol. The CHS response was assessed by obtaining 24-h ear thickness measurements. The average change in left ear thickness (ΔT) for each group was determined by substituting baseline ear thickness from 24-h ear thickness. The immunomodulatory effect of the antibody treatment was evaluated by calculating the percentage inhibition as follows: % Inhibition=[1-ΔT treatment/ΔT control]×100%.

VCAM-IgG was administered to mice in varying intraperitoneal doses, 4-6 hours prior to challenge. VCAM-IgG was administered in pyrogen-free, azide-free PBS. As shown in FIG. 2, VCAM-IgG tested over a dose range of 1.25 to 20 mg/kg (25-400 μg/mouse) maximally inhibited the mouse ear swelling response about 4/5 as well as the PS/2 antibody, which is a IgG2b rat anti-mouse α-4 antibody (anti-VLA4 antibody). Maximal inhibition was observed at the 10 mg/kg dose. The ED50 was about 2.5 mg/kg, i.p. (50 μg/mouse).

In a confirmation study (FIG. 3), VCAM-IgG tested at 20 mg/kg i.p. (400 μg/mouse) maximally inhibited the mouse ear swelling response as effectively as the PS/2 8 mg/kg i.v. antibody dose.

In summary, VCAM-IgG at optimal dose can block the skin CHR response by about 40-50%, just as well as does an optimal dose of the PS/2 antibody.

EXAMPLE VII The Effect of VCAM-Ig Treatment on Adoptive Transfer of Diabetes

For the adoptive transfer of diabetes experiments, NOD mice were obtained from Taconic Farms (Germantown, N.Y.) or from the Joslin Diabetes Center (Boston, Mass.). Spontaneously diabetic (D) females of recent onset (13-20 weeks of age) were used as spleen cell donors and 8 week old nondiabetic (Y) females served as recipients. Spleen cells from 4 week old nondiabetic (Y) female donors which fail to transfer disease were used as a negative control.

Recipient mice were placed on acidified water (1:8400 dilution of concentrated HCl in water) one week prior to sublethal irradiation (775 rad) performed in split doses (300 rad, 300 rad, and 175 rad) on each of three days (day -2, -1, and the day of transfer), in order to minimize any incidence of intestinal infection subsequent to high dose irradiation (Gamma Cell 1000 Cesium137 source, Nordion International, Inc., Ontario, Canada). Spleens were harvested from diabetic donors or from nondiabetic controls, cell suspensions made and red cells lysed with Hemolytic Geys solution. Spleen cells were injected intravenously (2-3×107 in 0.2 ml PBS) and were pretreated with either 100 μg VCAM 2D-IgG or 100 μg of irrelevant LFA-3Ig fusion protein control. Another group received PBS alone without cells transferred. The fusion protein LFA-3Ig (LFA-3TIP) was isolated and purified as described in PCT US92/02050 and Miller et al., 1993 [36]. The VCAM 2D-IgG fusion protein or irrelevant LFA-3Ig protein was administered at a dose of 100 μg/0.2 ml intraperitoneally twice weekly through day 17. This concentration was sufficient to provide a serum level of fusion protein sufficient to saturate VLA4-positive cells, the serum levels determined by ELISA as described above. Diabetes onset was monitored as described above.

The results of the evaluation are shown in FIG. 4. As shown in this Figure, VCAM 2D-IgG fusion protein significantly inhibits the onset of diabetes in recipients of cells from diabetic donor mice (D/VCAM-Ig, open circles) with 60% incidence by day 30 post-transfer, as compared to the mice which received cells from diabetic donor (data not shown) and LFA-3Ig irrelevant control Ig fusion protein (D/LFA-3 Ig) which had already achieved 60% incidence by day 15 post-transfer. Mice which received no cells (PBS only) did not develop disease. There were n=5 mice per experimental group.

EXAMPLE VIII The Effect of VCAM-Ig Treatment on the Sheep Model of Airways Hvper-Responsiveness

Experiments were performed essentially as described by Abraham et al. [33]. Briefly, allergic sheep having natural allergic cutaneous reaction to 1:1000 or 1:10,000 dilutions of Ascaris suum extract (Greer Diagnostics, Lenoir N.C.) were tested, and sheep demonstrating both early and late phase airway response (“dual responders”) to inhalation challenge with Ascaris suum antigen were selected. To measure breathing mechanics and physical changes in the airways, the sheep were restrained in a prone position with heads immobilized. A balloon catheter was advanced through one nostril under topical anesthesia with 2% lidocaine solution to the lower esophagus, and a cuffed endotracheal tube was advanced through the other nostril (using a flexible fiberoptic bronchoscope as a guide) for the measurement of airway mechanics and during aerosol challenges. Pleural pressure was estimated with the esophageal balloon catheter (filled with 1 ml of air) positioned 5-10 cm from the gastroesophageal junction. In this position, end expiratory pleural pressure ranged between −2 and −5 cm H2O. Once the balloon was placed, it was secured so that it remained in position for the duration of the experiment. Lateral pressure in the trachea was measured with a sidehole catheter, (inner diam. 2.5 mm) advanced through and positioned distal to the tip of the endotracheal tube. Transpulmonary pressure (the difference between tracheal and pleural pressure) was measured with a differential pressure transducer catheter system (MP45, Validyne, Northridge, Calif.). The pressure transducer catheter system showed no phase shift between pressure and flow to a frequency of 9 HZ. Pulmonary resistance (RL) was measured by connecting the proximal end of the endotracheal tube to a Fleich pneumotachograph (Dyna Sciences, Blue Bell Pa.). Signals indicating flow and transpulmonary pressure were recorded on an oscilloscope recorder (Model DR-12; Electronics for Medicine, White Plains, N.Y.) linked to a computer for automatic calculation of pulmonary resistance (RL) from transpulmonary pressure, respiratory volume (obtained by digital integration) and flow by the mid-volume technique, analyzed from 5-10 breaths. Thoracic gas volume (Vtg) was measured immediately after determination of RL in a constant volume body plethysmograph. Specific lung resistance (SRL) was calculated from these values (SRL=Vtg×RL).

Airway responsiveness was determined by performing dose response curves to inhaled carbachol. The dose response curves were plotted using measurements of SRL taken immediately after inhalation of buffer (PBS) alone and after each consecutive administration of 10 breaths of increasing concentrations of carbachol in PBS. The concentrations of carbachol were 0.25%, 0.5%, 1.0%, 2.0% and 4.0% wt/vol in PBS. The provocation test was discontinued when SRL increased over 400% from the post-PBS value or after the highest carbachol concentration had been administered. Airway responsiveness was determined by calculating from the dose response curves the cumulative carbachol dose in breath units (BU) that increased specific lung resistance 400% over the post buffer value (PD400%). One breath unit was defined as one breath of a 1% wt/vol carbachol solution. Thus, the greater the suppression of airway hyper-responsiveness, the greater the number of breath units would be required before observing the same bronchoconstriction as seen in the controls.

Each sheep was subjected to a trial as a control in which a placebo (PBS without additive) was used as a pretreatment 30 minutes before allergen challenge with Ascaris suum antigen (Greer Diagnostics, Lenoir, N.C.). The antigen solution was delivered as an aerosol using a disposable medical nebulizer (RAINDROP®, Puritan Bennett, Lenexa, Kans.) that provided an aerosol with a mass median aerodynamic diameter of 3.2 μM (geometric SD 1.9) as determined by an Andersen cascade impactor. The Ascaris suum extract was diluted in PBS to a concentration of 82,000 Protein Nitrogen Units(PNU)/ml. The output of the nebulizer was directed into a plastic T-tube, one end of which was connected to the inspiratory port of a Harvard respirator. A dosimeter connected to the nebulizer consisting of a solenoid valve and a 20 psi compressed air source and the solenoid valve was activated at the beginning of the inspiratory cycle of the Harvard respirator for one second. The aerosol delivered at a tidal volume of 500 ml and a rate of 20 breaths per min. for 20 min. Each sheep was challenged with an equivalent dose of antigen (400 breaths) in the control and VCAM1-IgG1 trials. Carbachol aerosols for the dose response curves were also generated by nebulizer as described above.

An airway challenge trial using five pairs of responder allergic sheep was conducted in order to investigate the efficacy of VCAM1-IgG1 (VCAM-Ig) fusion protein in the sheep model of airways hyper-responsiveness. The efficacy of the aerosol delivery of the VCAM-Ig was investigated. VCAM-Ig was delivered via nebulizer in the form of an aerosol.

In order to optimize therapeutic efficacy, aerosolized VCAM-Ig was first administered at different dosing regimens. These experiments are summarized in FIGS. 5-9. In all the experiments, the control sheep received placebo. In the first experiment, two animals were given 30 mg VCAM-Ig (1 mg/kg) at 30 minutes prior to antigen challenge, which is the standard time used for other therapeutic agents. Under these conditions significant but partial inhibition of the late phase response (LPR) but no effect on airways hyper-responsiveness (AHR) was observed (FIG. 5). This result was not surprising as VCAM-Ig was found previously to be a little less potent than mAb HP1/2. In the second experiment, therefore, the dose of VCAM-Ig was increased to 60 mg. This dose resulted in the partial blockage of LPR as in the previous experiment, but now AHR was blocked too (FIG. 6). However, due to the serious problems which resulted from attempting to aerosolize such a large volume, in subsequent experiments dosages were administered at different time intervals. In the third experiment, 30 mg of VCAM-Ig were administered at 30 minutes prior to and 8 hours after antigen challenge. Here the LPR was blocked completely but no blockage of the AHR was observed (FIG. 7). With respect to the LPR this represented a single dose 30 minutes prior to antigen challenge (equivalent to experiment 1) because the second dose at 8 hours was given after the LPR was largely over. In the fourth experiment, 15 mg of VCAM-Ig were administered at 2, 8 and 24 hours. Here partial blockage of the LPR and blockage of the AHR was observed (FIG. 8). In the final experiment, 30 mg of VCAM-Ig were administered at 2 and 24 hours and resulted in complete blockage of both the LPR and AHR (FIG. 9). This optimal dosage was tested on four animals with the same result.

In summary, ten animals have all shown partial or complete inhibition of the LPR versus a placebo control, and complete inhibition of both the LPR and AHR can be achieved under optimal conditions (30 mg of VCAM-Ig administered at 2 and 24 hours after antigen challenge).

The foregoing examples are intended as an illustration of the method of the present invention and are not presented as a limitation of the invention as claimed hereinafter. From the foregoing disclosure, numerous modifications and additional embodiments of the invention will be apparent to those experienced in this art. For example, actual dosage used, the type of antibody, antibody fragment or analog used, mode of administration, exact composition, time and manner of administration of the treatment, and many other features all may be varied without departing from the above description. All such modifications and additional embodiments are within the contemplation of this application and within the scope of the appended claims.

Cited Publications

  • [1] D. Podolsky, “Colonic Glycoproteins in Ulcerative Colitis: Potential Meaning in Heterogeneity, ” Inflammatory Bowel Diseases: Basic Research and Clinical Implications, Falk Symposium, Titisee, Germany, Jun. 7-9, 1987 (Kluwer Academic Publishers; Boston 1987) pp. 449-56.
  • [2] D. Podolsky and D. Fournier, “Alterations in Mucosal Content of Colonic Glycoconjugates in Inflammatory Bowel Disease Defined by Monoclonal Antibodies, Gastroenterology, 95, pp. 379-87 (1988).
  • [3] D. Podolsky and D. Fournier, “Emergence of Antigenic Glycoprotein Structures in Ulcerative Colitis Detected Through Monoclonal Antibodies,” Gastroenterology, 95, pp. 371-8 (1988).
  • [4] G. Malizia et al., “Expression of Leukocyte Adhesion Molecules by Mucosal Mononuclear Phagocytes in Inflammatory Bowel Disease,” Gastroenterology, 100, pp. 150-9 (1991).
  • [5] T. Springer, “Adhesion Receptors of the Immune System,” Nature, 346, pp. 425-34 (August 1990).
  • [6] A. Freedman et al., “Adhesion of Human B Cells to Germinal Centers In Vitro Involves VLA-4 and INCAM-110,” Science, 249, pp. 1030-33 (1990).
  • [7] G. E. Rice et al., “Vascular and Nonvascular Expression of INCAM-110, Amer. J. Pathology, 138(2), pp. 385-93 (1991).
  • [8] L. Burkly, et al., “Signaling by Vascular Cell Adhesion Molecule-1 (VCAM-1) Through VLA-4 Promotes CD3-dependent T Cell Proliferation,” Eur. J. Immunol., 21, pp. 2871-75 (1991).
  • [9] N. Damle and A. Aruffo, “Vascular Cell Adhesion Molecule 1 Induces T-cell Antigen Receptor-dependent Activation of CD4+ T Lymphocytes,” Proc. Natl. Acad. Sci. USA, 88, pp. 6403-7 (1991).
  • [10] G. van Seventer et al., “Analysis of T Cell Stimulation by Superantigen Plus Major Histocompatibility Complex Class II Molecules or by CD3 Monoclonal Antibody: Costimulation by Purified Adhesion Ligands VCAM-1, ICAM-1, but Not ELAM-1,” J. Exp. Medicine, 174, pp. 901-13 (1991).
  • [11] Kohler and Milstein, “Continuous Cultures of Fused Cells Secreting Antibody of Predefined Specificity,” Nature, 256, pp. 495-7 (1975).
  • [12] F. Sanchez-Madrid et al., “VLA-3: A novel polypeptide association within the VLA molecular complex: cell distribution and biochemical characterization,” Eur. J. Immunol., 16, pp. 1343-9 (1986).
  • [13] M. E. Hemler et al., “Characterization of the Cell Surface Heterodimer VLA-4 and Related Peptides,” J. Biol. Chem., 262(24), pp. 11478-85 (1987).
  • [14] M. Elices et al., “VCAM-1 on Activated Endothelium Interacts with the Leukocyte Integrin VLA-4 at a Site Distinct from the VLA-4/Fibronectin Binding Site,” Cell, 60, pp. 577-84 (1990).
  • [15] R. Pulido et al., “Functional Evidence for Three Distinct and Independently Inhibitable Adhesion Activities Mediated by the Human Integrin VLA-4,” J. Biol. Chem., 266(16), pp. 10241-5 (1991).
  • [16] D. Podolsky, et al., “Colonic Mucin Composition in Primates Selective Alterations Associated with Spontaneous Colitis in the Cotton-top Tamarin,” Gastroenterology, 88, pp. 20-5 (1985).
  • [17] D.-Podolsky et al., “Spontaneous Colitis In Cotton-Top Tamarins: Histologic, Clinical and Biochemical Features of an Animal Model of Chronic Colitis,” Digestive Diseases and Sciences, 30(4), Abstract, p. 396 (1985).
  • [18] J. Madara et al., “Characterization of Spontaneous Colitis in Cotton-Top Tamarin (Saguinus oedipus) and Its Response to Sulfasalazine,” Gastroenterology, 88, pp. 13-19 (1985).
  • [19] P. Jones et al., “Replacing the Complementarity-Determining Regions in a Human Antibody with Those From a Mouse,” Nature, 321, pp. 522-25 (1986).
  • [20] E. Ward et al., “Binding Activities of a Repertoire of Single Immunoglobulin Variable Domains Secreted From Escherichia coli,” Nature, 341, pp. 544-6 (1989).
  • [21] U.S. Pat. No. 4,816,397, Boss et al., “Multichain Polypeptides or Proteins And Processes For Their Production”, issued Mar. 28, 1989.
  • [22] L. Riechmann et al., “Reshaping Human Antibodies for Therapy,” Nature, 332, pp. 323-7 (1988).
  • [23] Man Sun Co et al., “Humanized Antibodies for Antiviral Therapy,” Proc. Natl. Acad. Sci. USA, 88, pp. 2869-73 (1990).
  • [24] P. Brown, Jr. et al., “Anti-Tac-H, a Humanized Antibody to the Interleukin 2 Receptor, Prolongs Primate Cardiac Allograft Survival,” Proc. Natl. Acad. Sci. USA, 88, pp. 2663-7 (1990).
  • [25] T. Clackson et al., “Making Antibody Fragments Using Phage Display Libraries,” Nature, 352, pp. 624-28 (1991).
  • [26] L. Osborn et al., “Direct Expression Cloning of Vascular Cell Adhesion Molecule 1, a Cytokine-induced Endothelial Protein That Binds to Lymphocytes,” Cell, 59, pp. 1203-11 (1989).
  • [27] J. Devlin et al., “Random Peptide Libraries: A Source of Specific Protein Binding Molecules,” Science, 249, pp. 400-406 (1990).
  • [28] J. Scott and G. Smith, “Searching for Peptide Ligands with an Epitope Library,” Science, 249, pp. 386-90 (1990).
  • [29] U.S. Pat. No. 4,833,092, Geysen, “Method For Determining Mimotopes”, issued May 23, 1989.
  • [30] S. Lichtiger and D. Present, “Preliminary Report: Cyclosporin in Treatment of Severe Active Ulcerative Colitis,” Lancet, 336, pp. 16-19 (1990).
  • [31] R. Lobb et al., “Expression and Functional Characterization of a Soluble Form of Endothelial-Leukocyte Adhesion Molecule 1,” J. Immunol., 147(1), pp. 124-29 (1991).
  • [32] R. Lobb et al., “Expression and Functional Characterization of a Soluble Form of Vascular Cell Adhesion Molecule 1,” Biochem. Biophys. Res. Commun., 178(3), pp. 1498-1504(1991).
  • [33] W. M. Abraham et al., “Cellular Markers of Inflammation in the Airways of Allergic Sheep with and without Allergen-induced Late Responses,” Am. Rev. Respir. Dis., 138, 1565-1571 (1988)
  • [34] Sambrook et al., “Polymerase Chain Reaction”, Molecular Cloning, 3, 370-31 (1989)
  • [35] J. Barsoum, DNA and Cell Biol., 9, 293-300 (1990)
  • [36] Miller et al., J. Exp. Med., 178, 211 (1993)

The foregoing documents are incorporated herein by reference.

Claims

1.-25. (canceled)

26. A method for the treatment of inflammatory bowel disease, comprising administering to a mammal suffering from inflammatory bowel disease a composition comprising a soluble fibronectin polypeptide comprising an alternatively spliced non-type III connecting segment or a fragment thereof.

27. The method according to claim 26, wherein the mammal is a human.

28. The method according to claim 26, wherein the mammal has ulcerative colitis.

29. The method according to claim 26, wherein the mammal has Crohn's Disease.

30. The method according to claim 26, wherein the composition is administered during an acute flareup of the inflammatory bowel disease.

31. The method according to claim 26, wherein the fibronectin polypeptide or fragment thereof is a component of a chimeric molecule.

32. The method according to claim 26, wherein the composition is administered intravenously.

33. The method according to claim 31, wherein the chimeric molecule further comprises a toxin moiety.

34. The method according to claim 33, wherein the toxin moiety is a cytotoxic peptide selected from the group consisting of Diphtheria toxin A, Pseudomonas Exotoxin, Ricin A, Abrin A, Schigella toxin, or Gelonin.

35. The method according to claim 33, wherein the toxin moiety is a radionucleotide or a chemotherapeutic agent.

36. The method according to claim 31, wherein the chimeric molecule further comprises an additional peptide that increases solubility or in vivo life time of the soluble fibronectin or fragment thereof.

37. The method according to claim 36, wherein the additional peptide is a heavy chain constant region of human IgG1.

38. The method according to claim 36, wherein the additional peptide comprises at least one of the CH2 or CH3 regions of the heavy chain of human IgG1.

Patent History
Publication number: 20070253902
Type: Application
Filed: Dec 6, 2006
Publication Date: Nov 1, 2007
Applicant:
Inventors: Roy Lobb (Westwood, MA), Linda Burkly (West Newton, MA)
Application Number: 11/634,822
Classifications
Current U.S. Class: 424/1.690; 424/134.100; 514/8.000
International Classification: A61K 38/16 (20060101); A61K 39/00 (20060101); A61K 51/00 (20060101); A61P 1/00 (20060101);