Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTI-CLTA4, ANTI-GLUT2 PROTEIN FOR THE TREATMENT OF TYPE 1 DIABETES
Document Type and Number:
WIPO Patent Application WO/2013/013029
Kind Code:
A1
Abstract:
The disclosure relates to a protein composed of a first polypeptide or polypeptide domain having a first specific binding activity for Cytotoxic T-lymphocyte Antigen 4 (CTLA-4) expressed on a T-cell cell surface and a second specific binding activity for Glucose Transporter 2 (GLUT2) or an extracellular ectodomain thereof expressed on a pancreatic β-cell surface, wherein binding of the first polypeptide or polypeptide domain to CTLA-4 induces a CTLA-4 specific agonist response in the T-cell, and binding of the second polypeptide or polypeptide domain to GLUT2 or an ectodomain thereof does not inhibit GLUT2 glucose transporter function, wherein said agonist response in the T-cell induces a response that reduces immunoreactivity against pancreatic β-cells.

Inventors:
PRABHAKAR BELLUR S (US)
VASU CHENTHAMARAKSHAN (US)
BHATTACHARYA PALASH (US)
Application Number:
PCT/US2012/047378
Publication Date:
January 24, 2013
Filing Date:
July 19, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV ILLINOIS (US)
PRABHAKAR BELLUR S (US)
VASU CHENTHAMARAKSHAN (US)
BHATTACHARYA PALASH (US)
International Classes:
C07K16/28; A61P3/10; C07K16/46
Domestic Patent References:
WO2005115419A12005-12-08
WO2000037504A22000-06-29
Other References:
VASU CHENTHAMARAKSHAN ET AL: "Targeted engagement of CTLA-4 prevents autoimmune thyroiditis.", INTERNATIONAL IMMUNOLOGY, vol. 15, no. 5, May 2003 (2003-05-01), pages 641 - 654, XP002683393, ISSN: 0953-8178
KARUMUTHIL-MELETHIL SUBHA ET AL: "Dendritic Cell-Directed CTLA-4 Engagement during Pancreatic beta Cell Antigen Presentation Delays Type 1 Diabetes", JOURNAL OF IMMUNOLOGY, vol. 184, no. 12, June 2010 (2010-06-01), pages 6695 - 6708, XP002683394, ISSN: 0022-1767
LI RUOBING ET AL: "Enhanced engagement of CTLA-4 induces antigen-specific CD4(+)CD25(+)Foxp3(+) and CD4(+)CD25(-) TGF-beta 1(+) adaptive regulatory T cells", JOURNAL OF IMMUNOLOGY, vol. 179, no. 8, October 2007 (2007-10-01), pages 5191 - 5203, XP002683395, ISSN: 0022-1767
CHATENOUD L: "CD3-SPECIFIC ANTIBODY-INDUCED ACTIVE TOLERANCE: FROM BENCH TO BEDSIDE", NATURE REVIEWS. IMMUNOLOGY, NATURE PUBLISHING GROUP, GB, vol. 3, no. 2, 1 February 2003 (2003-02-01), pages 123 - 132, XP009058496, ISSN: 1474-1733, DOI: 10.1038/NRI1000
CHUNG DENISE T ET AL: "Anti-thymocyte globulin (ATG) prevents autoimmune encephalomyelitis by expanding myelin antigen-specific Foxp3+ regulatory T cells", INTERNATIONAL IMMUNOLOGY, vol. 19, no. 8, August 2007 (2007-08-01), pages 1003 - 1010, XP002683396, ISSN: 0953-8178
ANDERSON ET AL., ANNUAL REVIEW OF IMMUNOLOGY, vol. 23, 2005, pages 447 - 85
KENT ET AL., NATURE, vol. 435, 2005, pages 224 - 8
ACHENBACH, CURRENT DIABETES REPORTS, vol. 5, 2005, pages 98 - 103
WAID ET AL., JOURNAL OF LEUKOCYTE BIOLOGY, vol. 84, 2008, pages 431 - 9
BRODE ET AL., JOURNAL OF IMMUNOLOGY, vol. 177, 2006, pages 6603 - 12
ALARD ET AL., DIABETES, vol. 55, 2006, pages 2098 - 105
MANIRARORA ET AL., PLOS ONE, vol. 3, 2008, pages E3739
CHEATEM ET AL., CLIN IMMUNOL, vol. 131, 2009, pages 260 - 70
GAUDREAU ET AL., J. IMMUNOL, vol. 179, 2007, pages 3638 - 47
KARUMUTHIL-MELETHIL ET AL., JOURNAL OF IMMUNOLOGY, vol. 184, 2010, pages 6695 - 708
YOU ET AL., PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OFAMERICA, vol. 104, 2007, pages 6335 - 40
CHATENOUD, NATURE REVIEWS. IMMUNOLOGY, vol. 3, 2003, pages 123 - 32
CHUNG ET AL., INTERNATIONAL IMMUNOLOGY, vol. 19, 2007, pages 1003 - 10
O'SHEA ET AL., NATURE REVIEWS. IMMUNOLOGY, vol. 2, 2002, pages 37 - 45
SILVA ET AL., IMMUNOTHERAPY, vol. 2, 2010, pages 817 - 33
TODD ET AL., IMMUNITY, vol. 15, 2001, pages 387 - 95
TIVOL ET AL., IMMUNITY, vol. 3, 1995, pages 541 - 7
SHRIKANT ET AL., IMMUNITY, vol. 11, 1999, pages 483 - 93
RATTS ET AL., INT IMMUNOL, vol. 11, 1999, pages 1889 - 96
SCHWARZ ET AL., JLMMUNOL, vol. 165, 2000, pages 1824 - 31
CHAI ET AL., JLMMUNOL, vol. 165, 2000, pages 3037 - 42
BLUESTONE, JLMMUNOL, vol. 158, 1997, pages 1989 - 93
LINSLEY ET AL., IMMUNITY, vol. 1, 1994, pages 793 - 801
LONDRIGAN ET AL., TRANSPLANTATION, vol. 90, 2010, pages 951 - 7
VERGANI ET AL., DIABETES, vol. 59, 2010, pages 2253 - 64
FIFE ET AL., THE JOURNAL OF CLINICAL INVESTIGATION, vol. 116, 2006, pages 2252 - 61
LI ET AL., JLMMUNOL, vol. 179, 2007, pages 5191 - 203
VASU ET AL., INT IMMUNOL, vol. 15, 2003, pages 641 - 54
THORENS ET AL., BIOCHEM SOC TRANS, vol. 22, 1994, pages 684 - 7
LI ET AL., JIMMUNOL, vol. 179, 2007, pages 5191 - 203
; VASU ET AL., INT IMMUNOL, vol. 15, 2003, pages 641 - 54
TODD; WICKER, IMMUNITY, vol. 15, 2001, pages 387 - 95
TANG ET AL., NATURE IMMUNOLOGY, vol. 7, pages 83 - 92
TREMBLEAU ET AL., THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 181, 1995, pages 817 - 21
CHEN ET AL., J EXP MED, vol. 202, 2005, pages 1387 - 97
BENDING ET AL., THE JOURNAL OF CLINICAL INVESTIGATION, vol. 119, 2009, pages 565 - 72
WAGNER ET AL., PROC. NAT. ACAD. SIC U.S.A., vol. 99, 2002, pages 3782 - 7
KARUMUTHIL-MELETHIL ET AL., JOURNAL OF IMMUNOLOGY, vol. 184, 2010, pages 6695 - 708
WILLIAMS; RUDENSKY, NATURE IMMUNOLOGY, vol. 8, 2007, pages 277 - 84
WAN; FLAVELL, NATURE, vol. 445, 2007, pages 766 - 70
WANG ET AL., PLOS ONE, vol. 3, 2008, pages E2705
BHATTACHARYA ET AL., JOURNAL OFLEUKOCYTE BIOLOGY, vol. 89, 2011, pages 235 - 49
Attorney, Agent or Firm:
MENSAH, Kwame, N. (300 South Wacker DriveChicago, IL, US)
Download PDF:
Claims:
WHAT IS CLAIMED:

1. A protein comprising a first polypeptide or polypeptide domain having a first specific binding activity for Cytotoxic T-lymphocyte Antigen 4 (CTLA-4) expressed on a T-cell cell surface and a second specific binding activity for Glucose Transporter 2 (GLUT2) or an extracellular ectodomain thereof expressed on a pancreatic beta-cell surface, wherein binding of the first polypeptide or polypeptide domain to CTLA-4 induces a CTLA-4 specific agonist response in the T cell, and binding of the second polypeptide or polypeptide domain to GLUT2 or an ectodomain thereof does not inhibit GLUT2 glucose transporter function, wherein said agonist response in the T cell reduces immunoreactivity against pancreatic beta cells.

2. The protein of claim 1, wherein the first and second polypeptide or polypeptide domain are independently an antibody molecule or a specific -binding fragment thereof.

3. The protein of claim 2 wherein the first and second polypeptide or polypeptide domain independently comprise at least one immunoglobulin heavy chain and one immunoglobulin light chain.

4. The protein of claim 2, wherein the first and second polypeptide or polypeptide domain independently comprise a monovalent antibody fragment, an F(ab) fragment, an F(ab)2 fragment, F(ab)' fragment , or an Fv fragment.

5. The protein of claim 2, wherein the second antibody molecule is a monoclonal antibody identified by ATCC Accession No. or an antigen-binding fragment thereof.

6. The protein of claim 1, wherein the first and second polypeptide or polypeptide domains are covalently attached to one another by a linker moiety.

7. A pharmaceutical composition comprising the protein of any of claims 1, 2, 3, 4, 5, or 6 with a pharmaceutically acceptable carrier or adjuvant.

8. A method for preventing diabetes in an animal comprising the step of administering to the animal a therapeutically effective amount of the pharmaceutical composition of claim 9.

9. A method for protecting pancreatic β-cells in an animal comprising the step of administering to the animal a protective amount of the pharmaceutical composition of claim 9.

10. A method of producing Treg cells that down-regulate autoimmune responses to pancreatic β-cells in an animal comprising the step of administering to the animal an effective amount of the pharmaceutical composition of claim 9.

1 1. The method of claim 13 wherein the pancreatic tissue specific T cells are down modulated and induced Treg cells are predominantly pancreatic tissue-specific Treg cells.

12. A method of producing immune tolerance to pancreatic β-cells in an animal comprising the step of administering to the animal an effective amount of the pharmaceutical composition of claim 9.

13. The method of claim 14, wherein the immune tolerance is pancreatic islet cell specific T cell tolerance.

Description:
ANTI-CLTA4, ANTI-GLUT2 PROTEIN FOR THE TREATMENT OF TYPE 1

DIABETES

This application claims the benefit of priority to U.S. Provisional Patent Application Serial No: 61/509,316, filed July 19, 2011, and is incorporated by reference herein in its entirety.

BACKGROUND OF THE INVENTION

[0001] Type-1 diabetes (T1D) is a chronic autoimmune disorder thought to be caused by autoreactive T-cells that participate in destruction of insulin-producing pancreatic β-cells in the islets of Langerhans. Complete destruction of β-cells results in a lifelong dependence on exogenous insulin. Specifically, CD4 + T-cells are known in the art to play a critical role in T1D pathology (Anderson, et al, 2005, Annual review of immunology 23: 447-85). Prior to diagnosis of T1D, pancreatic islets are infiltrated by inflammatory cells including CD4 + T- cells and antibodies to various β-cell antigens. These proteins are present in the sera of patients at risk (Kent et al, 2005, Nature 435: 224-8; Achenbach et al, 2005, Current diabetes reports 5:98-103).

[0002] It has been suggested that autoimmune destruction of β-cells and diabetes onset may be associated with a reduction in regulatory T-cell (Treg) numbers and/or functions (Waid et al, 2008, Journal of leukocyte biology 84:431-9; Brode et al, 2006, Journal of immunology 177:6603-12). On the other hand, antigen presenting cells (APCs) in NOD mice and T1D patients have been shown to be defective in their ability to stimulate CD4 + CD25 + Treg function although the Tregs themselves appear to be functional (Alard et al, 2006, Diabetes 55:2098-105; Manirarora et al, 2008, PloS one 3 : e3739). The inventors and others in the art have shown that restoring Treg function or inducing adaptive Tregs may be an effective method for preventing and or stabilizing autoimmune diabetes (Cheatem et al, 2009, Clin Immunol 131 : 260-70; Gaudreau et al, 2007, J Immunol 179: 3638-47;

Karumuthil-Melethil et al, 2010, Journal of immunology 184:6695-708; You et al, 2001 , Proceedings of the National Academy of Sciences of the United States of America 104:6335- 40).

[0003] Existing and emerging therapies in the art utilize broad-based

immunoregulatory strategies, such as inhibition or deletion of lymphocytes subsets and/or establishing immune tolerance via activation of Tregs and include for example non-mitogenic anti-CD3 or anti-thymocyte globulin (Chatenoud 2003, Nature reviews. Immunology 3 : 123- 32; Chung et al, 2007 ' , International immunology 19: 1003-10). These approaches, however, result in global attenuation of the immune response and render the patient susceptible to opportunistic infections and cancers. In this regard, biologies including monoclonal antibodies (mAbs) and tumor necrosis factor (TNF) inhibitors are commonly prescribed to individuals suffering from autoimmune diseases (O'Shea et ah, 2002, Nature reviews.

Immunology 2:37-45). Three licensed mAbs (adalimumab, etanercept and infliximab) are currently on the market for the treatment of immune mediated inflammatory diseases, including diabetes mellitus (Silva et ah, 2010, Immunotherapy 2: 817-33). However, in September 2008, the FDA announced that manufacturers of TNF inhibitors must strengthen existing warnings on the risk of fungal infections, in particular, histoplasmosis.

[0004] As a result, safer, targeted therapeutics are needed in the art. A highly desired alternative approach is the induction of T-cell tolerance to β-cell antigens for prevention of disease development in patients at risk or with recent onset of disease. Central to the immune response are T-cells whose activity can be up- or down-regulated resulting in an immune response that attacks or ignores an antigen, respectively. The ability to down-regulate or become tolerant to specific antigens is crucial for preventing or treating autoimmune diseases. Current clinical approaches for these indications are not target specific and result in global attenuation of the immune response, however. It is therefore beneficial to selectively suppress self-reactive T-cells while leaving the rest of the immune system intact. To do so, T- cell expressed cell surface markers provide a convenient and specific target for inducing desired immunomodulation.

[0005] Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) is a cell surface marker molecule expressed on activated T-cells that plays a critical role in maintaining peripheral tolerance and has been linked to insulin-dependent diabetes (IDD) disease susceptibility in both human patients and NOD mice (Todd et ah, 2001, Immunity 15: 387-95;

Karumuthil-Melethil et ah, Id.; Tivol et ah, 1995, Immunity 3 : 541-7). It has been demonstrated that peripheral T-cell tolerance in vivo requires CTLA-4 engagement (Shrikant et ah, 1999, Immunity 1 1 : 483-93; Ratts et ah, 1999, Int Immunol 1 1 : 1889-96; Schwarz et ah, 2000, J Immunol 165: 1824-31; Chai ei a/., 2000, J Immunol 165: 3037-42). T-cell activation requires T-cell receptor engagement of its cognate antigen-MHC complex and CD28 binding to the B7 ligands (i.e. CD80 and CD86) on APCs. Activation of T-cells results in increased expression of the T-cell surface molecule CTLA-4 (Bluestone, 1997, J Immunol 158: 1989-93; Linsley et ah, 1994, Immunity 1 : 793-801). Use of CTLA-4-Ig which blocks co-stimulation has been widely explored for treating autoimmunity and transplant rejection (Londrigan et al, 2010, Transplantation 90: 951-7; Vergani et ah, 2010, Diabetes 59: 2253- 64). However, this approach down-modulates all activated T-cells and can cause generalized immune suppression. Therfore, this exploration has not been very successful. Because signaling through CTLA-4 down modulates T-cell responses, engagement of CTLA-4 in a targeted fashion has been explored for treating several experimental autoimmune diseases (Karumuthil-Melethil et al, Id.; Fife et al., 2006, The Journal of clinical investigation 1 16: 2252-61 ; Li et al, 2007 ' , J Immunol 179: 5191-203; Vasu et al, 2003 , Int Immunol 15: 641- 54).

[0006] Although various pancreatic β-cell specific antigens have been described in the art, none of them is expressed on the cell surface plasma membranes. GLUT2 is a molecule that is expressed on pancreatic β-cell surfaces predominantly (although not completely) where it forms a part of "the glucose sensor" (Thorens et al, 1994, Biochem Soc Trans 22: 684-7). Encoded by the SLC2A2 gene in mammals, GLUT2 is a 524 amino acid multi-pass transmembrane carrier protein. Id. The longest stretch of extracellular domain is 67 amino acids long (UniProtKB/Swiss-Prot entry PI 1168). GLUT2 is involved in passive glucose transport while not being directly involved in disease pathogenesis and thus is an available target for specific binding to, inter alia, an antibody.

SUMMARY OF THE INVENTION

[0007] This invention as disclosed provides reagents and methods for treating and preventing Type 1 diabetes in an animal, particularly a human. The reagents of the invention comprise proteins that specifically bind to GLUT2 protein expressed on the cell surface of a pancreatic β-cell and CTLA-4 expressed on the cell surface of an activated T-cell. As set forth in greater detail herein, binding of the protein to both the GLUT2 and CTLA-4 induces Treg cells that down-regulate the autoimmune responses to pancreatic β-cells that are involved in the development of Type 1 diabetes.

[0008] In one embodiment, the invention as disclosed herein is a protein comprising a first polypeptide or polypeptide domain having a first specific binding activity for Cytotoxic T-lymphocyte Antigen 4 (CTLA-4) expressed on a T-cell cell surface and a second specific binding activity for Glucose Transporter 2 (GLUT2) or an extracellular ectodomain thereof expressed on a pancreatic β-cell cell surface, wherein binding of the first polypeptide or polypeptide domain to CTLA-4 induces a CTLA-4 specific agonist response in the T cell, and binding of the second polypeptide or polypeptide domain to GLUT2 or an ectodomain thereof does not inhibit GLUT2 glucose transporter function, wherein said agonist response in the T cell induces a response that reduces immunoreactivity against pancreatic β-cells. In certain embodiments, the invention as disclosed has a first and second polypeptide or polypeptide domain that are independently an antibody molecule or a specific -binding fragment thereof, and may futher have a first and second polypeptide or polypeptide domain independently comprising at least one immunoglobulin heavy chain and one immunoglobulin light chain. In additional embodiments, the first and second polypeptide or polypeptide domain independently comprise a monovalent antibody fragment, an F(ab) fragment, an F(ab)2 fragment, F(ab)' fragment , or an Fv fragment. The invention as disclosed herein may also have the first and second polypeptide or polypeptide domains covalently attached to one another by a linker moiety.

[0009] In another embodiment, the invention as disclosed herein is a pharmaceutical composition comprising the protein described above with a pharmaceutically acceptable carrier or adjuvant. In a certain embodiments, the pharmaceutical composition is used in methods for preventing diabetes in an animal comprising the step of administering to the animal a therapeutically effective amount of a pharmaceutical composition of the invention. In further embodiments, the methods are used for protecting pancreatic β-cells in an animal by administering to the animal a protective amount of the pharmaceutical composition.

[0010] In one embodiment, the invention as disclosed herein comprises methods for producing Treg cells that down-regulate autoimmune responses to pancreatic β-cells in an animal comprising the step of administering to the animal an effective amount of a pharmaceutical composition disclosed herein. In certain embodiments of the methods, the pancreatic tissue-specific T cells are down modulated and induced Treg cells are

predominantly pancreatic tissue-specific Treg cells.

[0011] In another embodiment, the invention as disclosed herein provides methods for producing immune tolerance to pancreatic β-cells in an animal comprising the step of administering to the animal an effective amount of a pharmaceutical composition as set forth herein. In certain embodiments of the methods, immune tolerance is pancreatic islet β-cell specific T cell tolerance.

Additional features and advantages are described herein, and will be apparent from the following Detailed Description, Drawings and the claims. BRIEF DESCRIPTION OF THE DRAWINGS

[0012] Figure 1 shows nickel nitriloacetic acid (Ni-NTA) affinity and reverse phase

HPLC purification of recombinant ecto-GLUT2 from E. coli lysate (upper panels) and Ni- NTA purification of recombinant full-length GLUT2 from HEK-293 cell lysate (lower panels).

[0013] Figure 2(a) shows fluorescence-activated cell sorting (FACS) analysis of three anti-GLUT mAbs disclosed herein binding to native GLUT2 expressed on Min-6 cells.

Figure 2(b) shows that anti-GLUT2 mAbs from D2, E7 and H5 hybridoma clones do not interfere with GLUT2 function as a glucose sensor.

[0014] Figure 3(a) shows that anti-GLUT2, anti-CTLA-4 protein is capable of binding to pancreatic islets in mice. Figure 3(b) shows that this protein is able to detect

GLUT2 in the membrane fraction of Min-6 (mouse insulinoma cells).

[0015] Figure 4 shows FACS analysis indicating that anti-GLUT2, anti-CTLA-4 protein is capable of binding CTLA-4 on CD4 + T-cells.

[0016] Figure 5 shows FACS analysis indicating that anti-GLUT2, anti-CTLA-4 protein is capable of binding to GLUT2 on Min-6 cells.

[0017] Figure 6 shows that anti-GLUT2, anti-CTLA-4 protein is capable of binding to insulin producing islet cells in vivo.

[0018] Figure 7(a)-(d) shows that treatment of NOD mice with anti-GLUT2, anti- CTLA-4 protein did not cause interference of liver or kidney functions.

[0019] Figure 8 shows effects of biweekly anti-GLUT2, anti-CTLA-4 protein treatment of NOD mice started at age 10 weeks.

[0020] Figure 9 shows effects of biweekly anti-GLUT2, anti-CTLA-4 protein treatment of NOD mice started at age 8 weeks.

[0021] Figure 10 shows effects of biweekly anti-GLUT2, anti-CTLA-4 protein treatment of NOD mice started at age 6 weeks.

[0022] Figure 11 shows a comparison between anti-hamster IgG response in 8 week and 6 week anti-GLUT2, anti-CTLA-4 protein treatment in NOD mice.

[0023] Figure 12 shows FACS analysis showing increased Foxp3+ regulatory T cells in anti-GLUT2, anti-CTLA-4 protein-treated (T-Glut2-CTLA-4) NOD mice relative to mice that were left untreated or treated with a control antibody (C-Ab).

[0024] Figure 13 shows FACS analysis showing reduced proliferation of effector T cells in response to pancreatic β-cell antigens in the presence of Tregs from anti-GLUT2, anti-CTLA-4 protein-treated NOD mice (T-Glut2-CTLA-4 (Low), T-Glut2-CTLA-4 (High)) relative to Tregs from untreated and C-Ab treated mice. The lower panel also shows increased Tregs in cultures of cells in vitro from T-Glut2-CTLA-4 treated NOD mice relative to the controls.

[0025] Figure 14 shows hemtoxylin and eosin stained sections of pancreas from untreated, control Ab-treated and therapeutic T-Glut2-CTLA4 treated mice. Islets from untreated and control Ab treated mice show considerable lymphocytic infiltration, while the islet from T-Glut2-CTLA4 treated mice shows very little or no infiltration into the islet. DETAILED DESCRIPTION

[0026] Provided herein are reagents and methods for treating or preventing Type 1

(insulin-dependent) diabetes (T1D) in a mammal, most particularly a human. The invention provides proteins made up of two different specific binding domains, including in certain embodiments monoclonal antibodies or specifc -binding fragments thereof, wherein one specific -binding domain binds to Glucose transporter 2 (GLUT2) expressed predominantly on pancreatic β-cells, while the other specific binding domain binds to Cytotoxic T- Lymphocyte Antigen 4 (CTLA-4), a cell surface molecule on activated T-cells. Also provided are pharmaceutical compositions of the disclosed proteins and methods for using said pharmaceutical compositions for administration to mammals, particularly humans, for therapy or prophyllaxis of T ID.

[0027] Without being bound to any particular description regarding mechanism of action, it is envisioned that, upon binding to pancreatic β-cells with the targeting (GLUT2- specific) arm, the anti-CTLA-4 arm is available to bind to activated antigen-specific T-cells that localize to the pancreatic islets. Binding of a protein of the invention can suppress effector (pathogenic) T-cell function, thereby inducing Tregs in a β-cell antigen-specific manner and overcoming the limitations of generalized immunosuppressants that have been used for treating other diseases and disorders having an autoimmunity component. Earlier studies by the inventors have shown that concurrent enhanced engagement of T-cell receptor and CTLA-4 on activated T-cells leads to induction of antigen-specific Tregs (Karumuthil- Melethil et al, Id.; Li et al. , 2007 ' , J Immunol 179: 5191-203; Vasu et al, 2003, Int Immunol 15: 641-54). As disclosed herein, anti-GLUT2/anti-CTLA-4 proteins of the invention can bind simultaneously to GLUT2 expressed on β-cells and engage CTLA-4 expressed on islet- infiltrating T-cells, resulting in down-modulation of β-cell specific T-cell responses, thereby inducing Tregs that can suppress autoimmunity in TID.

[0028] CTLA-4 engagement down-regulates T-cell responses through different mechanisms including induction of IL-10 and TGF-β (Karumuthil-Melethil et ah, Id.).

Therefore, targeted engagement of CTLA-4 on activated T-cells can be used to generate antigen/tissue specific Tregs for, inter alia, the treatment of diabetes. Disclosed herein is a novel approach to engage CTLA-4 on islets-infiltrating T-cells by, in certain embodiments, β-cell surface bound CTLA-4 specific agonistic antibodies. Since GLUT2 is predominantly found on pancreatic β-cells, this marker is capable of selectively anchoring to pancreatic β- cells molecules that specifically bind to CTLA-4, in certain embodiments being anti-CTLA-4 antibodies, onto the pancreatic β-cell surface. It is understood in the art that most activated autoreactive T-cells in TID reside in the pancreatic microenvironment. Therefore, pancreatic islet specific T-cell tolerance can be induced by administration of a protein of the invention. Advantageously, minimal and transient non-tissue specific protein binding and related side effects are produced because CTLA-4 is expressed at a very low level on resting T-cells. In addition, any such binding would be, in most instances, remote from any ongoing T-cell receptor engagement and would not inhibit normal immune surveillance. Hence, the methods disclosed herein provide an advantageous alternative to current experimental therapies that lack specificity (such as anti-CD3, anti-CD20, etc.).

[0029] The section headings are used herein for organizational purposes only, and are not to be construed as in any way limiting the subject matter described. The invention as disclosed herein is not limited to the particular methodology, protocols, cell lines, vectors, or reagents described herein because they may vary without departing from the spirit and scope of the invention.

[0030] Conventional and standard techniques may be used for recombinant DNA molecule, protein, and antibody production, as well as for tissue culture and cell

transformation. Enzymatic reactions and purification techniques are typically performed according to the manufacturer's specifications or as commonly accomplished in the art using conventional procedures known in the art, or as described herein. Unless specific definitions are provided, the nomenclature utilized in connection with, and the laboratory procedures and techniques of analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques may be used for chemical syntheses, chemical analyses,

pharmaceutical preparation, formulation, and delivery, and treatment of patients.

[0031 ] Further, the terminology used herein is for the purpose of exemplifying particular embodiments only and is not intended to limit the scope of the invention as disclosed herein. Any method and material similar or equivalent to those described herein can be used in the practice of the invention as disclosed herein and only exemplary methods, devices, and materials are described herein.

[0032] All patents and publications mentioned herein are incorporated by reference in their entirety for the purpose of describing and disclosing the proteins, enzymes, vectors, host T-cells, and methodologies reported therein that might be used with and in the invention as disclosed herein. However, nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.

1. Definitions

[0033] As utilized in accordance with the present disclosure, the following terms unless otherwise indicated, shall be understood to have the following meanings:

[0034] The term "antibody" is used in the broadest sense, and specifically covers monoclonal antibodies (including full-length monoclonal antibodies), polyclonal antibodies, antibody fragments, or synthetic polypeptides carrying one or more CDR or CDR-derived sequences so long as the polypeptides exhibit the desired biological activity. Antibodies

(Abs) and immunoglobulins (Igs) are glycoproteins having the same structural characteristics. Generally, antibodies are considered Igs with a defined or recognized specificity. Thus, while antibodies exhibit binding specificity to a specific target, immunoglobulins include both antibodies and other antibody-like molecules that lack target specificity. The antibodies of the invention can be of any class (e.g., IgG, IgE, IgM, IgD, IgA and so on), or subclass (e.g., IgGi, IgG 2 , IgG 2a , IgG 3 , IgG 4 , IgAi, IgA 2 and so on) ("type" and "class," and "subtype" and "subclass," are used interchangeably herein). Native or wildtype (obtained from a non-artificially manipulated member of a population) antibodies and immunoglobulins are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at the other end. The term "non- artificially manipulated" means not treated to contain or express a foreign antigen-binding molecule. Wildtype can refer to the most prevalent allele or species found in a population or to the antibody obtained from a non-manipulated animal, as compared to an allele or polymorphism, or a variant or derivative obtained by a form of manipulation, such as mutagenesis, use of recombinant methods and so on to change an amino acid of the antigen- binding molecule.

[0035] As used herein, "anti-CTLA-4 antibody" means an antibody or polypeptide derived therefrom (a derivative) that binds specifically to CTLA-4 as defined herein, including, but not limited to, molecules which inhibit or substantially reduce the binding of CTLA-4 to its receptor or inhibit CTLA-4 activity.

[0036] As used herein, "anti-GLUT2 antibody" means an antibody or polypeptide derived therefrom (a derivative) that binds specifically to GLUT2 as defined herein. In certain embodiments, the term includes molecules that do not inhibit or substantially GLUT2 activity.

[0037] As used herein, in certain embodiments "protein" means a first polypeptide or polypeptide domain having a first specific binding activity for Cytotoxic T-lymphocyte

Antigen 4 (CTLA-4) expressed on a T-cell cell surface and a second specific binding activity for Glucose Transporter 2 (GLUT2) or an extracellular ectodomain thereof expressed on a pancreatic β-cell cell surface.

[0038] "Agonist" or "agonist activity" refers to the ability of an anti-CTLA-4 antibody to induce signaling through CTLA-4 expressed on activated T cells that results in down modulation of T-cell response, and induction of T-reg cells that can down- modulate or suppress the activity of other effector T-cells.

[0039] "Antagonist" refers to a molecule capable of inhibiting one or more biological activities of a target molecule, such as signaling by CTLA-4. Antagonists may interfere with the binding of a receptor to a ligand and vice versa, by incapacitating or killing cells activated by a ligand, and/or by interfering with receptor or ligand activation (e.g., tyrosine kinase activation) or signal transduction after ligand binding to a receptor. The antagonist may completely block receptor-ligand interactions or may substantially reduce such interactions.

[0040] The term "antigen" as used herein refers to a molecule or a portion of a molecule capable of being bound by the antibodies of the invention. An antigen can have one or more than one epitope.

[0041] The term "antigen binding domain," "antigen binding site," or "antigen binding region" refers to that portion of a protein molecule, in particular an antibody, which contains the specific amino acid residues (or other moieties) that interact with an antigen and confer on the binding agent its specificity and affinity for the antigen. In an antibody, the antigen-binding domain is commonly referred to as the "complementarity-determining region" (or "CDR"). Where an antigen is large, an antibody may only bind to a particular part of the antigen, which part is called an epitope. An antigen-binding domain may be provided by one or more antibody variable domains. In certain aspects, an antigen-binding domain is made of the association of an antibody light chain variable domain (VL) and an antibody heavy chain variable domain (VH).

[0042] The term "epitope" refers to that portion of any molecule capable of being recognized by and bound by an antibody, at one or more of the binding agent's antigen binding regions. Epitopes usually consist of chemically active surface groupings of molecules, such as for example, amino acids or carbohydrate side chains, and have specific three-dimensional structural characteristics as well as specific charge characteristics.

Epitopes as used herein may be contiguous or non-contiguous. Moreover, epitopes may be mimetic in that they comprise a three-dimensional structure that is identical to the epitope used to generate the antibody, yet comprise none or only some of the amino acid residues found in the antigen molecule (in particular, CTLA-4 or GLUT2) used to stimulate the antibody immune response.

[0043] The term "antibody fragment" refers to a portion of an intact or a full-length chain or an antibody, generally comprising a target binding or variable region. Examples of antibody fragments include, but are not limited to, Fab, Fab', F(ab')2 and Fv fragments. A "functional fragment" or "analog of an anti-CTLA-4/GLUT2 antibody" is one that can prevent or substantially reduce the ability of the receptor to bind to a ligand or to initiate signaling. As used herein, the term "functional fragment" generally is synonymous with "antibody fragment," and with respect to antibodies, can refer to fragments such as Fv, Fab, F(ab')2, and so on which can prevent or substantially reduce the ability of the receptor to bind to a ligand or to initiate signaling. An "Fv" fragment consists of a dimer of one heavy and one light chain variable domain in a non-covalent association (VH-VL dimer). In that configuration, the three CDRs of each variable domain interact to define a target binding site on the surface of the VH-VL dimer, as in an intact antibody. Collectively, the six CDRs confer target binding specificity on the intact antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for a target) can have the ability to recognize and to bind target. The Fab fragment contains the variable and constant domains of the light chain and the variable and first constant domain (CHI) of the heavy chain. Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxyl terminus of the CHI domain to include one or more cysteines from the antibody hinge region. Fab' fragments can be produced by cleavage of the disulfide bond at the hinge cysteines of the F(ab')2 pepsin digestion product. Additional enzymatic and chemical treatments of antibodies can yield other functional fragments of interest.

[0044] The term "autoimmune disease" as used herein refers to a non-malignant disease or disorder arising from and directed against an individual's own tissues. In certain embodiments, the autoimmune disease or disorder of the invention includes, but is not limited to, diabetes mellitus, specifically Type I diabetes mellitus or insulin dependent diabetes mellitus.

[0045] The term, "carrier" refers to a diluent, adjuvant, excipient or vehicle with which the therapeutic is administered. Such physiological carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a suitable carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions also can be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. The compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations, depots, and the like. Such compositions will contain an effective amount of the antibody, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. As known in the art, specific formulations are constructed to suit the mode of administration.

[0046] The terms "cell," "cell line," and "cell culture" include progeny thereof. It is also understood that all progeny may not be precisely identical, such as in DNA content, due to deliberate or inadvertent mutation. Variant progeny that have the same function or biological property of interest, as screened for in the original cell, are included. [0047] The term "CDR grafted antibody" refers to an antibody in which the CDR from one antibody of a particular species or isotype is recombinantly inserted into the framework of another antibody of the same or different species or isotype.

[0048] "Derivatives" include those proteins and particularly antibodies that have been chemically modified in some manner distinct from insertion, deletion, or substitution variants.

[0049] As used herein, the terms "effective amount" and "therapeutically effective amount" refer to the amount of a therapy (e.g., a prophylactic or therapeutic agent), which is sufficient to reduce the severity and/or duration of TID, ameliorate one or more symptoms thereof, prevent the advancement of TID or cause regression of TID, or which is sufficient to result in the prevention of the development, recurrence, onset, or progression of TID or one or more symptoms thereof, or enhance or improve the prophylactic and/or therapeutic effects of another therapy (e.g., another therapeutic agent) useful for treating TID.

[0050] The term "expression vector" refers to a plasmid, phage, virus, or other recombinant genetic vector, for expressing a polypeptide from a DNA (or in some instances an R A) molecule. An expression vector can comprise a transcriptional unit comprising an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, promoters, or enhancers, (2) a structural genetic component or sequence that encodes the binding agent which is transcribed into mRNA and translated into protein, and (3) appropriate transcription initiation and termination sequences. Structural genetic components intended for use in yeast or eukaryotic expression systems preferably include a leader sequence enabling extracellular secretion of translated protein by a host T-cell.

Alternatively, where a recombinant antibody is expressed without a leader or transport sequence, it may include an amino terminal methionine residue. This residue may or may not be subsequently cleaved from the expressed recombinant protein to provide a final antibody product.

[0051 ] Also included within the scope of the invention are functional equivalents of antibody -comprising embodiments of the proteins disclosed herein. The term "functional equivalents" includes antibodies with homologous sequences, antibody homologs, chimeric antibodies, artificial antibodies and modified antibodies, for example, wherein each functional equivalent is defined by the ability to bind to CTLA-4 and/or GLUT2, inhibiting CTLA-4 signaling ability or function, or inhibiting binding of CTLA-4 to its receptor. The skilled artisan will understand that there is an overlap in the group of molecules termed "antibody fragments" and the group termed "functional equivalents." Methods of producing functional equivalents that retain CTLA-4 binding ability are known to the person skilled in the art.

[0052] The terms "functional fragment," "variant," "derivative," "analog," and the like, as well as forms thereof, of an antibody or antigen refer to a compound or molecule having qualitative biological activity in common with a full-length antibody or antigen of interest. For example, a functional fragment or analog of an anti- CTLA-4 and/or GLUT2 antibody is one that can bind to a CTLA-4 and/or GLUT2 molecule or one that can prevent or substantially reduce the ability of a ligand, or an agonistic or antagonistic antibody, to bind to CTLA-4 and/or GLUT2.

[0053] The term "heavy chain" when used in reference to an antibody collectively refers to five distinct types, called alpha, delta, epsilon, gamma, and mu, based on the amino acid sequence of the heavy chain constant domain. The combination of heavy and light chains give rise to five known classes of antibodies: IgA, IgD, IgE, IgG, and IgM, respectively, including four known subclasses of IgG, designated as IgGi, IgG 2 , IgG 3 , and IgG 4 .

[0054] The term "hinge" or "hinge region" as used herein refers to the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody.

[0055] "Antibody homolog" or "homolog" refers to any molecule which specifically binds to a particular antigen as taught herein and in particular CTLA-4 or GLUT2. Thus, an antibody homolog includes native or recombinant antibodies, whether modified or not, portions of antibodies (such as an Fab or Fv molecule) that retain the biological properties of interest, such as binding to CTLA-4 or GLUT2, a single chain antibody, a polypeptide carrying one or more CDR regions and so on. The amino acid sequence of the homolog need not be identical to that of the naturally occurring antibody but can be altered or modified to carry substitute amino acids, inserted amino acids, deleted amino acids, amino acids other than the twenty normally found in proteins, and so on to obtain a polypeptide with enhanced or other beneficial properties.

[0056] The terms "identity" or "homology" refer to the percentage of nucleotide bases or amino acid residues in the candidate sequence that are identical with the residue of a corresponding sequence to which it is compared after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity for the entire sequence, and not considering any conservative substitutions as part of the sequence identity. Neither N- terminal or C-terminal extensions nor insertions shall be construed as reducing identity or homology. Methods and computer programs for the alignment are available and well known in the art. Sequence identity may be measured using sequence analysis software.

[0057] The term "inhibiting and/or neutralizing epitope" is an epitope which when bound by an antibody, results in the loss of (or at least the decrease in) biological activity of the molecule, cell, or organism containing such epitope, in vivo, in vitro, or in situ. In the context of the invention disclosed herein, the neutralizing epitope is located on or is associated with a biologically active region of CTLA-4 or GLUT2. Alternatively, the term "activating epitope" is an epitope, which when bound by an antibody of the invention, such as an antibody, results in activation, or at least maintenance of a biologically active

conformation, of CTLA-4 or GLUT2.

[0058] The term "isolated" when used in relation to proteins and particularly antibodies of the invention refers to a compound that is free from at least one contaminating polypeptide or compound that is found in its natural environment, and preferably

substantially free from any other contaminating mammalian polypeptides that would interfere with its therapeutic or diagnostic use.

[0059] An "isolated" or "purified" protein and particular an antibody is substantially free of cellular material or other contaminating proteins from the cell or tissue source or medium from which the protein is derived, or substantially free of chemical precursors or other chemicals when chemically synthesized. Generally, "purified" will refer to a protein and particular an antibody composition that has been subjected to fractionation to remove various other components, and which composition substantially retains its expressed biological activity. The language "substantially free of cellular material" includes preparations of a protein and particular an antibody in which the polypeptide is separated from cellular components of the cells from which a protein and particular an antibody is isolated or recombinantly produced. Thus, a protein and particular an antibody that is substantially free of cellular material includes preparations of the protein and particular antibody having less than about 30%, 20%, 10%, 5%, 2.5%, or 1% (by dry weight) of contaminating protein. Where the term "substantially purified" is used, this designation will refer to an antibody composition in which the protein and particular antibody forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, or more of the proteins in the composition. [0060] When the protein and particular an antibody is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, 10%, 5%, 2.5%, or 1% of the volume of the protein preparation. When a protein and particular an antibody is produced by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals and reagents, i.e., the protein and particular antibody of interest is separated from chemical precursors or other chemicals that are involved in the synthesis of the protein. Accordingly, such preparations of the protein and particular antibody have less than about 30%, 20%, 10%, 5%, or 1% (by dry weight) of chemical precursors or compounds other than antibody of interest. In a preferred

embodiment of the invention, proteins and particularly antibodies are isolated or purified.

[0061] The proteins and particular antibodies of interest can be screened or can be used in an assay as described herein or as known in the art. Often, such assays require a reagent to be detectable, which is, for example, labeled. The word "label" when used herein refers to a detectable compound or composition that can be conjugated directly or indirectly to a molecule or protein, e.g., an antibody. The label may itself be detectable (e.g., radioisotope labels, particles, or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition that is detectable.

[0062] The term "light chain" when used in reference to an antibody collectively refers to two distinct types, called kappa (k) or lambda (1) based on the amino acid sequence of the constant domains.

[0063] The term "linear Fab" refers to a tetravalent antibody. The "linear Fab" is composed of a tandem of the same CHI-VH domain, paired with the identical light chain at each CHI-VH position.

[0064] The term "linker" as used herein refers to a molecule and particularly a peptide adapted to connect the variable domains of the protein and particularly antibody constructs of the invention. A peptide linker may contain one or a plurality of any amino acids, the amino acids glycine (G) and serine (S) being preferred. The linkers may be equal or differ from each other between and, when used with antibodies within the heavy chain polypeptide and the light chain polypeptide. Furthermore, the linker may have a length of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids. A preferred peptide linker unit for antibody heavy chain and light chain domains is GGGGS (SEQ ID NO.: 1). The numbers of linker units of the heavy chain and of the light chain may be equal (symmetrical order) or differ from each other (asymmetrical order). A peptide linker is preferably long enough to provide an adequate degree of flexibility to prevent the protein and particularly antibody moieties from interfering with each other's activity, for example by steric hindrance, to allow for proper protein folding and, if necessary, to allow the molecules to interact with two or more, possibly widely spaced, receptors on the same cell; yet it is preferably short enough to allow moieties such as the antibody moieties to remain stable in the cell.

[0065] As used herein, the phrase "low to undetectable levels of aggregation" refers to samples containing no more than 5%, no more than 4%, no more than 3%, no more than 2%, no more than 1%, and often no more than 0.5% aggregation, by weight protein, as measured by, for example, high performance size exclusion chromatography (HPSEC).

[0066] As used herein, the term "low to undetectable levels of fragmentation" refers to samples containing equal to or more than 80%, 85%, 90%, 95%, 98%, or 99%, of the total protein, for example, in a single peak, as determined by HPSEC, or in two (2) peaks (heavy chain and light chain) as determined by, for example, reduced capillary gel electrophoresis (rCGE), and containing no other single peaks having more than 5%, more than 4%, more than 3%, more than 2%, more than 1%, or more than 0.5% of the total protein, each. rCGE as used herein refers to capillary gel electrophoresis under reducing conditions sufficient to reduce disulfide bonds in an antibody or antibody-type or derived molecule.

[0067] "Animal" for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals, non-human primates, and zoo, sports or pet animals, such as dogs, horses, cats, and cows. In particular, NOD mice represent a well-established mouse model of TID that resembles many features of human TID including lymphocytic infiltration into pancreatic tissue with eventual β-cell destruction resulting in hyperglycemia, abrupt onset of overt diabetes, ketosis proneness, absence of obesity and the dependence on exogenous insulin to sustain life (Todd and Wicker, 2001, Immunity 15: 387-95; Tang et ah, 2006, Nature immunology 7: 83-92; Trembleau et ah, 1995, The Journal of experimental medicine, 181 : 817-21). Certain evidence known in the art suggests that regulatory T (Treg) cells control the progression of diabetes (Chen et ah, 2005, J Exp Med 202: 1387-97.

[0068] The term "monoclonal antibodies" refers to a collection of antibodies encoded by the same nucleic acid molecule(s), which are optionally produced by a single hybridoma or other cell line or by a transgenic mammal such that each monoclonal antibody will typically recognize the same epitope on the antigen. The term "monoclonal" is not limited to any particular method for making the antibody, nor is the term limited to antibodies produced in a particular species, e.g., mouse and rat. Monoclonal antibodies are highly specific, being directed against a single target site, epitope, or determinant. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes) of an antigen, each monoclonal antibody is directed against a single determinant on the target. In addition to their specificity, monoclonal antibodies are advantageous by being synthesized by a host cell, uncontaminated by other immunoglobulins, which provides for cloning the relevant gene and mRNA encoding the antibody chains thereof. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies for use with the invention as disclosed herein may be isolated from phage antibody libraries using well-known techniques or can be purified from a polyclonal preparation, as well as by conventional hybridoma technologies. The parent monoclonal antibodies to be used in accordance with the invention may be made by classic hybridoma methods or may be made by recombinant techniques.

[0069] The term "naturally occurring" when used in connection with biological materials such as nucleic acid molecules, proteins and polypeptides, host cells, and the like, refers to those which are found in nature and not modified by a human being.

[0070] The term "pharmaceutical composition" as used herein refers to formulations of various preparations for administration to an animal, particularly a human. The formulations containing therapeutically effective amounts of the proteins, particularly antibodies of the invention are sterile liquid solutions, liquid suspensions, or lyophilized versions, and optionally contain stabilizers or excipients.

[0071 ] Proteins of the invention, including certain embodiments comprising antibodies or antigen-binding fragments thereof, may be provided in pharmaceutically acceptable compositions as known in the art or as described herein. The term

"physiologically acceptable," "pharmacologically acceptable," and so on mean approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals and more particularly in humans.

[0072] "Single-chain Fv," "sFv," or "scAb" antibody fragments comprise the VH and

VL domains of an antibody, wherein these domains are present in a single polypeptide chain. Generally, the Fv polypeptide further comprises a polypeptide linker, often a flexible molecule, between the VH and VL domains, which enables the sFv to form the desired structure for target binding.

[0073] "Specifically binds CTLA-4" or "specifically binds GLUT2" refers to the ability of a specific binding agent (such as an antibody or fragment thereof) of the invention to recognize and bind mature, full-length or partial-length mouse CTLA-4 or GLUT2 polypeptide, or an ortholog thereof, such that its affinity (as determined by, e.g., affinity ELISA or BIAcore assays as described herein) or its neutralization capability (as determined by, e.g., neutralization ELISA assays described herein, or similar assays) is at least 10 times as great, but optionally 50 times as great, 100, 250, or 500 times as great, or even at least 1000 times as great as the affinity or neutralization capability of the same for any other interleukin or other peptide or polypeptide.

[0074] The phrase "substantially identical" with respect to an antibody chain polypeptide sequence may be construed as an antibody chain exhibiting at least 70%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to the reference polypeptide sequence. The term with respect to a nucleic acid sequence may be construed as a sequence of nucleotides exhibiting at least about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more sequence identity to the reference nucleic acid sequence.

[0075] "Substitutional" variants are those that have at least one amino acid residue in a native sequence removed and replaced with a different amino acid inserted in its place at the same position. The substitutions may be single, where only one amino acid in the molecule is substituted, or may be multiple, where two or more amino acids are substituted in the same molecule. The plural substitutions may be at consecutive sites. Also, one amino acid can be replaced with plural residues, in which case such a variant comprises both a substitution and an insertion. "Insertional" variants are those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a native sequence. Immediately adjacent to an amino acid means connected to either the a-carboxyl or a-amino functional group of the amino acid. "Deletional" variants are those with one or more amino acids in the native amino acid sequence removed. Ordinarily, deletional variants will have one or two amino acids deleted in a particular region of the molecule.

[0076] The term "treatment" as used herein refers to both therapeutic treatment and prophylactic or preventative measures. It refers to preventing, curing, reversing, attenuating, alleviating, minimizing, suppressing, or halting the deleterious effects of a disease state, disease progression, disease causative agent (e.g., bacteria or viruses), or other abnormal condition.

[0077] The term "variants," as used herein, include those polypeptides wherein amino acid residues are inserted into, deleted from, and/or substituted into the naturally occurring (or at least a known) amino acid sequence for the binding agent. Variants of the invention include fusion proteins as described below.

[0078] The term "variable" in the context of a variable domain of antibodies refers to certain portions of the pertinent molecule which differ extensively in sequence between and among antibodies and are used in the specific recognition and binding of a particular antibody for its particular target. However, the variability is not evenly distributed through the variable domains of antibodies. The variability is concentrated in three segments called complementarity determining regions (CDRs; i.e., CDR1, CDR2, and CDR3) also known as hypervariable regions, both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework (FR) regions or sequences. The variable domains of native heavy and light chains each comprise four FR regions, largely adopting a β-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the β-sheet structure. The CDRs in each chain are held together often in proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the target (epitope or determinant) binding site of antibodies.

2. Antibody Types and Components

[0079] Antibodies and antibody fragments that specifically bind CTLA-4 and GLUT2 polypeptides are within the scope of the invention disclosed herein. The antibodies may be monoclonal (mAbs), recombinant, chimeric, or humanized embodiments thereof such as CDR-grafted antibodies, or human, single chain, catalytic, multi-specific and/or bi-specific embodiments thereof, as well as fragments, variants, and/or derivatives thereof.

[0080] Alternatively, transgenic animals (e.g., mice) that are capable of producing a repertoire of antibodies in the absence of endogenous immunoglobulin production can be used to generate such antibodies. This can be accomplished by immunization of the animal with a CTLA-4 or GLUT2 antigen or fragments thereof where the CTLA-4 or GLUT2 fragments have an amino acid sequence that is unique to CTLA-4 or GLUT2. Such immunogens can be optionally conjugated to a carrier. Antibodies may also be produced by the expression of recombinant DNA in host-cells or by expression in hybridoma cells as described herein.

[0081] Antibodies of the invention may be described or specified in terms of the epitopes or portions of CTLA-4 or GLUT2 that the antibody recognizes or specifically binds. The epitopes or polypeptide portions may be specified as described herein, e.g., by N- terminal and C-terminal positions, by size in contiguous amino acid residues, conformational epitopes and so on.

[0082] Antibody fragments that recognize specific epitopes may be generated by known techniques. Traditionally, these fragments were derived via proteolytic digestion of intact antibodies. For example, Fab and F(ab¾ fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments). F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain. However, those fragments can be produced directly by recombinant host cells. For example, the antibody fragments can be isolated from an antibody phage library. According to another approach, F(ab¾ fragments can be isolated directly from recombinant host cell culture. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner. In other embodiments, the antibody of the invention is a single chain Fv fragment (Fv).

[0083] To determine whether a particular antibody homo log binds to a CTLA-4 or

GLUT2 polypeptide, any conventional binding assay may be used. Useful binding assays include FACS analysis, ELISA assays, Surface Plasmon Resonance, radioimmunoassays, and the like, which detect antibody binding and functions resulting therefrom, to a CTLA-4 or GLUT2 polypeptide. Full-length and soluble forms of CTLA-4 or GLUT2 polypeptides are useful in such assays. The binding of an antibody or homolog to CTLA-4 or GLUT2, or to soluble fragments thereof, may conveniently be detected through the use of a second antibody specific for immunoglobulins of the species from which the antibody or homolog is derived.

[0084] To determine whether a particular antibody or homolog significantly blocks binding to CTLA-4 or GLUT2, any suitable competition assay may be used. Useful assays include, for example, ELISA assays, FACS assays, radioimmunoassays, and the like that quantify the ability of the antibody or homolog to compete with CTLA-4 or GLUT2. 3. Variants of Antibodies

[0085] Variants of antibodies of the invention include insertion, deletion, and/or substitution variants. In one aspect of the invention, insertion variants are provided wherein one or more amino acid residues supplement an antibody amino acid sequence. Insertions may be located at either or both termini of the protein, or may be positioned within internal regions of the antibody amino acid sequence. Insertional variants with additional residues at either or both termini can include, for example, fusion proteins and proteins including amino acid tags or labels. Insertion variants also include antibody polypeptides wherein one or more amino acid residues are added to an antibody amino acid sequence, or fragment thereof.

[0086] In still another aspect, the invention provides substitution variants of proteins particularly antibodies of the invention. Substitution variants are generally considered to be "similar" to the original polypeptide or to have a certain "percent identity" to the original polypeptide, and include those polypeptides wherein one or more amino acid residues of a polypeptide are removed and replaced with alternative residues. Relating to a CTLA-4 or GLUT2 antibody in particular, percent identity refers to percent identity outside of the complimentary determining regions of the antibody. In one aspect, the substitutions are conservative in nature; however, the invention embraces substitutions that are also non- conservative.

[0087] Antibody fragments include those portions of the antibody that bind to an epitope on the antigen polypeptide. Examples of such fragments include Fab and F(ab')2 fragments generated, for example, by enzymatic or chemical cleavage of full-length antibodies. Other binding fragments include those generated by recombinant DNA techniques, such as the expression of recombinant plasmids containing nucleic acid sequences encoding antibody variable regions. The invention also embraces polypeptide fragments of a CTLA-4 or GLUT2 antibody wherein the fragments maintain the ability to specifically bind a CTLA-4 or GLUT2 polypeptide. Fragments comprising at least 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 or more consecutive amino acids of a peptide or polypeptide of the invention are comprehended herein. Preferred polypeptide fragments display specific binding and particularly

immunological properties unique to or specific for the antigen-binding agent of the invention. Fragments of the invention having the desired specific binding and particularly

immunological properties can be prepared by any of the methods well known and routinely practiced in the art. [0088] The antibody fragments and functional equivalents comprising certain

embodiments of the invention as disclsosed herein encompass those molecules with a detectable degree of specific binding to CTLA-4 or GLUT2. A detectable degree of binding includes all values in the range of at least 10-100%, preferably at least 50%, 60% or 70%, more preferably at least 75%, 80%, 85%, 90%, 95% or 99% of the binding ability of an antibody of interest. Also included are equivalents with an affinity greater than 100% that of an antibody of interest.

[0089] The CDRs generally are of importance for epitope recognition and antibody binding. However, in certain instances changes may be made to residues that comprise the CDRs without interfering with the ability of the antibody to recognize and to bind the cognate epitope. For example, changes that do not impact epitope recognition, yet increase the binding affinity of the antibody for the epitope, can be made. Several studies have surveyed the effects of introducing one or more amino acid changes at various positions in the sequence of an antibody, based on the knowledge of the primary antibody sequence, on the properties thereof, such as binding and level of expression.

[0090] Thus, equivalents of an antibody of interest can be generated by changing the sequences of the heavy and light chain genes in the CDR1, CDR2 or CDR3, or framework regions, using methods such as oligonucleotide-mediated site-directed mutagenesis, cassette mutagenesis, error-prone PCR, DNA shuffling or mutator-strains of E. coli. The methods of changing the nucleic acid sequence of the primary antibody can result in antibodies with improved binding specificity, for example.

[0091] Antibodies with homologous sequences are those antibodies with amino acid sequences that have sequence identity with the amino acid sequence of a CTLA-4 or GLUT2 antibody of the invention, particularly in one, two or three CDR portions of the protein amino acid sequence. Preferably, identity homology is with the amino acid sequence of the variable regions of an antibody of the invention. "Sequence identity" as applied to an amino acid sequence herein is defined as a sequence with at least about 90%, 91%, 92%, 93%, 94% or more sequence identity, and more preferably at least about 95%, 96%, 97%, 98% or 99% sequence identity to another amino acid sequence, as determined, for example, by the.

[0092] Preferred methods to determine the relatedness or percent identity of two polypeptides are designed to give the largest match between the sequences tested. Methods to determine identity are described in publicly available computer programs. Preferred computer program methods to determine identity between two sequences include, but are not limited to, the GCG program package, including GAP (Genetics Computer Group, University of Wisconsin, Madison, WI), BLASTP, BLASTN, and FASTA. The BLASTX program is publicly available from the National Center for Biotechnology Information (NCBI) and other sources. The well-known Smith Waterman algorithm may also be used to determine identity.

[0093] As used herein, the twenty conventional amino acids and their abbreviations follow conventional usage.

[0094] The amino acids may have either L or D stereochemistry (except for Gly, which is neither L nor D) and the polypeptides and compositions of the invention may comprise a combination of stereochemistries. However, the L stereochemistry is preferred.

[0095] Stereoisomers (e.g., D-amino acids) of the twenty conventional amino acids, unnatural amino acids such as α-, α-disubstituted amino acids, N-alkyl amino acids, lactic acid, and other unconventional amino acids may also be suitable components for

polypeptides of the invention. Examples of unconventional amino acids include, without limitation: aminoadipic acid, beta-alanine, beta-aminopropionic acid, aminobutyric acid, piperidinic acid, aminocaprioic acid, aminoheptanoic acid, aminoisobutyric acid,

aminopimelic acid, diaminobutyric acid, desmosine, diaminopimelic acid, diaminopropionic acid, N-ethylglycine, N-ethylaspargine, hyroxylysine, allo-hydroxylysine, hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarcosine, N-methylisoleucine, N- methylvaline, norvaline, norleucine, orithine, 4-hydroxyproline, γ-carboxyglutamate, ε- Ν,Ν,Ν-trimethyllysine, ε-Ν-acetyllysine, O-phosphoserine, N-acetylserine, N- formylmethionine, 3-methylhistidine, 5 -hydroxy lysine, σ-Ν-methylarginine, and other similar amino acids and amino acids (e.g., 4-hydroxyproline).

[0096] Similarly, unless specified otherwise, the left-hand end of single-stranded polynucleotide sequences is the 5' end; the left-hand direction of double-stranded

polynucleotide sequences is referred to as the 5' direction. The direction of 5' to 3' addition of nascent RNA transcripts is referred to as the transcription direction; sequence regions on the DNA strand having the same sequence as the RNA and which are 5' to the 5' end of the RNA transcript are referred to as "upstream sequences"; sequence regions on the DNA strand having the same sequence as the RNA and which are 3' to the 3' end of the RNA transcript are referred to as "downstream sequences."

[0097] Conservative amino acid substitutions may encompass non-naturally occurring amino acid residues, which are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems. These include peptidomimetics and other reversed or inverted forms of amino acid moieties.

[0098] Even more substantial modification in an antibody range and presentation of biological properties can be accomplished by selecting an amino acid that differs more substantially in properties from that normally resident at a site. Thus, such a substitution can be made while maintaining: (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation; (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.

[0099] Naturally occurring residues may be divided into classes based on common side chain properties:

1) hydrophobic: Met, Ala, Val, Leu, He;

2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gin;

3) acidic: Asp, Glu;

4) basic: His, Lys, Arg;

5) residues that influence chain orientation: Gly, Pro; and

6) aromatic: Trp, Tyr, Phe.

[00100] Preferred amino acid substitutions include those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity and (4) confer or modify other physico-chemical or functional properties of such analogs. Analogs can include various muteins of a sequence other than the naturally occurring peptide sequence. For example, single or multiple amino acid substitutions (preferably conservative amino acid substitutions) may be made in the naturally occurring sequence (preferably in the portion of the polypeptide outside the domain (s) forming intermolecular contacts. A conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence) unless of a change in the bulk or conformation of the R group or side chain.

[00101] For example, non-conservative substitutions may involve the exchange of a member of one of these classes for a member from another class. Such substituted residues may be introduced into regions of the antibody that are homologous with antibodies to other orthologs, or into the non-homologous regions of the molecule.

[00102] Antibodies comprising certain embodiments of this invention that are polypeptide or peptide substitution variants may have up to about ten to twelve percent of the original amino acid sequence replaced. For antibody variants, the heavy chain may have 50, 49, 48, 47, 46, 45, 44, 43, 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid replaced, while the light chain may have 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid replaced.

[00103] In making such changes, according to certain embodiments, the hydropathic index of amino acids may be considered. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics. They are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9);

tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (- 3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).

[00104] The importance of the hydropathic amino acid index in conferring interactive biological function on a protein is understood in the art. It is known that certain amino acids may be substituted for other amino acids having a similar hydropathic index or score and still retain a similar biological activity. In making changes based upon the hydropathic index, in certain embodiments, the substitution of amino acids whose hydropathic indices are within ±2 is included. In certain embodiments, those which are within ±1 are included, and in certain embodiments, those within ±0.5 are included.

[00105] It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity; particularly where the biologically functional protein or peptide thereby created is intended for use in immunological embodiments, as in the present case. In certain embodiments, the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and antigenicity, i.e., with a biological property of the protein.

The following hydrophilicity values have been assigned to these amino acid residues:

arginine (+3.0); lysine (+3.0); aspartate (+3.0 ± 1); glutamate (+3.0 ± 1); serine (+0.3);

asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5 ± 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8);

isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5) and tryptophan (-3.4). In making changes based upon similar hydrophilicity values, in certain embodiments, the substitution of amino acids whose hydrophilicity values are within ±2 is included, in certain embodiments, those which are within ±1 are included, and in certain embodiments, those within ±0.5 are included. One may also identify epitopes from primary amino acid sequences on the basis of hydrophilicity. These regions are also referred to as "epitopic core regions."

[00106] A skilled artisan will be able to determine suitable variants of the polypeptide as set forth herein using well-known techniques. In certain embodiments, one skilled in the art may identify suitable areas of the molecule that may be changed without destroying activity by targeting regions not believed to be important for activity. In certain embodiments, one can identify residues and portions of the molecules that are conserved among similar

polypeptides. In certain embodiments, even areas that may be important for biological activity or for structure may be subject to conservative amino acid substitutions without destroying the biological activity or without adversely affecting the polypeptide structure.

[00107] In certain embodiments, antibody variants include glycosylation variants wherein the number and/or type of glycosylation site has been altered compared to the amino acid sequences of the parent polypeptide. In certain embodiments, protein variants comprise a greater or a lesser number of N-linked glycosylation sites than the native protein. An N- linked glycosylation site is characterized by the sequence: Asn-X-Ser or Asn-X-Thr, wherein the amino acid residue designated as X may be any amino acid residue except proline. The substitution of amino acid residues to create this sequence provides a potential new site for the addition of an N-linked carbohydrate chain. Alternatively, substitutions that eliminate this sequence will remove an existing N-linked carbohydrate chain. Also provided is a rearrangement of N-linked carbohydrate chains wherein one or more N-linked glycosylation sites (typically those that are naturally occurring) are eliminated and one or more new N- linked sites are created. Additional preferred antibody variants include cysteine variants wherein one or more cysteine residues are deleted from or substituted for another amino acid (e.g., serine) as compared to the parent amino acid sequence. Cysteine variants may be useful when antibodies must be refolded into a biologically active conformation such as after the isolation of insoluble inclusion bodies. Cysteine variants generally have fewer cysteine residues than the native protein, and typically have an even number to minimize interactions resulting from unpaired cysteines.

[00108] The functional equivalents of the present application also include modified antibodies, e.g., antibodies modified by the covalent attachment of any type of molecule to the antibody. For example, modified antibodies include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, deamidation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand, linkage to a toxin or cytotoxic moiety or other protein. The covalent attachment need not yield an antibody that is immune from generating an anti-idiotypic response. The modifications may be achieved by known techniques, including, but not limited to, specific chemical cleavage, acetylation, formylation, or metabolic synthesis. Additionally, the modified antibodies may contain one or more non-classical amino acids.

4. Pharmaceutical Compositions

[00109] Pharmaceutical compositions of the disclosed proteins of the invention are within the scope of the invention as disclosed herein. Pharmaceutical compositions comprising antibodies are described generally in the art. Such compositions comprise a therapeutically or prophylactically effective amount of a protein, particularly an antibody or a specific binding fragment thereof, variant, derivative or fusion thereof as described herein, in admixture with a pharmaceutically acceptable agent.

[00110] Therapeutic formulations of the protein particularly antibodies or a specific binding fragments thereof may be prepared for storage as lyophilized formulations or aqueous solutions by mixing a protein particularly antibodies or a specific binding fragments thereof having the desired degree of purity with optional "pharmaceutically acceptable" carriers, diluents, excipients or stabilizers typically employed in the art, i.e., buffering agents, stabilizing agents, preservatives, isotonifiers, non-ionic detergents, antioxidants and other miscellaneous additives. Such additives are generally nontoxic to the recipients at the dosages and concentrations employed, hence, the excipients, diluents, carriers and so on are pharmaceutically acceptable.

[0011 1] Formulation of the proteins particularly antibodies or a specific binding fragments thereof disclosed herein also may contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely impact each other. For example, it may be desirable to further provide an immunosuppressive agent. Such molecules suitably are present in combination in amounts that are effective for the purpose intended.

[00112] Non-ionic surfactants or detergents (also known as "wetting agents") may be added to help solubilize the therapeutic agent, as well as to protect the therapeutic protein against agitation-induced aggregation, which also permits the formulation to be exposed to shear surface stresses without causing denaturation of the protein. Suitable non-ionic surfactants include polysorbates (e.g., 20, 80), polyoxamers (e.g., 184, 188), Pluronic ® polyols and polyoxyethylene sorbitan monoethers (e.g., TWEEN-20 ® , TWEEN-80 ® ). Non-ionic surfactants may be present in a range of about 0.05 mg/ml to about 1.0 mg/ml, preferably about 0.07 mg/ml to about 0.2 mg/ml.

[00113] The pharmaceutical composition may contain formulation materials for modifying, maintaining or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition. Suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates, other organic acids); bulking agents (such as mannitol or glycine), chelating agents (such as ethylenediamine tetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or

hydroxypropyl-beta-cyclodextrin); fillers; monosaccharides; disaccharides and other carbohydrates (such as glucose, mannose, or dextrins); proteins (such as serum albumin, gelatin or immunoglobulins); coloring; flavoring and diluting agents; emulsifying agents; hydrophilic polymers (such as polyvinylpyrrolidone); low molecular weight polypeptides; salt-forming counter ions (such as sodium); preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine, sorbic acid or hydrogen peroxide); solvents (such as glycerin, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); suspending agents; surfactants or wetting agents (such as pluronics, PEG, sorbitan esters, polysorbates such as polysorbate 20, polysorbate 80, triton, tromethamine, lecithin, cholesterol, tyloxapal); stability enhancing agents (sucrose or sorbitol); tonicity enhancing agents (such as alkali metal halides (preferably sodium or potassium chloride, mannitol sorbitol); delivery vehicles; diluents; excipients and/or pharmaceutical adjuvants.

[00114] Optimal pharmaceutical composition can be appreciated by one skilled in the art depending upon, for example, the intended route of administration, delivery format, and desired dosage. Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the protein particularly antibodies or a specific binding fragments thereof

[00115] The primary vehicle or carrier in a pharmaceutical composition may be either aqueous or non-aqueous in nature. For example, a suitable vehicle or carrier may be water for injection, physiological saline solution possibly supplemented with other materials common in compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. Other exemplary pharmaceutical compositions comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, which may further include sorbitol or a suitable substitute therefore. In one embodiment of the invention, protein compositions may be prepared for storage by mixing the selected composition having the desired degree of purity with optional formulation agents in the form of a lyophilized cake or an aqueous solution. Further, the antibody product may be formulated as a lyophilizate using appropriate excipients such as sucrose.

[00116] Particular embodiments of the invention encompass liquid formulations having stability at temperatures found in a commercial refrigerator and freezer found in the office of a physician or laboratory, such as from about -20° C to about 5° C, said stability assessed, for example, by high performance size exclusion chromatography (HPSEC), for storage purposes, such as for about 60 days, for about 120 days, for about 180 days, for about a year, for about 2 years or more. The liquid formulations of the invention also exhibit stability, as assessed, for example, by HSPEC, at room temperatures, for at least a few hours, such as one hour, two hours or about three hours prior to use.

[00117] Pharmaceutical compositions as provided herein can be selected for parenteral delivery. The preparation of such pharmaceutically acceptable compositions is within the skill of the art.

[00118] The formulation components are present in concentrations that are acceptable to the site of administration. For example, buffers are used to maintain the composition at physiological pH or at slightly lower pH, typically within a pH range of from about 5 to about 8.

[00119] When parenteral administration is contemplated, the therapeutic compositions for use in this invention may be in the form of a pyrogen- free, parenterally acceptable aqueous solution comprising the desired antibody in a pharmaceutically acceptable vehicle. A particularly suitable vehicle for parenteral injection is sterile distilled water in which a binding agent is formulated as a sterile, isotonic solution, properly preserved. Yet another preparation can involve the formulation of the desired molecule with an agent, such as injectable microspheres, bio-erodible particles, polymeric compounds (polylactic acid, polyglycolic acid), beads, or liposomes that provides for the controlled or sustained release οφ the product which may then be delivered via a depot injection. Hyaluronic acid may also be used, and this may have the effect of promoting sustained duration in the circulation. Other suitable means for the introduction of the desired molecule include implantable drug delivery devices.

[00120] In a preferred embodiment, an aqueous solution of therapeutic protein including antibodies or specific -binding fragments thereof can be administered by subcutaneous injection. Each dose may range from about 1 mg to about 10 mg per kilogram of body weight. The dosage can be ascertained empirically for the T1D patient population, mode of administration and so on, practicing pharmaceutical methods known in the art.

[00121] The pharmaceutical composition to be used for in vivo administration typically must be sterile. This may be accomplished by filtration through sterile filtration membranes. Where the composition is lyophilized, sterilization using this method may be conducted either prior to or following lyophilization and reconstitution. The composition for parenteral administration may be stored in lyophilized form or in solution. In addition, parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.

[00122] For any compound, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models such as mice, rats, rabbits, dogs, or pigs. An animal model, specifically a NOD mouse model, may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.

[00123] The exact dosage can be determined in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide sufficient levels of the active compound or to maintain the desired effect. Factors that may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combination(s), reaction sensitivities, and response to therapy. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, every week, or biweekly depending on the half-life and clearance rate of the particular formulation.

[00124] The frequency of dosing will depend upon the pharmacokinetic parameters of the binding agent molecule in the formulation used. Typically, a composition is administered until a dosage is reached that achieves the desired effect. The composition may therefore be administered as a single dose or as multiple doses (at the same or different

concentrations/dosages) over time, or as a continuous infusion. Further refinement of the appropriate dosage is routinely made. Appropriate dosages may be ascertained through use of appropriate dose-response data.

[00125] To prolong the serum circulation of an antibody in vivo, various techniques can be used. For example, inert polymer molecules, such as high molecular weight polyethylene glycol (PEG), can be attached to an antibody with or without a multifunctional linker either through site-specific conjugation of the PEG to the N-terminus or to the C-terminus of the antibody or via ε amino groups present on lysine residues. Linear or branched polymer derivatization that results in minimal loss of biological activity can be used. The degree of conjugation can be closely monitored by SDS-PAGE and mass spectrometry to ensure proper conjugation of PEG molecules to the antibodies. Unreacted PEG can be separated from antibody-PEG conjugates by size-exclusion or by ion exchange chromatography. PEG- derivatized antibodies can be tested for binding activity as well as for in vivo efficacy using methods known to those of skilled in the art, for example, by immunoassays described herein.

[00126] An antibody having an increased half-life in vivo can also be generated by introducing one or more amino acid modifications (i.e., substitutions, insertions or deletions) into an IgG constant domain, or F C R binding fragment thereof (such as an F e or hinge F e domain fragment).

[00127] Further, an antibody can be conjugated to albumin to make an antibody more stable in vivo or have a longer half-life in vivo. The techniques are known in the art. The antibody also can be modified, for example, by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein and so on.

EXAMPLES

[00128] The invention now will be exemplified for the benefit of the artisan by the following non-limiting examples that depict some of the embodiments by and in which the invention can be practiced.

EXAMPLE 1: Producing recombinant GLUT2, generating and characterizating anti- GLUT2 mAbs

[00129] Although some antibodies (mainly polyclonal) to mouse GLUT2 are commercially available, there is no disclosure on whether those antibodies would possess antagonistic or neutralizing activities for GLUT2 activity (i.e., regarding whether they would interfere with glucose transporter activity), in which case they cannot be used as part of an antibody that binds both GLUT2 and CTLA-4 for therapy. Therefore, there is a need in the art for anti-GLUT2 mAbs that do not interfere with GLUT2 glucose transport activity.

Although GLUT2 is a transmembrane protein, an antibody that recognizes only its 67 amino acid ectoplasmic domain would be beneficial because the antibody would be able to bind to an extracellular epitope expressed specifically on the surface of pancreatic β-cells.

[00130] To ensure display of native epitopes and to retain native conformation, both full length GLUT2 (FL) and its ectodomain (Ecto) region were produced. The cDNA corresponding to the ecto-GLUT2 (-200 bp) was PCR amplified using primers having the sequence:

5 '-GCGCCATATGAATGCACCTCAAGAGGTAATAATA-3 ' (SEQ ID NO.: 02) and 5 '-GCGCGGATCCTTAAGACCAGAGCATAGTGACTATGTG-3 ' (SEQ ID NO.: 03). The ecto-GLUT2 was cloned into an E. coli expression vector (pET15b) in frame with the N- terminal 6XHis tag coding sequence. Recombinant Ecto-GLUT2 was purified (>95% purity) from E. coli lysate by Ni-NTA agarose, followed by reverse phase HPLC, dialyzed and concentrated (Fig. 1, upper panel). The peptide identity was confirmed by mass

spectrometry. The FL-GLUT2 cDNA (-1.5 kb) was PCR amplified using primers having the sequence:

5 ' -GCGCGGATCCATCAGAAGACAAGATCACCGG-3 ' (SEQ ID NO.: 04) and

5 '-GCGCGAATTCTCACACACTCTCTGAAGACGC-3 ' (SEQ ID NO.: 05) and cloned into a mammalian expression vector (pCDNA3.1-HisB) in frame with the coding sequence for an N-terminal 6xHis tag. HEK 293 cells were transfected with the plasmid and G418 resistant stable clones were selected. The membrane was solubilized in 2% Tween-20 in 50 mM phosphate buffer and the FL-GLUT2 was purified on a Ni-NTA agarose column and confirmed by western blot using anti-GLUT2 polyclonal antibodies (Fig. 1, lower panel).

[00131] In order to generate anti-GLUT2 antibodies, Balb/c mice were repeatedly immunized subcutaneously with 50 mg of purified Ecto-GLUT2 emulsified in complete Freund's adjuvant (CFA) and monitored for anti-Ecto-GLUT2 antibody levels. When animals showed high titers of antibodies (i.e., 1 :90,000), they were given a final dose of Ecto- GLUT2 without the adjuvant and were euthanized after 4 days. Splenocytes were then fused to SP2 myeloma cells and selected with hypoxantine/aminopterin/thymidine (HAT) medium. Hybridoma supernatants were tested for the presence of anti-GLUT2 antibodies by ELISA against Ecto-GLUT2. Positive hybridomas were cloned and further screened for anti-GLUT2 IgG by western blot against FL-GLUT2. Three clones (named D2, E7 and H5) were selected that were highly specific for Ecto-GLUT2 and FL-GLUT2 by western blot. These Abs were tested for their capacity to recognize native GLUT2 expressed on the surface of Min-6 cells (a mouse insulinoma cell line) by FACS using a secondary anti-mouse antibody. The 3B11 hybridoma supernatant (non-specific) was used as a negative control. All three anti-GLUT2 monoclonal antibodies recognized native GLUT2 expressed on Min-6 cells (Fig. 2, upper panel).

[00132] To test if mAb binding interfered with GLUT2 function, β-ΤΟ-6 (mouse insulinoma cell line) cells were seeded in 96 well plates and incubated overnight with media alone (control) or media plus one of the 3 different purified mAbs (i.e. D2, E7 and H5). Cells were then washed in a modified Krebs-Ringer HEPES buffer (pH= 7.4, 0.5% BSA) and insulin secretion was stimulated using Krebs 2.8 mM glucose solution (basal) or 16.7 mM glucose solution for 1 h. The amount of insulin in the incubation buffers and cell extracts were measured using an ultra sensitive mouse-insulin ELISA kit (Crystal Chem INC).

Insulin secretion was expressed as a percentage of the total insulin, which was the sum of insulin in basal and stimulated buffers, and cell extracts (Fig. 2, lower panel). Similar insulin secretion in the control and anti-GLUT2 Ab treated cells indicated that none of the mAbs disturbed GLUT2 function.

[00133] Anti-Glut2 mAb H5 was chosen for initial construction of the anti-CTLA4, anti-GLUT2 protein. This protein was tested for its capacity to bind to mouse pancreatic islets by immunofluorescence (Fig 3(a)). Anti-insulin staining showed co-localization of anti-GLUT2 and anti-insulin antibodies on pancreatic islets. The H5 mAb was compared with a commercial rabbit anti-GLUT2 Ab (abeam # ab54460) raised against a synthetic peptide derived from the cytoplasmic domain of rat GLUT2, for its binding to endogenous GLUT2 expressed in Min-6 cells in Western Blots (Fig. 3(b)). The commercial Ab reacted with membrane bound as well as the membrane bound GLUT2.

EXAMPLE 2: Constructing an anti-GLUT2, anti-CTLA-4 protein

[00134] Hamster anti-CTLA-4 Ig (hybridoma UC10 B-4-F-10-1 1; ATCC, Rockville, MD) was purified using protein-A agarose beads. Purified anti-GLUT2 (H5) and anti-CTLA- 4 IgGs were chemically coupled using Pierce Protein-Protein Cross-linking Kit to generate the protein, which was then followed by purification. The anti-CTLA4, anti-GLUT2 protein was used to stain mouse splenocytes and Min-6 insulinoma cells, and probed with anti-mouse and anti-hamster secondary Abs. Splenocytes and Min-6 cells were co-stained with anti-CD4 and anti-insulin Abs respectively. Approximately 9.7% of the CD4+ T cells were double positive for CD4 and CTLA-4 expression (Fig. 4, upper right panel) which was comparable to that detected using the anti-CTLA4, anti-GLUT2 protein (Fig. 4, lower middle and right panels). FACS analysis also revealed that nearly 90% of the Min-6 cells were double positive for insulin and GLUT2 expression as detected by a control as well as the anti- CTLA4, anti-GLUT2 protein (Fig. 5). These results showed that the anti-CTLA4, anti- GLUT2 protein can bind to native CTLA-4 and GLUT2 expressed on the cell mebrane. EXAMPLE 3: Examining the ability of an anti-Glut2, anti-CTLA-4 protein to target β- cells and suppress activated T-cells in vivo

[00135] While infiltrating T-cells recognize β-cell specific antigens through T-cell receptors and migrate to islets of Langerhans in the pancreas, anti-GLUT2, anti-CTLA-4 protein engages CTLA-4 on infiltrating T-cells. This process leads to suppression of effector T-cell function and generation of β-cell specific Tregs which inhibit the autoimmune effector T-cells from destroying β-cells. Thus, the efficacy of the anti-GLUT2, anti-CTLA-4 protein depends on how efficiently it can bind to GLUT2 expressing pancreatic β-cells. The anti- GLUT2, anti-CTLA-4 protein was tested to determine if it is capable of homing to β-cells by administering injections and performing immunohistochemistry on pancreatic tissue. To test this efficacy, 100 μg purified anti-CTLA4, anti-GLUT2 protein was injected intravenously into CB 17-SCID mice (devoid of endogenous Igs), and the pancreas harvested after 3 hours. Direct immunoflurescence staining using an anti-mouse secondary Ab showed that anti- CTLA4, anti-GLUT2 protein can bind to insulin secreting pancreatic islet cells in vivo (Fig. 6).

EXAMPLE 4: Analysis of anti-GLUT2, anti-CTLA-4 protein associated pathology.

[00153] To ensure that anti-CTLA4, anti-GLUT2 protein can be used safely without harmful side-effects, 6-week old NOD mice were treated three times with 100 ug of the anti- GLUT2, anti-CTLA-4 protein at 2-week intervals. Blood sugar was monitored weekly to assess deregulation in glucose metabolism as an indicator of perturbation in GLUT2 function. Through the end of a 10-week period, both groups of treated mice failed to show any perturbation in glucose levels as compared to untreated controls. The sera from these NOD mice were then tested for Alanine Aminotransferase (ALT), (Fig. 7(a)) and Blood Urea Nitrogen (BUN), (Fig. 7(c)) to detect to detect damage to hepatocytes and kidney

respectively. These results showed no difference in the levels of these markers (Fig. 7(a) and 7(c)) between control and the anti-CTLA4, anti-GLUT2 protein-treated groups.

Histopathological examination of the liver and kidney tissues from these mice also failed to reveal any damage (Fig. 7(b) and 7(d)). These results indicate that no harmful effects result from the treatment, and that an anti-CTLA4, anti-GLUT2 protein can be used safely without any apparent off target effects.

EXAMPLE 5: Determination of therapeutic potential of anti-GLUT2, anti-CTLA-4 protein in preventing Type 1 diabetes.

[00136] To determine the efficacy (whether treatment can stabilize the glucose levels or reverse ongoing diabetes) of the anti-GLUT2, anti-CTLA-4 protein treatment in mice at various stages of disease, groups of mice at starting at 6, 8 and 10 weeks of age were treated with anti-GLUT2, anti-CTLA-4 protein (100 μg/mouse/dose, referred in Figs. 7-12 as T- Glut2-CTLA-4) every two weeks until at least 23 weeks of age and monitored for disease progression as indicated by increasing blood glucose levels. The results were compared against one group of untreated mice (referred in the Figs 7-12 as control or untreated) and one group of mice treated with a non-specific mAb, anti-CTLA-4 protein (referred in the Figs 7- 12 as C-Ab or Non-sp Ab). The best protective effect was seen when the treatment was initiated at 10-weeks of age (Fig. 8), followed by initiation of treatment at 8-weeks age (Fig. 9); followed by initiation of treatment at 6 weeks of age (Fig. 10). These results show that anti-GLUT2, anti-CTLA-4 protein therapy can delay development of hyperglycemia in NOD mice. Table 1 shows summary of the results (therapeutic efficacy of biweekly anti-GLUT2, anti-CTLA-4 protein treatment of NOD mice started at 6 weeks, 8 weeks and 10 weeks of age).

Table 1

is calculated against the control group. [00137] These results indicated that the anti-CTLA-4 mAb is a hamster antibody and thus will be immunogenic in the recipient mice. Therefore, the more times the treatment is administered, the higher the resultant anti-hamster antibody titer (see Fig. 11). Evidently, the anti-hamster antibodies thus elicited in recipient NOD mice can neutralize the effect of subsequent anti-GLUT2, anti-CTLA-4 protein treatments. Since the 6, 8 and 10 week old recipients received 4, 3 and 2 anti-GLUT2, anti-CTLA-4 protein treatments respectively, prior to onset of diabetes, the 10 week study showed the highest efficacy. This problem can be readily overcome by using a mouse anti-CTLA-4 IgG.

EXAMPLE 6: Stability and persistence of anti-GLUT2, anti-CTLA-4 protein binding.

[00138] To test the stability of anti-GLUT2, anti-CTLA-4 protein in vitro, normal mouse sera are spiked with anti-GLUT2, anti-CTLA-4 protein at day 0. Spiked sera are incubated at 37°C for 7 days. Samples are then taken out every 24 hours and stored at -20°C. After 7 days, all samples are analyzed by ELISA against FL-GLUT2 to measure any loss of Ab integrity. The capacity of the Abs to simultaneously recognize CTLA-4 and GLUT2 is determined by FACS as described above for Figure 4.

[00139] To test the stability and persistence of anti-GLUT2, anti-CTLA-4 protein binding in vivo, 4 groups of 3 mice each are given a single intravenous dose of 100 μg anti- GLUT2, anti-CTLA-4 protein. One group of mice is euthanized at different time-points (1, 2, 4, and 8 days post- injection) and pancreatic tissue harvested. Fixed pancreatic tissue sections are further incubated with FITC labeled anti-hamster IgG and analyzed to determine the stability and persistence of anti-GLUT2, anti-CTLA-4 protein binding to pancreatic islets in vivo. EXAMPLE 7: Analysis of potential anti-GLUT2, anti-CTLA-4 protein associated pathology.

[00140] Although the in vitro studies showed that the anti-GLUT2 mAb is functionally neutral and mice immunized with GLUT2 to generate hybridomas showed no apparent signs of disease, a comprehensive pathological study of different tissues is performed to ensure lack of pathology. Groups of mice are treated with different doses of the anti-GLUT2, anti- CTLA-4 protein and observed for 2 weeks for weight loss or any sign of distress. Tail bleed is used to monitor blood glucose levels and serum insulin levels. Because GLUT2 acts as a vital glucose sensor in liver, kidney and β-cells, any perturbation of GLUT2 function could destabilize glucose levels and hence insulin release. Additionally, vital organs (brain, liver, kidney, heart and intestine) are harvested and analyzed for tissue damage.

EXAMPLE 8: Anti-GLUT2, anti-CTLA-4 protein modulation of T-cell function in vivo

[00141] The anti-GLUT2, anti-CTLA-4 protein is evaluated for its ability to modulate pathogenic T-cell function in vivo. It has been shown that in vitro activated NOD-BDC2.5 T- cells, as well as T-cells from wild-type NOD mice, can induce hyperglycemia rapidly in young NOD mice and NOD-SCID mice (Bending et ah, 2009, The Journal of Clinical Investigation 1 19:565-72; and Wagner et ah, 2002, Proc. Nat. Acad. Sic U.S.A. 99:3782-7). These adoptive transfer models are used to examine the ability of in vivo injected anti- GLUT2, anti-CTLA-4 protein to modulate pathogenic T-cell function in vivo. In the first set of experiments, spleen cells from NOD-BDC2.5 TCR transgenic mice are activated in vitro using BDC2.5 peptide (YVRPLWVRME, SEQ ID NO.: 06) for 4 days, CD4 + cells purified, labeled using Carboxyfluorescein succinimidyl ester (CFSE), and injected (lxlO 6 T- cells/mouse) intravenously into control antibody- and anti-GLUT2, anti-CTLA-4 protein- treated young (3-4 week old) NOD mice. Anti-GLUT2, anti-CTLA-4 protein is injected as described above 2 hours prior to T-cell injection. A control group that received T-cells but is treated with a control antibody is also included to determine whether anti-CTLA-4 portion of control antibody can influence T-cell properties in vivo. As reported in the inventor's earlier study (Karumuthil-Melethil et ah, 2010, Journal of Immunology 184:6695-708), adoptively transferred BDC2.5 TCR-Tg cells infiltrate the pancreas and destroy islets and induce hyperglycemia within 6-8 days. Therefore, one set of mice from each group is monitored for blood glucose levels every day for up to 10 days and every 3 days following that for 30 days to determine whether pathogenic properties of islet-infiltrating T-cells are suppressed in the anti-GLUT2, anti-CTLA-4 protein- injected mice. Upon anti-GLUT2, anti-CTLA-4 protein modulation of the pathogenic properties of islet infiltrating T-cells, the mice that received the protein then remain free of hyperglycemia for a longer period of time. A second set of mice is euthanized 96 hours post-cell transfer, immune cells isolated from pancreatic tissue, pancreatic lymph nodes and spleen are examined for Carboxyfluorescein succinimidyl ester (CFSE) + (/low) cells for proliferation profile, intracellular cytokines IL-10, IFN-gamma, IL- 17, TGF-βΙ levels, and expression of transcription factor Foxp3 by FACS. Upon anti- GLUT2, anti-CTLA-4 protein induction of T-cells with regulatory properties, BDC2.5 TCR- Tg T-cells recovered from this anti-GLUT2, anti-CTLA-4 protein treated mice then express Foxp3 and/or high levels of IL-10 or TGF-betal.

[00142] A range of doses from 20μg to 200μg of anti-GLUT2, anti-CTLA-4 protein are tested for the treatment. Single or multiple injections of the anti-GLUT2, anti-CTLA-4 protein is used to to suppress homogenous population of adoptively transferred pathogenic BDC2.5 T-cells. Multiple injections mitigate degradation of the anti-GLUT2, anti-CTLA-4 protein in vivo. Therefore, based on results from stability studies described above, experiments are repeated by daily injections with effective dose of the anti-GLUT2, anti- CTLA-4 protein for 3 days. In vitro activated T-cells of hyperglycemic NOD mice are injected to young NOD mice to advance hyperglycemia and study the effect of anti-GLUT2, anti-CTLA-4 protein treatment. This process mitigates β-cell destruction and hyperglycemia. Spleen cells from diabetic NOD mice are activated using immunodominant β-cell antigen peptides (such as: 1. Insulin B (9-23); 2. GAD65 (206-220); 3. GAD65 (524-543); 4. IA- 2beta (755-777) and 5; IGRP (123-145) as described in the inventor's earlier study

(Karumuthil-Melethil et ah, 2010, Journal of Immunology 184:6695-708) before injecting into anti-GLUT2, anti-CTLA-4 protein recipient and control mice.

EXAMPLE 9: Optimization of anti-GLUT2, anti-CTLA-4 protein treatment.

Whether a single injection of the anti-GLUT2, anti-CTLA-4 protein is sufficient or intermittent injections are needed is determined using the protocol described below. Five mice are used per group to determine the minimum number of injections of the anti-GLUT2, anti-CTLA-4 protein needed to achieve effective protection. In one set of experiments, eight week old pre-diabetic NOD mice are injected intravenously with the anti-GLUT2-anti- CTLA-4 protein or control antibody either only once on day 1 or every other day (i.e. on days 1, 3, 5 and 7). In the second set of experiments, injections are repeated at two-week intervals. The effect of the anti-GLUT2, anti-CTLA-4 protein injection is assessed by weekly monitoring of blood glucose levels for up to 25 weeks

EXAMPLE 10: Determining the efficacy of anti-GLUT2, anti-CTLA-4 protein treatment in mice at various stages of disease

[00143] The experiments above indicated that anti-GLUT2, anti-CTLA-4 protein treatment can prevent the onset of Type 1 diabetes in NOD mice. However, whether anti- GLUT2, anti-CTLA-4 protein treatment can also stabilize glucose levels or reverse ongoing diabetes can be determined. Groups of mice are treated at 8, 10, 12 weeks of age using an optimized anti-GLUT2, anti-CTLA-4 protein dose and disease progression monitored. In addition, pre-hyperglycemic mice (glucose levels between 150-250 mg/dl) and

hyperglycemic mice (glucose levels >250 mg/dl) selected from mice of ages between 12 and 25 weeks are also treated with the optimized anti-GLUT2, anti-CTLA-4 protein dose and regimen. Prior to, and every 7 days after, the initiation of the treatment, the blood glucose levels are monitored in these mice for up to 25 weeks post-treatment. To ensure normal immune cell function and to avoid prolonged hyperglycemia-induced complications, diabetic test and control groups of mice are injected subcutaneous ly with insulin every day

(2U/mouse/day) for the first 30 days. This provides a sufficient period of time for the therapy to be effective under a euglycemic state and for the potential restoration of euglycemia under the condition of suppressed autoimmunity. It is anticipated that mice treated with the anti- GLUT2, anti-CTLA-4 protein would show significant delay in the onset of hyperglycemia when treated at pre-diabetic stage. Diabetic mice treated with this anti-GLUT2, anti-CTLA-4 protein may remain euglycemic even after withdrawing insulin treatment on day 30. Mice are continually treated bi-weekly with the anti-GLUT2, anti-CTLA-4 protein and determine blood glucose levels to determine the optimum amount of the protein that is used to keep the mice disease free. The optimum anti-GLUT2, anti-CTLA-4 protein treatment cannot protect mice beyond a certain point of disease progression if the treatment is stopped.

EXAMPLE 11: Type 1 diabetes suppression by anti-GLUT2, anti-CTLA-4 protein.

[00144] Disease suppression can result from either direct suppression of effector T- cell (Teff) response due to CTLA-4 mediated down modulation or induction of Tregs, which in turn can suppress Teff cells. Based on the inventor's previous studies on CTLA-4 engagement studies (Karumuthil-Melethil et ah, 2010, Id.; Li et ah, 2001 , Id.; Vasu et ah, 2004, Id.; Perez et ah, 2008, Id.), disease suppression is believed to involve both mechanisms of action. To investigate these mechanisms of action, T-cells from anti-GLUT2, anti-CTLA- 4 protein-treated and control mice are examined for their phenotypic and functional properties. Eight week old mice are treated with an optimal dose of therapeutic anti-GLUT2, anti-CTLA-4 protein and control antibody twice at bi-weekly interval. Two weeks after the last injection mice are sacrificed; spleen and pancreatic draining lymph node cells from these mice are used for multiple immunological analyses. Pancreata are also isolated for histopathological studies. EXAMPLE 12: Analysis of Treg frequency and function

[00145] Foxp3 is a marker for Tregs and maintenance of its sustained expression determines the dominant suppressor function of Tregs (Williams & Rudensky, 2007, Nature Immunology 8:277-84; Wan & Flavell, 2007, Nature 445:766-70). Therefore, to determine the levels of induction of Tregs, cells from the spleen and pancreatic lymph nodes of mice that were repeatedly treated with anti-GLUT2, anti-CTLA-4 protein and control antibody at bi-weekly interval were analyzed by staining for intra-cellular Foxp3 expression. FACS analysis revealed increased Foxp3+ Treg percentages in anti-GLUT2, anti-CTLA-4 protein (T-Glut2-CTLA-4) treated mice compared to either untreated or control antibody (C-Ab) treated mice (Fig. 12).

[00146] IL-10 and TGF-beta have been implicated in the induction of Treg mediated tolerance. Therefore, to determine the levels of IL-10 production by Tregs, cells from the spleen and pancreatic lymph nodes of mice that were repeatedly treated with anti-GLUT2, anti-CTLA-4 protein and control antibody at bi-weekly interval were analyzed by staining for intra-cellular IL-10 expression. FACS analysis revealed increased CD4+IL-10+ percentages in anti-GLUT2, anti-CTLA-4 protein treated mice than either untreated or control antibody treated mice.

[00147] Since a majority of T-cells with immunoregulatory properties express Foxp3,

IL-10 and/or TGF-beta, expression levels of TGF-betal are also examined in these cells by FACS. Moreover, recently induced Tregs have been shown to express Glycoprotein A Repetitions Predominant (GARP) (Wang et ah, 2008, PLoS One 3 :e2705; Bhattacharya et ah, 201 1, Journal of Leukocyte Biology 89:235-49). Therefore, cells are stained for GARP as well as Foxp3 expression. These analyses indicate the proportion of induced versus natural Tregs and if the Tregs are increased in anti-GLUT2, anti-CTLA-4 protein treated mice relative to controls.

[00148] To examine the effects of anti-GLUT2, anti-CTLA-4 protein on T cell activation in vitro, splenic CD4+ T-cells were isolated from hyperglycemic NOD mice, labeled with CFSE and co-cultured with splenic APCs, immunodominant β-cell Ag peptides and total pancreatic cells in the absence or presence of different concentrations of anti- GLUT2, anti-CTLA-4 protein or control protein (C-Ab). Results showed suppression of CD4+ T-cell proliferation in the presence of anti-GLUT2, anti-CTLA-4 protein (T-Glut2- CTLA-4), but not the control antibody (C-Ab) (Fig. 13, upper panel). Additionally, after 96 hours of co-culture, an increase in the frequency of Foxp3+ Tregs in the anti-GLUT2, anti- CTLA-4 protein (T-Glut2-CTLA-4) treated cultures relative to the control (C-Ab) treated culture was noted (Fig. 13, lower panel).

[00149] If an increase in Treg frequency is observed, then the effector T-cell suppressive properties of CD4 + CD25 + isolated from spleen and draining lymph node cells from anti-GLUT2, anti-CTLA-4 protein treated and control mice are compared. Effector CD4 + CD25 " T-cells from control and treated mice are labeled with CFSE and cultured either alone or in the presence of Tregs from control and anti-GLUT2, anti-CTLA-4 protein treated mice, and in the presence or absence of a mixture of immunodominant β-cell antigen peptides indicated above and splenic dendritic cells (DCs) as APCs (Karumuthil-Melethil et al., 2010, Id.). Alternatively, cells are stimulated using plate bound anti-CD3 Ab and soluble CD28 Ab. Cell proliferation is examined by CFSE dilution assay. The Tregs are cultured at different ratios of 1 : 1, 1 :2, 1 :4, 1 :8, and 1 : 16 to evaluate their suppressive abilities.

T-cells with regulatory properties induced by anti-GLUT2, anti-CTLA-4 protein can be Foxp3 + but produce IL-10 and/or TGF-betal. Therefore, to compare the overall proliferative response of T-cells from treated and control mice, spleen and draining lymph node cells are labeled with CFSE, cultured in the presence of β-cell antigen peptides for 4 days, and proliferative response in CD4 + and CD8 + T-cells are examined by FACS.

EXAMPLE 13: Cytokine analyses

[00150] T-cells from spleen and pancreatic lymph nodes of anti-GLUT2, anti-CTLA-4 protein treated and untreated mice are isolated and cultured in the presence of β-cell antigen peptides or CD3/CD28 Abs as described above. Cultures are maintained for 3 days and the supernatants examined for cytokine levels. Pro-inflammatory and immunoregulatory cytokine levels are also measured in the serum samples collected from treated and untreated mice. Specifically levels of IL-2, IL-4, IL-6, IL-10, IL-12, IL-Ι β, IFN-gamma, IL-17, TGF- beta and TNF-alpha are examined to differentiate between Thl, Th2 and regulatory T-cell responses. This serves as an indicator of the qualitative difference in the immune response between anti-GLUT2, anti-CTLA-4 protein treated and control mice.

EXAMPLE 14: Histopathology of islets

[00151] Histopathological examination of Hematoxylin and Eosin (H&E) stained sections of pancreatic tissue from different experimental groups of mice (from the 10-week treatment groups selected at random) revealed extensive lymphocytic infiltration of pancreatic tissue in untreated and control Ab treated mice. In contrast, lymphocytic infiltration was essentially absent in the pancreata of mice that were treated with therapeutic anti-GLUT2, anti-CTLA-4 protein (T-Glut2-CTLA4). These results show that T-Glut2- CTLA4 treatment resulted not only in the expansion of regulatory T-cells but that those cells were capable of suppressing β-cell specific immune response in vitro as well as in vivo.

[00152] It should be understood that the foregoing disclosure emphasizes certain specific embodiments of the invention and that all modifications or alternatives equivalent thereto are within the spirit and scope of the invention as set forth in the appended claims.