Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PEPTIDES AND METHODS FOR TREATING DISEASES
Document Type and Number:
WIPO Patent Application WO/2021/011437
Kind Code:
A1
Abstract:
Bioactive peptides and methods for their use for preventing, treating, and/or reducing the incidence and/or symptoms of an autoimmune disease or disorder that may result from chronic inflammation, damage to β-cells of the pancreas, and other conditions that are implicated by the CD40-CD154 dyad. In particular, small peptides that are capable of interacting with CD40, thereby interfering with the ability of CD40 to interact with CD 154, which impacts inflammation, autoimmunity, and disease progression. The use of such peptides in reducing diabetes mellitus, and in particular, the autoimmune inflammatory response that may be a driving factor thereof. Methods and materials for preventing and modulating diabetes mellitus and autoimmune diseases especially in dogs, cats, and horses.

Inventors:
WAGNER JR (US)
YUSSMAN MARTIN GLENN (US)
HENRY CHARLES W (US)
Application Number:
PCT/US2020/041744
Publication Date:
January 21, 2021
Filing Date:
July 13, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
OP T LLC (US)
International Classes:
A61K38/00; C07K7/00; C07K7/06; C07K7/08; C07K14/00; C07K14/435
Domestic Patent References:
WO2019136307A12019-07-11
WO2019032945A12019-02-14
WO2019136307A12019-07-11
WO2008036675A22008-03-27
Foreign References:
US20110178000A12011-07-21
US20050202531A12005-09-15
US4642295A1987-02-10
US20190194290A12019-06-27
US20160200823A12016-07-14
US20170319671A12017-11-09
US20120282291A12012-11-08
Other References:
LEIGHTON ET AL.: "A Practical Review of C-Peptide Testing in Diabetes", DIABETES THE, vol. 8, 8 May 2017 (2017-05-08), pages 475 - 487, XP055717509, DOI: 10.1007/s13300-017-0265-4
BANCHEREAU, J. ET AL., ANNU. REV. OF IMMUNOL., vol. 12, 1994, pages 881
SAMBROOK, J.FRITSCH, E.F.MANIATIS, T: "Molecular Cloning: A Laboratory Manua", December 1989, COLD SPRING HARBOR LABORATORY PRESS
See also references of EP 3996731A4
Attorney, Agent or Firm:
SCULL, Peter B. (US)
Download PDF:
Claims:
What is claimed:

1. A peptide less than 20 amino acids in length, for the treatment of diabetes, said peptide comprising an amino acid sequence selected from the group of: SEQ ID NOs: 7-15, 21-26, 41-48, and 50-52.

2. A peptide of claim 1 that is administered via intramuscular (IM) delivery, intravenous (IV) delivery, subcutaneous (SC) delivery, oral delivery, gavage delivery, emollient/skin delivery, transdermal patch, and/or intranasally.

3. A peptide of claim 1 that inhibits, affects, interferes, or interacts with CD40.

4. The peptide of claim 1 that is produced synthetically.

5. The peptide of claim 1 that is conjugated or fused to a second peptide or polypeptide.

6. A pharmaceutical composition comprising the peptide of claim 1 and a

pharmaceutically acceptable carrier.

7. A peptide that affects the binding of CD40 to CD 154, wherein said peptide is less than 20 amino acids in length and comprises at least one sequence selected from SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15.

8. A peptide that interacts with CD40, wherein said peptide is less than 30 amino acids in length and comprises at least one sequence selected from SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO: 50, SEQ ID NO: 51, and SEQ ID NO: 52.

9. The peptide of claim 8 that disrupts or inhibits interaction between CD40 and CD 154.

10. The peptide of claim 8 that includes a C-terminal amidated amino acid.

11. The peptide of claim 8 that includes an N-terminal acetylated amino acid.

12. The peptide of claim 8 that is produced synthetically.

13. The peptide of claim 8 that is conjugated or fused to a second peptide or polypeptide.

14. A pharmaceutical composition comprising the peptide of any of claims 8-13 and a pharmaceutically acceptable carrier.

15. A method for preventing, modulating, and/or reducing diabetes mellitus in a subject, the method comprising administering a peptide selected from the group of: SEQ ID NO:

7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15.

16. A method of claim 15 wherein the subject is a dog, cat, or horse.

17. A method of claim 15 wherein the subject is a mouse or human.

18. A method of claim 15, wherein the peptide binds to CD40.

19. A method of claim 15, wherein said peptide binds to a CD40 protein with a Kd of greater than 10 6.

20. A method of claim 15, wherein said peptide affects the interaction of CD40 and CD154.

21. A method of claim 15, wherein the peptide affects the interaction of CD40 with CD 154 in such a manner as to alter the cytokine expression profile of a cell population treated with said peptide.

22. A method of claim 15, wherein the subject does not produce autoantibodies responsive to the peptide and/or CD 154.

23. The method of any of the prior claims, wherein said peptide binds to multiple bone marrow derived cell types which express CD40 of approximately 45kDa, including splenic CD4hi cells, CD8 cells, and other antigen presenting cells.

24. The method of any of the prior claims, wherein said peptide is obtained from natural sources via degradation or it is synthesized.

25. A method to identify a subject at risk for developing autoimmune disease and/or diabetes mellitus, the method comprising obtaining a sample containing T-cells from a patient to be tested, contacting the sample with a peptide that binds the CD40 protein, detecting the CD-40 bound peptide, and determining the level of Th40 cells from the amount of CD40 bound, wherein a level of Th40 cells greater than 25% of the total T-cell population indicates the patient is at risk for developing cardiovascular disease and/or developing atherosclerosis.

26. A method of claim 25, wherein the peptide is selected from the group of: SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID N0:41, SEQ ID NO:42, SEQ ID NO:43, SEQ ID NO:44, SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO: 50, SEQ ID N0:51, and SEQ ID NO:52.

27. A method of administering a CD40 peptide to prevent, modulate, and/or reduce diabetes mellitus, comprising:

selecting a peptide from the group of SEQ ID NO: 7-15, 21-26, 41-48, and 50-52; selecting a delivery method from the group comprising intramuscular (IM) delivery, intravenous (IV) delivery, subcutaneous (SC) delivery, oral delivery, gavage delivery, emollient/skin delivery, or transdermal patch;

using the selected delivery method to administer the selected peptide to a subject.

28. A method of administering a CD40 peptide to prevent, modulate, and/or reduce diabetes mellitus, comprising:

selecting a peptide from the group of SEQ ID NO: 7-15, 21-26, 41-48, and 50-52; selecting a an administration from the group comprising: an implantable device, a hydrophilic polymer formulation, a permeable polymeric membrane, injectable gel implants, solvent extraction system, phase inversion system, thermosensitive gels, pH dependent in situ gels, microparticles, microspheres, nanoparticles, nanospheres, bio degradable implants, or photoactivated depot;

using the selected administration to administer the selected peptide to a subject.

29. The method of any of the prior claims, wherein said peptide binds to multiple bone marrow derived cell types which express CD40 of approximately 45kDa, including splenic CD4hi cells, CD8 cells, and other antigen presenting cells.

30. A method to modulate and/or reduce IFN-g in a cell or a subject comprising administering a peptide selected from SEQ ID NOs 7-15, 21-26, 41-48, and 50-52, in an amount sufficient to reduce or inhibit IFN-g signaling, wherein, the IFN-g signaling.

31. A method of claim 30, wherein the IFN-g signaling is associated with diabetes mellitus or an autoimmune disease, disorder, or syndrome.

32. A method to modulate and/or reduce interleukin-2 signaling in a cell or a subject comprising administering a peptide selected from SEQ ID NOs 7-15, 21-26, 41-48 and 50- 52, in an amount sufficient to reduce or inhibit interleukin-2 signaling.

33. A method of claim 32, wherein the interleukin-2 signaling is associated with diabetes mellitus or an autoimmune disease, disorder, or syndrome.

34. A method to modulate and/or reduce interleukin 17 (IL-17) in a cell or a subject comprising administering a peptide selected from SEQ ID NOs 7-15, 21-26, 41-48 and 50- 52, in an amount sufficient to reduce or inhibit interleukin 17 (IL-17) signaling.

35. A method of claim 34, wherein the interleukin- 17 signaling is associated with diabetes mellitus or an autoimmune disease, disorder, or syndrome.

36. A method to modulate and/or reduce blood glucose of a subject comprising:

selecting a peptide from the group of SEQ ID NO: 7-15, 21-26, 41-48 and 50-52, administering to a subject in need thereof, a therapeutically effective amount of the peptide selected from the group of SEQ ID NO: 7-15, 21-26, 41-48 and 50-52 that reduces, modulates, and/or affects the blood glucose levels of the subject.

37. A method to modulate and/or reduce fructosamine serum values in a subject comprising:

selecting a peptide from the group of SEQ ID NO: 7-15, 21-26, 41-48 and 50-52, administering to a subject in need thereof, a therapeutically effective amount of the peptide selected from the group of SEQ ID NO: 7-15, 21-26, 41-48 and 50-52 that reduces, modulates, and/or affects the fructosamine serum values of the subject.

38. A method to modify, change, or affect the c-peptide levels of a subject comprising: selecting a peptide from the group of SEQ ID NO: 7-15, 21-26, 41-48 and 50-52, administering to a subject in need thereof, a therapeutically effective amount of the peptide selected from the group of SEQ ID NO: 7-15, 21-26, 41-48 and 50-52 that modifies, changes, or affects the c-peptide levels of the subject.

39. A peptide for the treatment of diabetes, said peptide comprising an amino acid sequence selected from the group of: SEQ ID NOs: 7-15, 21-26, 41-48, and 50-52, wherein a modification is made at the N-terminus.

40. A peptide of claim 37 the modification selected from the group of: 5-FAM, 5-FAM- ahx, Abz, Acetylation, Acryl, Alloc, Benzoyl, Biotin, Biotin-Ahx, BOC, Br-Ac-, BSA, CBZ, Dansyl, Dansyl-Ahx, Decanoic acid, DPTA, Fatty Acid, FITC, FITC-Ahx, Fmoc, Formylation, Hexanoic acid, HYNIC, KLH (-NH2 of N-terminal), Laurie acid, Lipoic acid, Maleimide, MCA, Myristoyl, Octanoic acid, OVA (-NH2 of N-terminal), Palmitoyl, PEN, Stearic acid, Succinylation, and/or TMR.

41. The peptide for the treatment of diabetes, said peptide comprising an amino acid sequence selected from the group of: SEQ ID NOs: 7-15, 21-26, 41-48, and 50-52, wherein a modification is made at the C-terminus.

42. The peptide of claim 39 the modification selected from the gourp of: AFC, AMC, Amidation, BSA (-COOH of C-terminal), Bzl, Cysteamide, Ester (Oet), Ester (OMe), Ester (OtBu), Ester (OTBzl), KLH (-COOH of C-terminal), MAPS asymmetric 2, 4, or 8 branches, Me, NHEt, NHisopen, NHMe, OSU, OVA (-COOH of C terminal), p- Nitroanilide, and/or tBu.

43. A peptide according to any of the previous claims, the peptide binding to live cells for less than 1 day.

44. A peptide according to any of the previous claims, the peptide binding to live cells for more than 1 day.

45. A peptide according to any of the previous claims, the peptide binding to live cells for more than 2 days.

46. A peptide according to any of the previous claims, wherein administration of the peptide reduces IFNy.

47. A peptide according to any of the previous claims, wherein administration of the peptide reduces IL-2.

48. A peptide according to any of the previous claims, wherein administration of the peptide reduces IL-17a.

49. A pharmaceutical composition for the treatment of diseases selected from the group of type 1 diabetes, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosa, chronic obstructive pulmonary disease, atherosclerosis, and/or acute respiratory distress syndrome, comprising at least one bioactive peptide selected from SEQ ID NOs: 7-15, 21- 25, 41-48, and 50-52, a sterile solubilizing agent, and a buffering agent.

50. A pharmaceutical composition according to claim 47 further comprising a sugar.

51. A pharmaceutical composition according to claim 47, the solubilizing agent being water.

52. A pharmaceutical composition for the treatment of hyperglycemia, comprising at least one bioactive peptide selected from SEQ ID NOs 7-15, 21-26, 41-48, and 50-52, wherein the peptide is dissolved in a sterile solution in phosphate buffered saline (PBS).

53. A composition, method or system as described herein.

Description:
Title: PEPTIDES AND METHODS FOR TREATING DISEASES

Sequence Listing

The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy created on July 11, 2020 is named SOP0_00901WO_Sequence_Listing_ST25.txt and is 33 kilobytes in size.

The present developments relate to peptides and methods and/or uses thereof for prevention, modulation, and treatment of disease in subjects. Further, the present disclosure relates to peptides that affect, interact, and/or inhibit the interaction of the CD40 complex and CD154, and the use of such compounds in modulating T-cell activity and treating disease.

Background

Inflammation may occur when inflammatory cells, such as neutrophils, eosinophils, basophils, mast cells, macrophages, platelets, and endothelial cells, respond to inflammatory events or harmful stimuli, such as, invading microorganisms, damages cells, or other irritants. The body’s inflammatory response is beneficial because for example, in the case of invading microorganisms, the inflammatory response is an important step in localizing the infecting agent for removal by the immune system. However, in autoimmunity there is no infection, yet severe inflammation is present or persistent. The inflammation in this case, referred to as aseptic chronic inflammation (ACI), is detrimental since it destroys normal tissues. The results of this aseptic inflammation are life-altering and in some cases life-threatening. Moreover, as with acute inflammation, this process is mediated by immune cells, including T-cells.

A major concern for modem medicine is how to control ACI such as that which occurs during autoimmune diseases, as well as how to control acute inflammation resulting from trauma. Inflammation, both chronic and acute, leads to tissue degeneration and eventual loss of function of major organs. ACI is not limited to a single disease, but is instrumental in numerous autoimmune diseases, including, but not limited to: type 1 diabetes (T1D), multiple sclerosis (MS), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Crohn’s disease, inflammatory bowel disease (IBS), chronic obstructive pulmonary disease (COPD) including types of autoimmune asthma, atherosclerosis, vasculitis, hypertension, thyroiditis including Hashimoto’s and Graves diseases, primary biliary cirrhosis, Paget’s disease, Addison’s disease, acute respiratory distress syndrome (ARDS), acute lung injury, and aseptic chronic inflammation (ACI) associated with organ transplantation.

Autoimmune disorders are classified into two types: organ-specific (directed mainly at one organ) and non-organ-specific (widely spread throughout the body). Examples of organ-specific autoimmune disorders are insulin-dependent Type 1 diabetes (T1D) which affects the pancreas; Hashimoto's thyroiditis and Graves' disease, which affect the thyroid gland; pernicious anemia, which affects the blood; Addison's disease, which affects the adrenal glands; chronic active hepatitis, which affects the liver; myasthenia gravis which affects the muscle; and multiple sclerosis (MS), which affects tissue of the nervous system. An example of a non-organ-specific autoimmune disorders is rheumatoid arthritis (RA). Autoimmune diseases are often chronic, debilitating, and life-threatening. The National Institutes of Health (NIH) estimates that up to 23.5 million Americans suffer from autoimmune disease and that the prevalence is rising. It has been estimated that autoimmune diseases are among the ten leading causes of death among women in all age groups up to 65 years.

Acute inflammation, as observed during trauma or sepsis, is also immune cell mediated. While a comprehensive, complete, and exhaustive list of the molecular mediators in this process have not yet been identified, a prominent role for T-cells, lymphocytes, neutrophils, macrophages, monocytes, neutrophils, eosinophils, basophils, mast cells, and other inflammatory cells is strongly implicated. Therefore, a process to modulate these cell types may control the inflammatory response.

It is known that canines, felines, and equine suffer from a variety of autoimmune diseases. Some of these diseases may have molecular drivers that are similar to those of humans, while others may have causes that are quite different, and many are not yet known. In any event, the market for disease therapeutics for companion animals is a growing field and researchers and pet owners, and veterinarians alike are in search of therapies for diseases that afflict these valued and treasured animals.

Diabetes mellitus is one of the most common endocrine diseases that a canine may develop (Catchpole, B., et ah, Diabetologia, 2005, 48: 1948-56). Further, the incidence of canine diabetes mellitus is increasing at substantial rates, with one study showing the incidence increasing from 19 cases per 10,000 dogs in 1970 to 64 cases per 10,000 dogs in 1999 (Gruptill, L., et ah, Vet. T, 2003, 165:240-47). The standard of care usually requires that a dog suffering from the disease initially start an insulin therapy, irrespective of the underlying cause and/or the classification of the diabetes (O’Kell, A, et al., Diabetes, 2017, 66(6): 1443-52). Currently, cases of canine diabetes are classified into two different types: insulin deficiency diabetes and insulin resistance diabetes (Catchpole, B., et al., Diabetologia , 2005, 48: 1948-56). It is currently postulated that canines are not susceptible to developing diabetes that is equivalent to human type 2 diabetes; however, it is noted that obesity is associated with insulin resistance diabetes in some cases (Id).

Most diabetic dogs suffer from insulin deficiency diabetes, where the reason for the disease is loss or destruction of pancreatic b-cells results in the underproduction or complete lack of production of insulin (Id.). The role of autoimmunity is currently less clear in canine diabetes development than that of human T1D and the non-obese diabetic (NOD) mouse development; however, the inflammatory process and autoimmunity is still suspected in at least some cases of the disease (Id ; Matthews, CE., Pediatr. Diabetes, 2005, 6: 165-77; Amer. Diabetes Association Diagnosis and Classification of Diabetes Mellitus, Diabetes Care , 2014, 37, Suppl. l :S81-S90). Canine diabetes is often diagnosed when hyperglycemia (usually > ~ 250 mg/dL) and glucosuria are identified; however, no single diagnostic indicators, benchmarks, or criteria currently exist (O’Kell, A, et al., Diabetes, 2017, 66(6): 1443-52).

Diabetes mellitus is also a common endocrine disease in cats (Gottlieb, S., et al., Vet. Med. (Auckl), 9:33-42, 2018). The current classification of feline diabetes is based on human diabetes including the mechanisms and causes involved in pancreatic b-cell failure thereof. Thus, it is currently postulated that cats suffer from both type 1 and type 2 diabetes; however, type 1 diabetes is considered less common in felines, than in humans or dogs (Id.).

Horses also suffer from diabetes mellitus and again the model for this disease state is based off what is understood about human diabetes (Johnons, P.J., et al., J Diabetes Sci. Tecnol. 6(3):534-40, 2012). Horses are susceptible to develop equine metabolic syndrome (EMS) which may in turn cause the development of debilitating conditions such as laminitis, which affects the equine hoof (Id.). Development of laminitis sometimes necessitates euthanasia. (Id.) Therefore, a therapy which may improve conditions related to disease and inflammation may be useful to prevent complications in equine subjects that are afflicted with EMS.

The present subject matter is also related to the discovery that a unique subset of T-cells, which express CD40 protein, and thus are referred to as Th40 cells, that may be instrumental in autoimmune inflammation. Moreover, involvement of Th40 cells in the autoimmune process may be dependent on the interaction between CD40 protein expressed on the surface of the T-cell, and CD 154 protein. Interaction of CD40 and CD 154 results in activation signals being delivered between the cells, and subsequent activation of the Th40 cell. Such activation results in propagation of the Th40 cell and an increase in inflammation (e.g., an increase in the number of immune cells and immunoregulatory molecules, present in the system). Accordingly, inhibition of the CD40/CD154 interaction may modulate Th40 cell activity, and thereby affect inflammation.

Thus, there exists a need in the art for safer and more effective methods for treatment and prevention of autoimmune diseases in canines, felines, and equines, including for diabetes mellitus. The present disclosure provides novel bioactive peptides, variants, and derivatives thereof. Moreover, the present developments may address this need by describing peptides and methods for treatment of autoimmune diseases by administration of a therapeutically effective amount of a CD40 peptide. Further, the present developments may provide the added benefit of preventing auto-antibody generation, and thus may allow the resumption of normal immune function.

This statement of background is for information purposes only and is not intended to be a complete or exhaustive explication of all potentially relevant background.

Summary

The present developments relate to bioactive peptides and their use in methods for treating subjects before, during, and/or after the development of diabetes mellitus and/or an autoimmune condition. The present developments demonstrate that the peptides hereof may impact inflammatory conditions that are present or concurrent with a number of diseases. The peptides hereof may be used to provide therapeutic results to help control, manage, or impact conditions and diseases that are caused by inflammation, the inflammatory cascade, or autoimmune- autoinflammatory pathways. Thus, the present developments relate to the use of peptides hereof to control one or more of blood glucose, fructosaime serum levels/values, c-peptide levels, and other measured markers of disease, disorders, or inflammation and described and disclosed herein.

These developments may be related to the understanding that the CD40-CD154 inflammatory dyad may play and important role in the development and progression of diabetes mellitus and autoimmune diseases. The present developments also may relate to reducing Th40 cell levels and hyperglycemia. The present developments may also relate to the therapeutic effects of reducing frutcosamine serum values and increasing c-peptide levels.

Thus, this disclosure provides bioactive peptides and methods for their use for preventing, treating, and/or reducing the incidence and/or symptoms of an autoimmune disease or disorder that may result from chronic inflammation, damage to b-cells of the pancreas, and other conditions that are implicated by the CD40-CD154 dyad. The peptides disclosed herein provide variants, homologs, orthologs, and/or other derivatives that may be useful in the treatment of disease. The methods disclosed herein include administering to a subject in need of treatment an effective amount of a peptide that affects, modifies, inhibits, modulates, and/or interacts with CD40. This interaction may be in such a manner as to interfere with CD40 proteins and CD 154 proteins binding and interactions. In these methods, the autoimmune disease may include: type 1 diabetes (T1D), multiple sclerosis (MS), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Crohn’s disease, inflammatory bowel disease (IBS), chronic obstructive pulmonary disease (COPD) including types of autoimmune asthma, atherosclerosis, vasculitis, hypertension, thyroiditis including Hashimoto’s and Graves diseases, primary biliary cirrhosis, Paget’s disease, Addison’s disease, acute respiratory distress syndrome (ARDS), acute lung injury, and aseptic chronic inflammation (ACI) associated with organ transplantation.

The peptides hereof also may be used in methods of treating or preventing hyperglycemia in a subject by administering to the subject a composition comprising a peptide of this disclosure, and functional fragments and/or homologs and analogs thereof, in an amount and under conditions effective to decrease hyperglycemia.

The peptides useful in these methods may be a peptide that binds to, interacts with, and/or affects CD40. Further, the peptides hereof may affect or disrupt the interaction of CD40 with CD 154. Moreover, in some embodiments, administration of the peptides hereof may reduce the number of Th40 cells in the subject. These short interfering peptides (SIPs) may include an amino acid sequence selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO:

9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15. The inhibitor peptides may include a modification selected from phosphorylation and glycosylation, and/or may be linked to polyethylene glycol (PEG) molecule, and/or may be linked to one or more domains of an Fc region of mouse, human, canine, feline, or equine, IgG

immunoglobin, or the equivalent in each species, and/or may be linked to an epitope tag polypeptide comprising between 6 and 50 amino acid residues. Additionally, the inhibitor peptides hereof may also include modifications of termini such as acetylation and amidation. Additionally, peptides hereof may include several salt forms including hydrochloride salts, acetate salts, TFA salts, and sodium chloride salts.

This Summary is neither intended nor should it be construed as being representative of the full extent and scope of the present disclosure. Further, references made herein to“the present disclosure,” or aspects thereof, should be understood to mean certain embodiments of the present disclosure and should not necessarily be construed as limiting all embodiments to a particular description. The present disclosure is set forth in various levels of detail in this Summary as well as in the Drawings, Detailed Description, and no limitation as to the scope of the present disclosure is intended by either the inclusion or non-inclusion of elements. Moreover, the materials, method, and examples are illustrative only and are not intended to be limiting, nor should they be construed as limiting the embodiments.

Additional features and aspects of the developments hereof will be apparent from the following detailed description and claims.

Brief Description of the Figures

FIG. 1 is a chart of the effect of various peptides of CD 154 on the development of diabetes in NOD mice. The 6-mer (SEQ ID NO: 46), 8-mer (SEQ ID NO: 22), 10-mer (SEQ NO: 41), 13- mer (SEQ ID NO:42), 15-mer (SEQ ID NO: 24), and 24-mer (SEQ ID NO:43) were tested.

FIG. 2A is a chart of the effect of a 15-mer peptide from CD154 on the CD4/CD8 ratio in NOD mice.

FIG. 2B is a chart of the effect of 15-mer peptide in treated versus control pancreata excised, examined, and scored.

FIG. 3 is a graph of reversal of diabetes in NOD mice using a 15-mer peptide from CD 154.

FIG. 4 is a dot-plot of the detection of Th40 cells using a 15-mer peptide from CD 154.

FIG. 5 is a dot-plot of a screening of B cells using a 15-mer peptide from CD 154.

FIG. 6 is a chart demonstrating a comparison of Th40 cell levels in diabetic and non diabetic mice.

FIG. 7 is a chart demonstrating the effect of treatment with the 15-mer peptide on insulin granulation of the pancreas. FIG. 8 is a graph that shows the effect of mutations in the 15-mer peptide on the ability of the 15-mer peptide to inhibit development of diabetes in NOD mice.

FIG. 9 is a chart showing the number of cells (xlO 6 ) of CD3+CD4+CD40+ in different mice models.

FIG. 10 is a chart showing the percentage of TH40 cells in the peripheral blood in human subjects in control and diabetic subjects.

FIG. 11 is a dot-plot of detection of Th40 cells in canine blood (of canine known to have diabetes) using a 6-mer peptide (SEQ ID NO: 46) from CD154.

FIG. 12 is a graph of the blood glucose levels of diabetic dog subjects pre- and post treatment.

FIG. 13 is a graph of fructosamine serum values in diabetic dog subjects pre- and post treatment.

FIG. 14a, FIG. 14b, FIG. 14c, FIG. 14d, and FIG.14e provide graphs of c-peptide levels in diabetic dog subjects before, during, and at the end of treatment.

FIG. 15 provides a chart of Th40 percentage in canine peripheral blood.

FIG. 16 provides a graph showing the daily blood glucose average for a male diabetic dog subject prior to treatment, during treatment, and post-treatment.

FIG. 17 provides a graph showing the daily blood glucose average for a female diabetic dog subject prior to treatment, during treatment, and post-treatment.

FIG. 18 provides a graph comparing the peripheral Th40 cell percentages in mouse, human, and dog subjects.

FIG. 19 provides tables of blood counts for a diabetic dog subject, before and during treatment.

FIG. 20 provides tables of blood counts for a diabetic dog subject, before and during treatment.

FIG. 21 is a graph demonstrating the effect of a peptide treatment on hyperglycemia in diabetic NOD mice.

FIG. 22 is a graph of binding of a peptide to live cells in culture over time.

FIG. 23 provides a graph demonstrating that SEQ ID NO: 11 regulates the inflammatory cytokine IFNy without impacting production of non-inflammatory cytokine IL-4. FIG. 24 provides a graph showing that different concentrations of peptide of SEQ ID NO:46 affects or influences levels of IL-2, IFNy, and IL-17A.

FIG. 25 provides a graph showing the effect that treatment with SEQ ID NO: 13 had on ketones measured in the urine of the dog subjects.

Detailed Description

This disclosure provides bioactive peptides. These peptides are useful for preventing, modulating, and/or treating a variety of indications and diseases, which are discussed in detail below. In some embodiments, the peptides hereof affect the interaction and/or binding of the CD40 complex with its ligand CD 154.

The present developments provide novel compositions and methods for treating, modulating, and/or reducing the severity of diabetes in animal subjects. In some instances, diabetes may arise as a result of beta cells being infiltrated and destroyed which in turn results in the loss or reduction of ability to produce insulin. The developments hereof also relate to treatment of these conditions which is demonstrated in results of blood glucose measurements, fructosamine serum values, c-peptide levels, and peripheral Th40 levels. Thus the present developments also relate to methods of treating subjects before, during, and/or after the development of diabetes, with a specific peptide or pharmaceutical composition that may reduce signs and symptoms of diabetes, reduce Th40 levels in the blood, and reduce, modulate, and/or regulate hyperglycemia.

Thus, the developments and implementations disclosed herein and hereof may include compositions of matter and methods of treating and/or reducing the diabetes. These methods include administering to a subject in need of a treatment an effective amount of a peptide to inhibit, interfere, interact, and/or affect the binding or signaling between proteins associated with the CD40 complex, including but not limited to CD 154. It should also be appreciated, that the diseases or disorders implicated may also include autoimmune conditions such as type 1 diabetes (T1D), multiple sclerosis (MS), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Crohn’s disease, inflammatory bowel disease (IBS), chronic obstructive pulmonary disease (COPD) including types of autoimmune asthma, atherosclerosis, vasculitis, hypertension, thyroiditis including Hashimoto’s and Graves diseases, primary biliary cirrhosis, Paget’s disease, Addison’s disease, acute respiratory distress syndrome (ARDS), acute lung injury, and aseptic chronic inflammation (ACI) associated with organ transplantation. The present subject matter is based on the discovery that a unique subset of T-cells, which express CD40 protein, and thus are referred to as Th40 cells, may be instrumental in autoimmune inflammation. Moreover, involvement of Th40 cells in the autoimmune process may be dependent on the interaction between CD40 protein expressed on the surface of the T-cell, and CD 154 protein. Interaction of CD40 complex and CD 154 results in activation signals being delivered between the cells, and subsequent activation of the Th40 cell. Such activation results in propagation of the Th40 cell and an increase in inflammation (e.g., an increase in the number of immune cells and immunoregulatory molecules, present in the system). Accordingly, inhibition of the CD40/CD154 interaction can modulate Th40 cell activity, and thereby affect inflammation. Thus, the present subject matter relates to the peptides, and administration thereof, that may affect the interaction between a CD40 protein and a CD 154 protein, thereby modulating inflammation. Moreover, the present subject matter relates to peptides that affect the interaction between CD40 protein expressed on the surface of a T-cell, and a CD 154 protein, thereby affecting T-cell activity, controlling inflammation, and consequently preventing, modulating, and reducing diabetes mellitus and/or other autoimmune diseases. The present subject matter also encompasses the use of such peptides to detect Th40 cells.

A unique T cell subset has been shown to be instrumental in the development of autoimmune disease. These cells are phenotypically characterized as CD41oCD40+ (Waid, D.M., et al., Eur. J. of Immunol., 34: 1488, 2004; Vaitaitis, G.M., et al., Cutting Edge, ./. Immunol ., 170:3455, 2003; Wagner, D.H., Jr., et al., Proc. Nad Acad. Sci. USA, 99:3782, 2002; Wagner, D.H., Jr., et al., Int’l J. of Mol. Med. 4:231, 1999), and are referred to as Th40 cells. (Waid, D.M., et al. (2004) Eur. J. of Immunol. 34: 1488; Vaitaitis, G.M., et al., Cutting Edge, J. Immunol. 170:3455, 2003; Wagner, D.H., Jr., et al., Proc. Nat Acad. Sci. USA 99:3782, 2002; Wagner, D.H., Jr., et al., Int’l J. of Mol. Med. 4:231, 1999). Th40 cells may be any CD4+ T cell that co expresses CD40 and CD4hi or CD41o. CD40 expression typically is associated with antigen presenting cells and the majority of prior art describes CD40 as being expressed on B cells, macrophages, monocytes, and other cells; however, CD40 proteins are also expressed on T cells (Waid, D.M., et al., 2004. Eur. J. of Immunol., 34: 1488, 2004; Vaitaitis, G.M., et al., Cutting Edge, J. Immunol., 170:3455, 2003; Wagner, D.H., Jr., et al., Proc. Nat’lAcad. Sci. USA, 99:3782, 2002; Wagner, D.H., et al., Int’l. J. of Mol. Med., 4:231, 1999; Bourgeois, C., et al., Science, 297:2060, 2002; Fanslow, W.C., et al., J. oflmmun., 152:4262, 1994; Ramsdell, F., et al., J. of Immunol. 152:2190, 1994; Grabstein, K.H., et al., J. of Immunol., 150:3141, 1993; Armitage, R.J., et al., Sem. in Immun., 5:401, 1993; Cooper, C.J., et al., J of Immunol., 173:6532, 2004). While Th40 cells comprise a proportion of the peripheral CD4+ compartment in naive, non-autoimmune mice (Waid, D.M., et al., Eur. J. of Immunol., 34:1488, 2004; Wagner, D.H., Jr., et al., Int’lJ. of Mol. Med., 4:231, 1999), and in humans (Waid. D.M., et al., Clin. Immunol.. 124: 138, 2007), this proportion is drastically expanded to as much as 50% of the CD4+ compartment in autoimmune prone mice (Waid, D.M., et al., Eur. J. of Immunol. 34:1488, 2004; Wagner, D.H., Jr., et al., Proc. Nat Ί Acad. Sci. USA 99:3782, 2002; Wagner, D.H., et al., Int Ί J. of Mol. Med., 4:231, 1999) and humans (Waid, D.M., et al., Eur. J. of Immunol. 34: 1488, 2004; Waid. D.M., et al., Clin. Immunol. 124: 138, 2007). These T cells do not express early activation markers and occur in the naive phenotype of non-challenged mice.

In NOD (non-obese diabetic) mice, Th40 cells occur at exaggerated levels in spleen, lymph nodes and the pancreas, even prior to diabetes onset (Waid, D.M., et al., Eur. J. of Immunol. 34: 1488, 2004; Wagner, D.H., Jr., et al., Proc. Nat Ί Acad. Sci. USA 99:3782, 2002). An elevated number and percentage of these T cells are seen in peripheral blood of type 1 diabetic (T1D) patients when compared to non-autoimmune controls and type 2 diabetic patients (Waid. D.M., et al., Clin. Immunol., 124: 138, 2007).

The observed increase in Th40 cells could mean that those T cells are antigen responsive or that CD40 expression is activation induced. Furthermore, several diabetogenic T cell clones are CD40+ (Wagner, D.H., Jr., et al., Proc. Nat’l Acad. Sci. USA 99:3782, 2002). Purified primary Th40 cells from NOD mice and from pre-diabetic NOD (12 - weeks of age) mice successfully transfer type 1 diabetes to NOD/scid (Non-Obese Diabetic/Severe Combined Immunodeficiency) recipient mice, directly demonstrating pathogenicity of the Th40 T cell subset (Waid, D.M., et al., Eur. J. of Immunol. 34: 1488, 2004; Wagner, D.H., Jr., et al., 2002. Proc. Nat’l Acad. Sci. USA, 99:3782, 2002). It has been shown that Th40 cells infiltrate islet beta cells destroying insulin production thus suggesting islet antigen specificity (Waid, D.M., et al., Eur. J. of Immunol. 34: 1488, 2004; Wagner, D.H., Jr., et al., Proc. Nat’l Acad. Sci. USA 99:3782, 2002). It has also been shown that Th40 cells are required for diabetes transfer. Peripheral (spleen and regional lymph node) T cells that were CD40 depleted, then CD25, Treg, depleted were not capable of transferring diabetes to Scid (Severe Combined Immunodeficiency) recipients. Even though Treg cells were removed, if the auto-aggressive CD40+ T cells subset is absent, disease transfer does not occur.

While Th40 cells are important in the development of autoimmunity, another important factor is expression of the CD40-Ligand, CD 154. CD 154 is temporally induced on activated T- cells in response to CD3/TCR stimulation (Lederman, S. et al., ./. of Exp. Med., 175: 1091, 1992). CD 154 expression has also been demonstrated on platelets, monocytes, basophils, eosinophils, dendritic cells, fibroblasts, smooth muscle, and endothelial cells (Russo, S. et al., J. Immunol. 171 :5489, 2003; Stumpf, C., et al., Eur. J. Heart Fail., 5:629, 2003; Schonbeck, U., et al., Cell Mol. Life Sci. 58:4, 2001). CD154 is a member of the tumor necrosis factor (TNF) super-family and a soluble form of CD154 (sCD154) has been described (Russo, S., et al., J. Immunol. 171 :5489 2003; Stumpf, C., et al., Eur. J. Heart Fail 5:629, 2003; Toubi, E., et al., Autoimmunity 37:457, 2004). Therefore, sCD154 may act like a cytokine (Stumpf, C., et al., Eur. J. Heart Fail. 5:629, 2003). Even though CD154 has not been genetically linked in T1D studies, sCD154 is significantly elevated in T1D and may play a role in the disease process (Varo, N. et al., Circulation 107:2664, 2003; Cipollone, F., Q\ d\., Diabetologia %. \2\6, 2005; Devaraj, S., et al ., Diabetes 55:774, 2006). The importance of CD40-CD154 interaction in autoimmunity has been established (Wagner, D.H., Jr., et al., Proc. Nat Ί Acad. Sci. USA 99:3782, 2002; Kobata, T., et al., Rev. Immunogenet. 2:74, 2000; Homann, D., et al., Immunity 16:403, 2002; Goodnow, C. C., et al., Lancet 357:2115, 2001; Balasa, B., et al., J. of Immunol. 159:4620, 1997). Blocking CD40-CD154 interaction may prevent collagen induced arthritis, (Durie, F.H., et al., Science 281 : 1328, 1993) experimental autoimmune encephalitis (Howard, L.M., et al., Autoimmunity 37:411, 2004), prostatitis (Grossman, M.E., et al., J. Immunother. 24:237, 2001), and type-1 diabetes in the NOD mouse model (Durie, F.H. et al., Science 281 : 1328, 1993; Balasa, B. et al., Journal of Immunology 159:4620, 1997; Howard, L.M., et al., Autoimmunity 37:411, 2004; Grossman, M.E. et al., J. Immunother. 24:237, 2001). In the diabetes model, it was essential to administer a CD154 blocking antibody to NOD mice at 3- weeks of age because at 9-weeks, blocking antibodies had no effect on diabetes prevention (Balasa, B. et al., J. of Immunol. 159:4620, 1997).

Previous work has also demonstrated that the Th40 cell subset induces RAG1 and RAG2 (Recombination-Activating Genes) transcription, translation and nuclear translocation (Vaitaitis, G.M., et al., Cutting Edge, J. Immunol. 170:3455, 2003) when CD40 is engaged (Vaitaitis, G.M. et al., Cutting Edge, J. Immunol. 170:3455, 2003). CD3 engagement does not induce RAGl or RAG2 in T-cells (Vaitaitis, G.M., et al., Cutting Edge, J. Immunol. 170:3455, 2003). Subsequent to RAG1/RAG2 induction, CD40-mediated T-cell receptor (TCR) revision occurs in peripheral T cells (Vaitaitis, G.M. et al., Cutting Edge, J. Immunol. 170:3455, 2003). CD40 induction of TCR revision is RAG dependent. T cells isolated from a TCR-Tg mouse undergo TCR revision when CD40 engaged, but T-cells from the TCR-Tg.RAG-/- mouse do not TCR revise when CD40 engaged (Wagner, D.H., Jr. et al., Int’lJ. of Mol. Med. 4:231, 1999).

CD40 is a 50-kDa integral membrane protein of the tumor necrosis factor receptor (TNF- R) family. It is constitutively expressed as a homotrimer (Foy TM, et al., Ann. Rev. of Immunol., 14:591, 1996). In general, stimulation of all CD40-expressing cell types induces operations which contribute to inflammation, such as enhancement of costimulatory and adhesion molecules, and up-regulation of proteolytic enzymes (Mach, F. et al., Atherosclerosis. 137 Suppl:S89-95, 1998).

CD40’s ligand - CD 154 - is a 39-kDa protein that belongs to the tumor necrosis factor (TNF) family. CD40 forms a trimer that binds CD 154 at the interface of the three monomers. CD 154 is expressed commonly on cells beyond the surface-expressed CD 154, as CD 154 may also exist in a soluble biologically active form (sCD154) that is shed from the cell surface after activation. The main source of sCD154 is platelets. (Foy TM, et al., Ann. Rev. of Immunol., 14:591, 1996).

Genetically manipulated mouse models, including models such as the NOD mouse, are utilized for research and development concerning autoimmune diseases including diabetes because the NOD mouse spontaneously develops T1D. Prior studies in humans and mice have attempted to block the CD40/CD154 interaction by using monoclonal antibodies and this approach has proven efficacious in several mouse model studies and in several human studies; however, the use of monoclonal antibodies to target the CD40/CD154 dyad was abandoned due to thromboembolic events which may have been related to the functioning of CD 154 in thrombus stabilization. It is postulated that CD 154 stabilize thrombi by interaction with the integrin aiihb , and by inhibiting CD 154, thrombi may be less stable, and as a consequence shed emboli causing thrombotic events.

Multiple treatment options have been put forward to address and control both chronic and acute inflammation. Many approaches use non-steroidal anti-inflammatory drugs (NSAIDs) that attack the production of leukotrienes and prostaglandins, cellular products that cause localized inflammation. Other approaches use more powerful immunosuppressant drugs such as cyclophosphamide, methotrexate and azathioprine that suppress the immune response and stop the progression of the disease. Still other treatments involve the use of monoclonal antibodies (mAh) designed to alter the immune responses to self-tissues, as occurs during autoimmune diseases. However, all of these treatments often may have severe, long-term side effects.

Current immune-modulatory therapies may rely upon monoclonal antibody treatments that may give rise to complications. For example, antibodies administered to a subject may cross-react with unintended targets and cause severe nephritic complications and those that specifically act against CD 154 may cause embolic complications. Further, the CD40-CD154 interaction may play an important role in antibody generation which may indicate that administration of a monoclonal antibody could induce auto-antibody generation and further complications, which may inhibit the restoration of normal immune function (see generally Banchereau, J. et al., Annu. Rev. of Immunol. 12:881, 1994).

Before the present development is further described, it is to be understood that developments hereof are not strictly limited to particular embodiments described, as such may of course vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the claims.

It must be noted that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It should further be understood that as used herein, the term“a” entity or“an” entity refers to one or more of that entity. For example, a nucleic acid molecule refers to one or more nucleic acid molecules. As such, the terms“a”,“an”,“one or more” and”at least one” can be used interchangeably. Similarly, the terms“comprising”,“including” and“having” can be used interchangeably.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates, which may need to be independently confirmed.

It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the developments, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub combination. All combinations of the embodiments are specifically embraced by the present invention and are disclosed herein just as if each and every combination was individually and explicitly disclosed. In addition, all sub-combinations are also specifically embraced by the present invention and are disclosed herein just as if each and every such sub-combination was individually and explicitly disclosed herein.

It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as "solely," "only" and the like in connection with the recitation of claim elements, or use of a "negative" limitation.

Furthermore, as used herein the term animal refers to a vertebrate, preferably a mammal. Suitable mammals on which to use the methods of the present invention include but are not limited farm animals, sports animals, pets, primates, mice, rats, horses, dogs, cats, and humans. The term animal can be used interchangeably with the terms subject or patient.

One embodiment of the present subject matter is a peptide that interacts with a CD40 protein in such a manner as to prevent diabetes mellitus and/or autoimmune disease. As used herein, the terms interact, interaction, and the like, mean that two molecules come into sufficient physical proximity such that they cause a modulation of inflammation. One such type of interaction is a binding interaction. In such an interaction the peptide associates with CD40 to form a complex. An example of complex formation is the association of an antigen with an antibody. According to the present subject matter, binding of a peptide hereof to a CD40 protein can be reversible (e.g., non-covalent binding interactions) or non-reversible (e.g., covalent binding interactions). Moreover, a reversible interaction can be strong or weak, the strength of the interaction being determined by the forces (e.g., ionic charges, hydrogen binding, van der Walls interactions, etc.) exerted by each protein on the other protein in the complex. Factors affecting the strength of an interaction between two molecules are known to those skilled in the art. One useful measure of the strength of binding between two molecules, such as a peptide and a protein, is the dissociation constant (Kd). Preferred peptides of the present invention are those that bind to a CD40 protein with a Kd of no more than about 1 x 10 6 M, about 1 x 10 7 M, or about 1 x 10 8 M. Particularly preferred peptides are those having a Kd of less than about 1 x 10 9 M. In one embodiment, a peptide hereof binds to a CD40 protein with a Kd of less than 100 nM, less than 50 nM, less than 25 nM, less than 10 nM, less than 5 nM, less than 3 nM, less than 2 nM, or less than 1 nM. Methods of measuring and analyzing binding interactions between a peptide and a CD40 protein are known by those of skill in the art.

As used herein, to modulate inflammation means to modify or affect the interaction between CD40 and/or CD40 complex and/or modify or affect inflammation more generally. As used herein, the terms level, number, count and concentration can be used interchangeably.

Modulation of inflammation may result in an increase or decrease in the number of Th40 cells present in the inflammatory environment. Consequently, modulation can be referred to as positive or negative. Positive modulation (also referred to as up-regulation) of inflammation may refer to an increase in the number of Th40 cells in the inflammatory environment. Negative modulation (also referred to as down-regulation) of inflammation may refer to a reduction in the number of Th40 cells present in the inflammatory environment. A preferred peptide is one that down- regulates inflammation, and thereby may reduce the number of Th40 cells present in the inflammatory environment. Positive and negative modulation of inflammation may or may not result in a change in the type and amount of immunoregulatory molecules present in the inflammatory environment.

It will be appreciated by those skilled in the art that both a cell culture system and the immune system of an animal comprise basal levels of immune cells and immunoregulatory molecules. The phrases basal level and normal level can be used interchangeably. With regard to the immune system of an animal, as used herein, the basal level of a type of immune cell (e.g., Th40 cell), or a immunoregulatory molecule, refers to the average number of that cell type, or immunoregulatory molecule, present in a population of individuals considered healthy (i.e., free of metabolic, autoimmune, or infectious disease). With regard to a cell culture system, as used herein, the basal level of a type of immune cell, or an immunoregulatory molecule, refers to the average level of that cell type, or immunoregulatory molecule, present in a population of cells that is non-activated. Those skilled in the art are capable of determining if a T-cell, or a population of such cells, is activated. For example, in some instances the expression of CD69, CD25 and/or CD 154 proteins by a cell indicates that the cell has been activated.

The basal level of a cell or molecule can be a specific amount (e.g., a specific

concentration) or it can encompass a range of amounts. Basal levels, or ranges, of immune cells and immunoregulatory molecules are known to those in the art. For example, in a healthy individual, the normal level of CD4+ T-cells present in human blood is 500-1500 cells/ml.

Variability in this measurement can result from differences in the method used to determine the cell count. Furthermore, normal levels of cells can also be reported as a percentage of a total cell population. For example, in a healthy individual, Th40 cells may make up less than 25% of the total T cell population. Thus, as used herein, the term inflammation may refer to an inflammatory environment in which Th40 cells make up greater than about 25%, greater than about 30%, greater than about 35%, greater than about 40%, greater than about 45% , greater than about 50%, greater than about 55%, greater than about 60%, greater than about 65%, greater than about 70%, greater than about 75%, or greater than about 80% of the total T-cell population. Moreover, a preferred peptide herein is one that may reduce the level of Th40 cells to less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 27%, or equal to about 25% of the total T-cell population. Methods of measuring different types of T- cells in the T-cell population are known to those skilled in the art. Furthermore, a novel method for detecting Th40 cells using peptides hereof is disclosed herein.

As used herein, the phrase inflammatory environment refers to the overall population of immune cells, and related immunoregulatory molecules, that are present in a culture of cells, or in the body of an animal. As such, the phrase inflammatory environment encompasses the types, and/or the relative amounts of immune cells and immunoregulatory molecules (e.g., cytokines) present in a culture of cells, or in an animal, which are involved in affecting an inflammatory reaction. Examples of cells encompassed by the term inflammatory environment include, but are not limited to, T cells, neutrophils, macrophages, granulocytes, and the like. The inflammatory environment relates to cells and molecules that mediate both acute and chronic inflammation. It will be appreciated by those skilled in the art that the inflammatory environment refers to the system to which peptides hereof are administered. In one embodiment, the system is a cell culture system. In one embodiment, the system is a whole animal. A preferred peptide hereof is one that selectively interacts with a CD40 protein in solution, as determined using an assay such as an immunosorbent assay, or on the surface of a T-cell. As used herein, the terms selectively, selective, specific, and the like, indicate the peptide has a greater affinity for a CD40 protein than it does for proteins unrelated to the CD40 protein. More specifically, the terms selectively, selective, specific, and the like indicate that the affinity of the peptide for CD40 is statistically significantly higher than its affinity for a negative control (e.g., an unrelated protein such as albumin) as measured using a standard assay (e.g., ELISA). Suitable techniques for assaying the ability of a peptide to selectively interact with a CD40 protein are known to those skilled in the art. Such assays can be in vitro or in vivo assays. Examples of useful assays include, but are not limited to, an enzyme-linked immunoassay, a competitive enzyme-linked immunoassay, a radioimmunoassay, a fluorescence immunoassay, a

chemiluminescent assay, a lateral flow assay, a flow-through assay, an agglutination assay, a particulate-based assay (e.g., using particulates such as, but not limited to, magnetic particles or plastic polymers, such as latex or polystyrene beads), an immunoprecipitation assay, an immunoblot assay (e.g., a western blot), a phosphorescence assay, a flow-through assay, a chromatography assay, a polyacrylamide gel electrophoresis (PAGE)-based assay, a surface plasmon resonance assay, a spectrophotometric assay, a particulate-based assay, an electronic sensory assay and a flow cytometric assay. Methods of performing such assays are well known to those skilled in the art. In one embodiment, an assay can be performed using cells in culture, or it can be performed in a whole animal. Assays can be designed to give qualitative, quantitative or semi-quantitative results, depending on how they are used and the type of result that is desired.

One embodiment hereof is a peptide that interacts with a CD40 protein in such a manner as to affect the interaction of the CD40 protein with a CD 154 protein. The effect of the peptide on the CD40/CD154 interaction can be positive or it can be negative. For example, the peptide may interact with the CD40 protein in such a manner that the strength of the interaction between the CD40 protein and a CD 154 protein is increased. Alternatively, the peptide may interact with the CD40 protein such that the strength of the interaction between the CD40 protein and a CD 154 protein is decreased. Methods of measuring the strength of binding between the peptide and a CD40 protein are known to those skilled in the art. A preferred peptide hereof is one that reduces the strength of the interaction between a CD40 protein and a CD 154 protein. Preferred peptides hereof may reduce the strength of binding between a CD40 protein and a CD 154 protein by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%. A particularly preferred peptide is one that completely inhibits binding of CD40 to CD 154. Complete inhibition of binding between CD40 and CD 154 means that when a peptide hereof is brought into proximity with a CD40 protein and a CD 154 protein under conditions that would normally allow the interaction of CD40 and CD 154, no such interaction occurs and activation signals are not stimulated in the CD40-expressing cell.

Consequently CD40/CD154 mediated modulation of inflammation does not occur. In one embodiment, the peptide interacts with the CD40 protein in such a manner as to reduce the level of inflammation in the system. In one embodiment, the peptide interacts with the CD40 protein in such a manner as to inhibit the development of inflammation in the system.

While peptides hereof may interact with any site on the CD40 protein, preferred peptides interact with the CD40 protein at a location that overlaps with the CD 154 binding site. In one embodiment, a peptide hereof interacts with the CD40 protein at the CD 154 binding site. An example of such a peptide is a CD40 ligand competitive antagonist. As used herein, peptides that interfere with, or inhibit, the binding of a CD 154 protein to a CD40 protein are referred to as small interfering peptides (SIPs). As used herein a small interfering peptide is a peptide that, through physio-chemical properties, interferes with the interaction of a CD40 protein with a CD 154 protein, thereby preventing activation signals from being delivered to the CD40-bearing cell, thus limiting the activation of the CD40-bearing cell, and consequently, inflammation. As

demonstrated herein, the consequences of such interference are prevention of T-cell activation and propagation, and a prevention or reduction of inflammation. As demonstrated herein, in some instances the results of such inhibition or prevention of interaction between CD40 and CD 154 may include observable data that demonstrates that diabetes mellitus, and characteristics of diseases associated therewith, are prevented, modulated, and/or reduced.

Additionally, a small interfering peptide, may, through its physio-chemical properties, interfere with the interaction of a CD40 protein with a CD 154 protein, thereby preventing activation signals from being delivered to the CD40-bearing cell, thus limiting the activation of the CD40-bearing cell, and consequently, modulating, inhibiting, and preventing diabetes mellitus and/or autoimmune disease. As demonstrated herein, the consequences of such interference are prevention of T cell activation and propagation, and a prevention, reduction, or modulation of diabetes mellitus related developments. The peptides hereof may also be modified to contain, conjugated to, and/or administered with phosphorus, sulfur, manganese, magnesium, calcium, halogens, metals, etc. Amino acid mimics may be used to produce polypeptides, and therefore, the polypeptides of this disclosure may include amino acids mimics that have enhanced properties, such as resistance to degradation.

In the methods disclosed herein, the bioactive peptide may be administered via

intramuscular (IM) delivery, intravenous (IV) delivery, subcutaneous (SC) delivery, oral delivery, gavage delivery, emollient/skin delivery, transdermal patch, and/or intranasally.

The peptides and methods hereof may also comprise administering pro-drugs that metabolize to an active form of these peptides. As used herein, a“pro-drug” is a compound that a biological system metabolizes to an active compound as a result of spontaneous chemical reaction(s), enzyme catalyzed reaction(s), and/or metabolic chemical reaction(s), or a combination of each. Standard prodrugs are formed using groups attached to functionality, e.g. HO-, HS-, HOOC-, R2N-, associated with the drug, that cleave in vivo. Standard prodrugs include, but are not limited to, carboxylate esters where the group is alkyl, aryl, aralkyl, acyloxyalkyl,

alkoxycarbonyloxyalkyl as well as esters of hydroxyl, thiol and amines, where the group attached is an acyl group, an alkoxycarbonyl, aminocarbonyl, phosphate or sulfate. The groups illustrated are exemplary and not exhaustive, and one skilled in the art could prepare other known varieties of prodrugs. Prodrugs must undergo some form of a chemical transformation to produce the compound that is biologically active or is a precursor of the biologically active compound. In some cases, the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved bioavailability, pharmacodynamic half-life, etc.

The methods hereof may include the treatment of a subject developing diabetes mellitus, suspected of developing diabetes mellitus, or likely to develop diabetes mellitus, in order to decrease or prevent complications and problems associated with developing diabetes mellitus and/or autoimmune diseases. In these methods, the blood glucose levels of the subject may be reduced to less than 160 mg/dl, less than 150 mg/dl, less than 140 mg/dl, less than 130 mg/dl, less than 120 mg/dl, or less than 110 mg/dl.

The methods are not limited to use to only type 1 diabetes mellitus, as these methods may be used to treat hyperglycemia resulting from any condition.

When administered or co-administered, the compounds of the present developments are given in a therapeutically effective amount to the subject. As used herein, the phrase “therapeutically effective amount” means an amount that has any beneficial effect in treating the condition, disease, or symptoms, or preventing or reducing the development of disease, condition, syndromes, or disorders. Determining the therapeutically effective amount of the bioactive peptide to administer of the current developments is a matter of optimization and titration in the art and may vary depending on such factors as, but not limited to, the degree of progression or extent of the disease, condition, syndrome, or disorder to be treated, the age and condition of the subject, the weight and surface area of the subject, and other such ascertainable traits that may be pertinent and relevant to the treatment of the subject.

The present developments provide novel bioactive peptides, variants, homologs, orthologs, and/or derivates thereof related to peptides as disclosed herein. The sequences of some of such peptides are shown below in Table 1.

Table 1.

In one aspect, the developments feature peptides of 6 amino acids in length, wherein the peptide comprises the amino acid sequence of Formula I:

X 1 -X 2 -X 3 -X 4 -X 5 -X 6

Wherein,

X 1 is K, E, P, or Q or a small naturally or non-naturally occurring amino acid;

X 2 is K or a small naturally or non-naturally occurring amino acid;

X 3 is G or a small naturally or non-naturally occurring amino acid;

X 4 is Y;

X 5 is Y;

X 6 is T.

In some embodiments, the amino acid sequence of said peptides differs by at least one, or at least two, or at least three amino acids as compared to the amino acid sequences set forth in SEQ ID NOs: 7-10. In another aspect, the developments feature peptides of 15 amino acids in length, wherein the peptide comprises the amino acid sequence of Formula II:

gΐ g2 g3 g4 g5 g6 g7 g8 g9 gΐq gΐ ΐ g12 g13 g14 g15

L -L "L -L "L -L "L -L -L -L -L -L -L -L -L

Wherein,

X s V;

X 2 is L;

X 3 is Q or R;

X 4 is W;

X 5 is A;

X 6 is K, E, P, or Q;

X 7 is K;

X 8 is G;

X 9 is Y;

X 10 is Y;

X 11 is T;

X 12 is M or I;

X 13 is K or S;

X 14 is S or N;

X 15 is N.

In some embodiments, the amino acid sequence of said peptides differs by at least one, or at least two, or at least three amino acids as compared to the amino acid sequence set forth in SEQ ID Nos: 11-15.

The present developments provide additional novel bioactive peptides, variants, homologs, orthologs, and/or derivates thereof related to peptides as disclosed herein. The sequences of such peptides are shown below in Table 2.

Table 2.

In some embodiments, the peptides disclosed herein may be peptides that bind to CD40 and/or the CD40 complex. Further, the peptides hereof may be short interfering peptides and may disrupt, affect, or alter the interaction of CD40 with CD 154.

In some embodiments, the peptide is isolated from a protein recombinantly produced in a cell. The cell used for recombinant production may be a prokaryotic cell or a eukaryotic cell. In some embodiments the peptide is chemically synthesized. A preferred implementation is the peptide that is chemically synthesized.

In some embodiments, the peptide is phosphorylated at a serine, threonine, or tyrosine residue.

In some embodiments, the peptide is modified at its amino terminus. Examples of amino terminal modifications include an N-glycated, N-alkylated, N-acetylated, or N-acylated amino acid. In some embodiments, the peptides include a C-terminal amidated amino acid. In other embodiments, the peptide does not include an amidated amino acid at its carboxy terminus. In some implementations of the peptide the C-terminus may include a protecting group.

In some embodiments, the peptide is pegylated. The developments in another implementation includes a pharmaceutical composition comprising any one of the peptides of SEQ ID Nos: 7-15 and a pharmaceutically acceptable carrier.

The developments in another embodiment includes a pharmaceutical composition comprising any one of the peptides of SEQ ID Nos: 21-25; SEQ ID Nos:41-48; and SEQ ID NOs:50-52 in a pharmaceutically acceptable carrier.

In some embodiments, the peptide is less than 225, 200, 175, 150, 125, 100, 75, 50, 30, 25, 20, 15, 10, 9, 8, 7, 6, or 5 amino acids in length.

The peptides hereof may be modified to include one or more modifications. Thus, in some embodiments, the c-terminus is amidated. In other embodiments, the n-terminus is acetylated. In other embodiments, both modifications are present. In yet other implementations, neither modification is present.

The present developments may also relate to treating subjects, of human, canine, feline, and equine varieties for a variety of autoimmune diseases, including but not limited to diabetes mellitus whether insulin deficiency diabetes and insulin resistance diabetes, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosa, chronic obstructive pulmonary disease, and/or acute respiratory distress syndrome.

The present developments are based on the knowledge that interaction of CD40-ligand (CD 154 protein) with CD40 protein expressed on T-cells (Th40 cells), may be important in the development of autoimmune disease and type 1 diabetes mellitus. The present developments may be based on the elucidation of the critical residues in CD40 and CD 154 that may be important for this interaction. The present developments relate to affecting, modulating, and/or modifying the interaction between a CD40 protein and a CD 154 protein through the use of small peptides that interact with the CD40 protein at a site where the CD 154 protein would normally bind. In some aspects, the present developments may also relate to using such peptides to reduce the level of Th40 cells, thereby reducing the severity of disease.

One implementation of the present developments is a method for preventing diabetes mellitus in a subject, comprising contacting the CD40 protein with a peptide that interacts with the CD40 protein. Preferred peptides are those that are less than 25 amino acids in length, and that bind to a CD40 protein, thereby inhibiting its interaction with a CD 154 protein. One implementation of the present developments is a method for preventing autoimmune disease in a subject, comprising contacting the CD40 protein with a peptide that interacts with the CD40 protein. Preferred peptides are those that are less than 25 amino acids in length, and that bind to a CD40 protein, thereby inhibiting its interaction with a CD 154 protein.

One implementation of the present developments is a method for preventing, reducing, ameliorating, and/or modulating diabetes mellitus in a subject, comprising administering to a subject in need thereof, a therapeutically effective amount of a peptide that reduces, modulates, and/or affects the blood glucose level of the subject.

One implementation of the present developments is a method for preventing, reducing, ameliorating, and/or modulating diabetes mellitus in a subject, comprising administering to a subject in need thereof, a therapeutically effective amount of a peptide that reduces, modulates, and/or affects the serum fructosamine levels of the subject.

One implementation of the present developments is a method for preventing, reducing, ameliorating, and/or modulating diabetes mellitus in a subject, comprising administering to a subject in need thereof, a therapeutically effective amount of a peptide that reduces, modulates, and/or affects the c-peptide levels of the subject.

Yet another implementation of the present developments is a method for preventing, modulating, and/or reducing diabetes mellitus, the method comprising inhibiting interaction between a CD40 protein and a CD 154 protein with a peptide that interacts with the CD40 protein. Preferred peptides interact with the CD40 protein at the CD 154-binding site. Preferably such peptides are less than 25 amino acids in length. Even more preferred peptides are those amino acid sequences selected from SEQ ID NOs: 7-15, 21-25, 41-48, and 50-52.

One implementation of the present developments is a method for preventing, modulating, and/or reducing autoimmune disease, the method comprising inhibiting interaction between a CD40 protein and a CD 154 protein with a peptide that interacts with the CD40 protein. Preferred peptides interact with the CD40 protein at the CD 154-binding site. Preferably such peptides are less than 25 amino acids in length. Even more preferred peptides are those amino acid sequences selected from SEQ ID NOs: 7-15, 21-25, and 41-48.

One embodiment of the present developments is a method for preventing, modulating, and/or reducing diabetes mellitus, the method comprising administering to a subject in need thereof, a therapeutically effective amount of a peptide that affects the interaction of CD40 with CD154/CD40-ligand. An aspect of this development may be that the peptide binds to CD40. In this embodiment, the peptide may bind to a CD40 protein with a Kd of less than 10 6 . Further, in this embodiment, the peptide may affect the interaction between CD40 and CD 154. Additionally, a preferred embodiment may inhibit the binding of CD40 to CD 154. Moreover, in this embodiment, the peptide binds to CD40 at the site where CD40 interacts with CD 154. In this embodiment, the peptide may affect the interaction of CD40 with CD 154 in such a manner as to prevent the expansion of Th40 cells. In this embodiment, the peptide may affect the interaction of CD40 with CD 154 in such a manner as to reduce the number of Th40 cells. In this embodiment, the peptide may affect the interaction of CD40 with CD 154 in such a manner as to alter the cytokine expression profile of a cell population, treated with said peptide.

One embodiment of the present developments is a method for preventing, modulating, and/or reducing autoimmune disease, the method comprising administering to a subject in need thereof, a therapeutically effective amount of a peptide that affects the interaction of CD40 with CD154/CD404igand. An aspect of this development may be that the peptide binds to CD40. In this embodiment, the peptide may bind to a CD40 protein with a Kd of lessthan 10 6 . Further, in this embodiment, the peptide may affect the interaction between CD40 and CD 154. Additionally, a preferred embodiment may inhibit the binding of CD40 to CD 154. Moreover, in this embodiment, the peptide binds to CD40 at the site where CD40 interacts with CD 154. In this embodiment, the peptide may affect the interaction of CD40 with CD 154 in such a manner as to prevent the expansion of Th40 cells. In this embodiment, the peptide affects the interaction of CD40 with CD 154 in such a manner as to reduce the number of Th40 cells. In this embodiment, the peptide may affect the interaction of CD40 with CD 154 in such a manner as to alter the cytokine expression profile of a cell population, treated with said peptide.

One embodiment of the present developments is a method to modulate and/or reduce diabetes mellitus in an animal, the method comprising administering to the animal, a peptide that interacts with a CD40 protein in such a manner as to modulate IFNy (interferon gamma). Preferred peptides are those that interact with the CD40 protein at the CD 154-binding site, thereby modulating IFNy. Preferred peptides may modulate inflammation by reducing the level of Th40 cells to no more than 25% of the total T-cell population. Such methods can be used to prevent and/or reduce diabetes mellitus and symptoms that may accompany diabetes mellitus, more generally. In this implementation a peptide may be selected from SEQ ID NOs: 7-15, 21-25, 41- 48, and 50-52.

One embodiment of the present developments is a method to identify a subject at risk for developing diabetes and/or autoimmune disease, the method comprising obtaining a sample containing T-cells from a subject to be tested, contacting the sample with a peptide that binds the CD40 protein, detecting the CD40 bound peptide, and determining the level of Th40 cells from the amount of CD40 bound, wherein a level of Th40 cells greater than 25% of the total T-cell population indicates the subject is at risk for developing diabetes mellitus and/or autoimmune disease.

Another aspect of the present developments, is a method to administer a bioactive peptide hereof to prevent, modulate, and/or reduce diabetes mellitus and/or autoimmune disease, comprising selecting one or more of the peptides of SEQ ID NO: 7-15, 21-25, 41-48, and 50-52; selecting a delivery method selected from the group comprising intramuscular (IM) delivery, intravenous (IV) delivery, subcutaneous (SC) delivery, oral delivery, gavage delivery, emollient/skin delivery, or transdermal patch; and using one the said delivery methods to deliver a therapeutically effective dose of the peptide.

Another aspect of the present developments, is a method to administer a bioactive peptide hereof to prevent, modulate, and/or reduce diabetes mellitus and/or autoimmune disease, comprising selecting one or more of the peptides of SEQ ID NO: 7-15; selecting a delivery method selected from the group comprising intramuscular (IM) delivery, intravenous (IV) delivery, subcutaneous (SC) delivery, oral delivery, gavage delivery, emollient/skin delivery, or transdermal patch; and using one the said delivery methods to deliver a therapeutically effective dose of the peptide.

Another embodiment of the present developments, is a method to administer a CD40 peptide to prevent, modulate, and/or reduce diabetes mellitus and/or autoimmune disease, in an animal, comprising selecting a peptide that interacts with a CD40 protein and CD 154 binding site, using an extended delivery method selected from the group comprising an implantable device, a hydrophilic polymer formulation, a permeable polymeric membrane, injectable gel implants, solvent extraction system, phase inversion system, thermosensitive gels, pH dependent in situ gels, microparticles, microspheres, nanoparticles, nanospheres, bio-degradable implants, or photoactivated depot. A peptide useful for practicing methods of the present developments should be of a size sufficient to interact with CD40 protein / CD40 complex in such a manner as to modulate diabetes mellitus. It is understood by those skilled in the art that preferred peptides are relatively short since they are easier and may be less expensive to produce. Preferred peptides may be those that are less than 20 amino acids in length. A preferred peptide may be one that is 4, 6, 8, 10, 13, and 15 amino acids in length.

Interaction of a CD40 protein and a CD 154 protein has been shown to occur at particular regions within each protein. The inventors have now shown that, surprisingly, a peptide comprising only a short portion of the CD 154 region that interacts with CD40 is capable of binding, affecting, and interacting with a CD40 protein, thereby modulating diabetes mellitus. Thus, one embodiment hereof is a peptide that comprises at least a portion of the amino acid sequence of a CD 154 protein such that the peptide interacts with CD40 protein in such a manner as to modulate diabetes mellitus. In one embodiment, interaction of the peptide with CD40 protein results in negative modulation of diabetes mellitus. In one aspect, the peptide comprises at least a portion or a fragment of SEQ ID NO: 1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, or SEQ ID NO:5.

In one aspect, the peptide is as short as possible yet comprises enough of the CD 154 protein to allow interaction with a CD40 protein in such a manner as to diabetes mellitus. In one embodiment, a peptide hereof comprises 6, 13 or 15 contiguous amino acids from SEQ ID NO: l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, or SEQ ID NO:5, and interacts with CD40 in such a manner as to modulate diabetes. In one embodiment, a peptide hereof comprises 6, 13 or 15 contiguous amino acids from SEQ ID NO: l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, or SEQ ID NO:5, and interacts with CD40 in such a manner as to modulate autoimmune disease. A preferred peptide comprises a core sequence of lysine-glycine-tyrosine-tyrosine (KGYY; SEQ ID NO:6 and SEQ ID NO:20), which corresponds to amino acids 142-145 of SEQ ID NO: l, amino acids 143-146 of SEQ ID NO:2, amino acids 142-145 of SEQ ID NO:3, amino acids 142-145 of SEQ ID NO:4, and amino acids 143-146 of SEQ ID NO:5. Useful peptides can comprise additional regions of sequence from SEQ ID NO: l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, and SEQ ID NO:5 that are adjacent to the core sequence, so long as the peptide is capable of modulating diabetes mellitus and/or autoimmune disease. In one embodiment hereof, a peptide comprises at least one sequence selected from SEQ ID NOs:7-15, 21-25, 41-48 and 50-52, so long as the peptide interacts with CD40 protein in such a manner as to modulate diabetes.

While peptides of the present subject matter may be selected entirely of or from sequences that are responsible for the interaction of the peptide with a CD40 protein, they may additionally contain amino acid sequences that do not interact with a CD40 protein, but which have other useful functions. Any useful, additional amino acid sequence can be added to the CD40- interacting sequence, so long as the additional sequences do not have an unwanted effect on the ability of the CD40 interacting sequence to interact with a CD40 protein. For example, in addition to the amino acid sequence responsible for interacting with a CD40 protein, a peptide hereof can contain amino acid sequences that are useful for visualizing or purifying the peptide. Such sequences act as labels (e.g., enzymes) or tags (antibody binding sites). Examples of such labels and tags include, but are not limited to, B-galactosidase, luciferase, glutathione-s-transferase, thioredoxin, HIS-tags, biotin tags, and fluorescent tags. Other useful sequences for labeling and tagging proteins are known to those of skill in the art.

Likewise, peptides hereof may be conjugated or fused to another peptide, polypeptide, or nanoparticle, so long as such modification does not affect the ability of the peptide to modulate diabetes or autoimmune disease.

Likewise, peptides hereof can be modified, so long as such modification does not significantly affect or impact the ability of the peptide to modulate diabetes, autoimmune disease, or significantly affect the peptide’s functionality in controlling and affecting blood glucose control, blood serum fructosamine levels, and/or c-peptide levels. Such modifications can be made, for example, to increase the stability, solubility or absorbability of the protein. Examples of such modifications include, but are not limited to pegylation, glycosylation, acetylation, amidation, n-terminus protection, c-terminus protection, and chemical modification of the peptide.

N-terminal modifications may include modifications provided herein, so long as such a modification does not significantly affect the peptide’s functionality in performing its desired role and providing its intended therapeutic effects. Such N-terminal modifications may include, but are not limited to: 5-FAM, 5-FAM-ahx, Abz, Acetylation, Acryl, Alloc, Benzoyl, Biotin, Biotin-Ahx, BOC, Br-Ac-, BSA, CBZ, Dansyl, Dansyl-Ahx, Decanoic acid, DPTA, Fatty Acid, FITC, FITC- Ahx, Fmoc, Formylation, Hexanoic acid, HYNIC, KLH (-NH2 of N-terminal), Laurie acid, Lipoic acid, Maleimide, MCA, Myristoyl, Octanoic acid, OVA (-NH2 of N-terminal), Palmitoyl, PEN, Stearic acid, Succinylation, and/or TMR.

C-terminal modifications may include modification provided herein, so long as such a modification does not significantly affect the peptide’s functionality in performing its desired role and providing its intended therapeutic effects. Such C-terminal modifications may include, but are not limited to: AFC, AMC, Amidation, BSA (-COOH of C-terminal), Bzl, Cysteamide, Ester (Oet), Ester (OMe), Ester (OtBu), Ester (OTBzl), KLH (-COOH of C-terminal), MAPS asymmetric 2, 4, or 8 branches, Me, NHEt, NHisopen, NHMe, OSU, OVA (-COOH of C terminal), p-Nitroanilide, and/or tBu.

Peptides hereof may be derivatives or fragments from those that are found in nature (e.g., obtained from plants, animals or microorganisms) or they may be produced in a laboratory (e.g., recombinantly or synthetically). Preferred peptides are those that are synthesized. Also encompassed are peptides that are combinations of natural and synthetic molecules. General methods for producing and isolating recombinant or synthetic peptides are known to those skilled in the art. It should be noted that, as used herein, an isolated, or biologically pure, molecule, is one that has been removed from its natural milieu. As such the terms isolated, biologically pure, and the like, do not necessarily reflect the extent to which the protein has been purified.

As has been described herein, interaction of the CD40 protein and the CD 154 protein may be necessary for involvement of Th40 cells in diabetes mellitus and autoimmune diseases.

Consequently, inhibition of the interaction between a CD40 and CD 154 protein and/or CD40 complex using peptides hereof is a useful method of affecting diabetes mellitus and autoimmune diseases. Thus, one embodiment is a method to reduce the interaction between a CD40 protein and a CD 154 protein comprising introducing into an environment containing a CD40 protein and a CD 154 protein, a peptide, that interacts with the CD40 protein in such a manner as to reduce the interaction between the CD40 protein and the CD 154 protein. In one aspect hereof, the peptide reduces the interaction between the CD40 protein and the CD 154 protein by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%. In one embodiment, the peptide reduces the interaction between the CD40 protein and the CD 154 protein by a factor of at least 10, at least 100, at least 1,000, at least 10,000. Methods of measuring the strength of the interaction between the CD40 protein and the CD 154 protein have been discussed previously, and are also know to those of skill in the art.

One embodiment hereof is a method to modulate diabetes comprising contacting a CD40 protein with a peptide that interacts with the CD40 protein in such a manner as to modulate inflammation. In one aspect, interaction of the peptide with the CD40 protein may increase or decrease the number of Th40 cells by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%. In one embodiment, interaction of the peptide with the CD40 protein may increase or decrease the number of Th40 cells by a factor of at least 10, at least 100, at least 1,000, at least 10,000.

One aspect is a method to prevent, reduce, or modulate diabetes mellitus in a subject, the method comprising administering a peptide hereof to the patient. In one embodiment, the peptide comprises an amino acid sequence selected from SEQ ID NOs:7-15, 21-25, 41-48, and 50-52. In one embodiment, the peptide is an amino acid sequence selected from SEQ ID NOs:7-15. In a preferred embodiment, interaction of the peptide with the CD40 protein may decrease the number of Th40 cells by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%. In another embodiment, interaction of the peptide with the CD40 protein may decrease the number of Th40 cells by a factor of at least 10, at least 100, at least 1,000, at least 10,000. In a preferred embodiment, the level of Th40 cells is may be reduced so that Th40 cells comprise no more than about 20%, about 25%, about 30%, about 35%, or about 40% of the total T-cell population.

Peptides and methods hereof are suitable for use in cell culture as well as for treating a patient. As used herein the term patient refers to any animal in need of such treatment. The animal can be a human or a non-human animal. A preferred animal to treat is a mammal. A peptide can be administered or applied per se, or as pharmaceutical compositions. A peptide hereof, or a pharmaceutical composition thereof, can be administered to a patient by a variety of routes, including, but limited to, by injection (e.g., intravenous, intramuscular, subcutaneous, intrathecal, intraperitoneal), by inhalation, by oral (e.g., in a pill, tablet, capsule, powder, syrup, solution, suspension, thin film, dispersion or emulsion.), transdermal, transmucosal, pulmonary, buccal, intranasal, sublingual, intracerebral, intravaginal rectal or topical administration or by any other convenient method known to those of skill in the art. The amount of a peptide hereof and/or a pharmaceutical composition thereof that will be effective can be determined by standard clinical techniques known in the art. Such an amount is dependent on, among other factors, the patient being treated, including, but not limited to the weight, age, and condition of the patient, the intended effect of the compound, the manner of administration and the judgment of the prescribing physician or veterinarian. Also, in this context, it should be noted that in treating a patient exhibiting a disorder of interest, a therapeutically effective amount of an agent or agents such as these is administered. A therapeutically effective dose refers to that amount of the compound that results in amelioration of one or more symptoms or a prolongation of survival in a patient.

A peptide hereof, or a pharmaceutical composition thereof, can be administered alone or in combination with one or more other pharmaceutical agents, including other compounds of the present disclosure. The peptides used and utilized in the methods may be administered to the subject in a pharmaceutically acceptable carrier, adjuvant, or vehicle. Select examples of pharmaceutically acceptable carriers, adjuvants, and vehicles, which are well-known in the art disclosed in Remington: The Science and Practice of Pharmacy, 21 st Ed., Hendrickson, R., et ah, Eds., Lippincott Williams & Wilkins, Baltimore, Md. (2006). The selection of a pharmaceutically acceptable carrier, adjuvant, or vehicle will depend on a variety of factors including but not limited to, the route of administration, dosage levels, the age, weight and/or condition of the subject, etc. The specific pharmaceutical composition depends on the desired mode of

administration, as is well known to the skilled artisan.

Because the inventors have discovered that Th40 cells are intimately involved in the development of autoimmune diseases and diabetes mellitus, the peptides and methods disclosed herein can be used to affect inflammation and other symptoms resulting from such diseases. Thus, one embodiment hereof is a method to treat diabetes mellitus in a patient in need of such treatment, the method comprising administering to a patient a peptide that interacts with the CD40 protein, thereby reducing diabetes mellitus. In one embodiment the peptide interacts with the CD40 protein in such a manner as to affect the interaction of CD40 and CD 154, thereby reducing diabetes mellitus. In a preferred embodiment, interaction of the peptide with the CD40 protein reduces the number of Th40 cells in a patient to a level equal to that observed in subjects that do not have diabetes mellitus. The present developments may be suitable for treating any patient having an autoimmune disease and/or diabetes mellitus, the development of which is dependent on Th40 cells. More specifically, peptides hereof may be suitable for reducing the level of Th40 cells in such patients. In a preferred embodiment, a peptide hereof reduces the level of Th40 cells in a patient suffering from a diabetes mellitus to no more than about 25% of the total T-cell population.

The inventors have also shown that, surprisingly, peptides hereof can be used to reverse the disease process in individuals already showing signs of diabetes mellitus. Thus, one aspect of the present subject matter is a method to reverse diabetes mellitus comprising administering to a patient diagnosed as having diabetes mellitus, a peptide hereof. In one embodiment, the peptide comprises an amino acid sequence selected from SEQ ID NOs:7-15, 21-25, 41-48, and 50-52, so long as the peptide can down-regulate inflammation. As used herein the phrase to reverse diabetes mellitus means to reduce diabetic symptoms to a level comparable to the observably lower level of symptoms and measurements may be more common in a non-diabetic subject. The subject may be required to continue the therapy indefinitely to continue to achieve the desired results from the therapy. In other instances, it may be possible for the subject to be titrated off of the therapy, yet have the positive effects and changes induced by the therapy carry-forward. This may be dependent on a number of factors, including but not limited to the disease state when initially presented, the progression of the disease state, and other factors as may be determined by a physician or veterinarian as the case may be.

As has been described, peptides of the present invention selectively interact with and/or affect CD40 expressing cells. Consequently, peptides of the present subject matter may be used to identify Th40 cells. Thus, one embodiment hereof is a method to detect Th40-dependent diabetes mellitus, said method comprising contacting a T-cell population with a peptide hereof. In a preferred embodiment, the peptide is labeled with a detectable marker, such as, for example, luciferase or alkaline phosphatase. Such detection can be performed using assay techniques known to those skilled in the art. In general, an assay for detecting Th40 cells using a peptide hereof comprises (a) obtaining a sample of cells; (b) contacting a peptide hereof with said cells under condition suitable to allow binding of the peptide to Th40 cells, if present; (c) washing said cells using conditions that disrupt non-specific interactions, and that remove unbound peptide; and (d) detecting peptide bound to cells. Detection of bound peptide can be achieved directly or indirectly. For example, direct detection can be achieved using a peptide labeled using a detectable marker, as disclosed herein. Following the wash step listed above, the cells are then simply screened for the presence of detectable marker. The presence of detectable marker in the cell sample indicates the presence of Th40 cells, and thus Th40-dependent diabetes. Alternatively, indirect detection involves the use of a second molecule, such as an antibody, that binds to the peptide. In an indirect detection assay, following the wash step listed above, a detection molecule that binds the peptide is added to the cell sample. The detection molecule is labeled with a detectable marker. After washing away unbound detection molecule, the cells are screened for the presence of detectable marker. The presence of detectable marker in the cell sample indicates the presence of Th40 cells. It should be understood that the assays described herein are meant as examples of useful assays, and other assay techniques can be employed. Suitable assay techniques are known to those skilled in the art, and are also disclosed in, for example, Molecular Cloning: A Laboratory Manual, Sambrook, L, Fritsch, E.F., and Maniatis, T, Cold Spring Harbor Laboratory Press; 2nd Edition (December 1989). All references cited herein are incorporated herein in their entirety.

The peptide utilized and described in the above-mentioned detection method may also include modifications to the peptide such as stable isotope labeled peptides or fluorescent peptide modifications/FRET pairs that may allow the peptide to be utilized in several detection methods and applications. For example, stable isotope labeled peptides, such as: Arg(13C6, 15N4), lie (13C6, 15N), Leu (13C6, 15N), Lys (13C6, 15N2), and/or Val(13C5, 15N) may be utilized.

Moreover, fluorescent peptide modifications/FRET pairs, such as: 1-Pyrenemethylamine HCL, 5- FAM (N-Terminal), 5-FAM-Ahx (N-terminal), Abz (N-Terminal), Abz/DNP, Abz/Tyr (3-N02), DABCYL, DABCYL/Glu(EDANS)-NH2, Dansyl (N-Terminal), Dansyl-Ahx (N-Terminal), EDANS/DABCYL, FITC (N-Terminal), FITC-Ahx (N-Terminal), Glu(EDANS)-NH2, MCA (N- Terminal), MCA/DNP, Quenched fluorescent peptide, Tyr (3-N02), TMR, and/or AMC may be utilized. Thus, the peptides hereof may be modified in such a way whether with stable isotope peptides or fluorescent peptide modifications/FRET pairs that may allow the peptides of SEQ ID NOs:7-15, 21-25, 41-48, and 50-52, or others disclosed herein, to be used for assays or other useful procedures.

The assay technology described above can also be used to identify other molecules that affect the interaction of a CD40 protein with a CD 154 protein. Examples of such molecules include, but are not limited to, proteins, peptides and small molecules. For example, assays can be designed that test the ability of molecules to compete with a peptide of the present developments for binding to a Th40 cell. For instance, a peptide labeled with a detectable marker, can be mixed with a test molecule and a population of cells known to contain Th40 cells, under conditions that allow binding of the peptide to the Th40 cells. Following an appropriate incubation period, the cells are washed to remove unbound peptide, and the cells screened for the presence of detectable marker. Alternatively, the labeled peptide could be bound to Th40 cells first, and after a wash step to remove unbound peptide, the test molecule could be added to the cells containing bound peptide. Following an incubating period and a wash step to remove unbound molecule, or released peptide, the cells are screened for the presence of detectable marker. In either case, absence of the detectable marker in the cell sample indicates the test molecule is able to compete with the peptide for binding to the Th40 cells, while presence of the detectable marker would indicate the test molecule does not inhibit binding of the peptide to Th40 cells. Inhibition of binding need not be 100%, as such assay would also be useful for identifying molecules that partially inhibit binding of the peptide to Th40 cells. It is understood by those skilled in the art that such assays would involve the use of positive controls (e.g., unlabeled peptide) and negative controls (e.g., a protein/molecule that is known not to bind to Th40 cells).

Because increased levels of Th40 cells may be associated with the development of autoimmune disease, the present developments can be used to identify patients at risk for developing autoimmune disease and autoimmune related diabetes mellitus. Thus, one

embodiment of the present developments is a method to identify a patient at risk for developing autoimmune related diabetes mellitus. In one embodiment, patients at risk for developing diabetes mellitus are identified by obtaining a sample from a patient to be tested, contacting the T-cell portion of said sample with a peptide hereof, and determining the level of Th40 cells present in the sample, wherein a level of Th40 cells greater than about 25% of the total T-cell population indicates the patient is at risk for developing autoimmune disease or diabetes mellitus. In one embodiment, the peptide comprises an amino acid sequence selected from SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15, so long as the peptide binds to the CD40 protein. In one

embodiment, the peptide is an amino acid sequence selected from SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15. In another implementation, the amino acid sequence is selected from one of SEQ ID NOs:7-15, 21-25, 41-48, and 50-52. In one implementation, the peptide is labeled with a suitable detectable marker such as, for example, luciferase or alkaline phosphatase. The present developments also comprise kits useful for practicing the methods disclosed herein, the kit comprising a peptide that interacts with a CD40 protein in such a manner as to modulate diabetes mellitus. In one embodiment, the peptide comprises an amino acid sequence selected from SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO: l l, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15, so long as the peptide can down- regulate diabetes mellitus. In one embodiment, the peptide is an amino acid sequence selected from SEQ ID NO: 7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, and SEQ ID NO: 15. . In one embodiment, the peptides is an amino acid sequence selected from SEQ ID NOs:7-15, 21-25, 41-48, and 50-52. Another embodiment is a kit for determining the level of Th40 cells, the kit comprising a peptide that interacts with a CD40 protein, and methods for detecting CD40-bound peptide. Kits can also contain associated reagents and components, such as, but not limited to, buffers, labels, containers, inserts, tubing, vials, syringes, and the like. Kits may also contain peptides that have stable isotope labeled amino acids or fluorescent peptide modifications/FRET pairs, as described elsewhere herein.

Another embodiment of the present developments includes a method to modulate and/or reduce interleukin 17 (IL-17) in a cell or a subject comprising administering a peptide selected from SEQ ID NOs 7-15, 21-26, 41-48, and 50-52 in an amount sufficient to reduce or inhibit interleukin 17 (IL-17) signaling, wherein, the IL-17 signaling is associated with a condition selected from the group comprising type I diabetes, multiple sclerosis, systemic lupus erythematosa, rheumatoid arthritis, Crohn’s disease, inflammatory bowel disease, chronic obstructive pulmonary disease, asthma, atherosclerosis, vasculitis, hypertension, thyroiditis, primary biliary cirrhosis, Paget’s disease, Addison’s disease, acute respiratory distress syndrome, acute lung injury, and/or aseptic chronic inflammation, more generally.

Another embodiment of the present developments includes a method to modulate and/or reduce interleukin 17 (IL-17) in a cell or a subject comprising administering a peptide selected from SEQ ID NOs 7-15, 21-26, 41-48, and 50-52 in an amount sufficient to reduce or inhibit interleukin 17 (IL-17) signaling, wherein, the IL-17 is associated with diabetes mellitus.

One aspect of the developments hereof includes an implementation wherein the peptides of SEQ ID NOs 7-15, 21-26, 41-48, and 50-52 may be provided as a sterile solution in phosphate buffered saline (PBS). In this implementation, the pH is approximately 5.5. The following examples are provided for the purpose of illustration and are not intended to limit the scope of the present invention.

Examples

Example 1

This Example demonstrates the effect of various peptide fragments of CD 154 on

CD4/CD8 ratios and the development of diabetes in NOD mice.

Peptides were designed based on the amino acid sequence of mouse CD 154 protein (SEQ ID NO: 1) in the SwissPro database. The peptides (8-mer (SEQ ID NO: 22; SEQ ID NO: 23), 10- mer (SEQ ID NO:41), 13-mer (SEQ ID NO:42), 15-mer (SEQ ID NO: 24), 24-mer (SEQ ID NO:26), scrambled (SEQ ID NO: 40), and RGD (arginylglycylaspartic acid) were then ordered from New England Peptide. The RGD peptide is a 15-amino acid sequence from the CD 154 sequence that does not include the CD40 binding motif. The lyophilized peptides were suspended in sterile PBS at 1 mg/ml. 25 ug (lmg/kg) of a particular peptide was then injected into the tail vein of 6-week old NOD mice. Control mice received 100 ul of sterile PBS. This is well before the onset of diabetes (and atherosclerosis), but after damage to pancreatic islets has begun.

Weekly after the initial injection, another 25ug of peptide (or 100 ul of PBS in the case of the Control mice) was injected into the tail vein. At 10 weeks of age, mice were monitored for diabetes, as indicated by a blood glucose level greater than 250 mg/dL for three consecutive days. The results of this study are shown in Figure 1. During this time, blood was also taken from the tail vein, or by sub-mandibular venal puncture, and the level of CD4+ and CD8+ cells determined by flow cytometry using antibodies for CD4 protein and CD8 protein. The results of this analysis are shown in Figure 2 A.

Pancreata were excised and examined by histology for cellular infiltrates and assigned scores based on observable, measurable, and quantifiable data: 0 = no infiltrate; 1 = one pole infiltrate; 2 = peri-insulitis, bi-polar-infiltrates; 3 = 75% infiltrate and 4 = full infiltration. The results of this analysis are shown in Figure 2B.

The results demonstrate that treatment with a peptide unrelated to the CD 154 protein did not reduce the development of diabetes in NOD mice. In contrast, treatment of mice with a 15- mer peptide derived from the CD154 protein prevented the onset of diabetes. Further, the 13-mer peptides derived from the CD 154 protein had significant effects on the development of diabetes. In addition, the data demonstrates that the 15-mer peptide did not result in compromise of the immune system, as determined by the CD4/CD8 ratio.

Example 2

This Example demonstrates the effect of the 15-mer peptide on hyperglycemia in newly diabetic NOD mice.

Six mice that had received the 6-mer peptide in Example 1, and that had subsequently developed diabetes, were injected intravenously with 100 ug of the 15-mer peptide. These mice were then given weekly injections of the 15-mer peptide into their tail veins, and their blood glucose levels monitored twice-weekly. The 15-mer peptide was administered for a total of ten weeks, after which the treatment was stopped. The results of this study are shown in Figure 3.

This study demonstrates that injection of the 15-mer peptide into already diabetic mice can reverse hyperglycemia. It also demonstrates that cessation of the treatment results in return of hyperglycemia within 7 weeks.

Example 3

This study demonstrates the ability of the 15-mer peptide to bind to Th40 cell and B cells.

Total lymphocytes were isolated from 9 week old NOD mice. The lymphocytes were incubated with anti-CD4, anti-CD8, and an FITC-labeled 15-mer peptide, and then analyzed by flow cytometry. Cells were gated for CD4 (both CD4hi and CD41o populations were included) and CD40 versus the 15-mer peptide. The results of this analysis are shown in Figure 4.

B cells were isolated from the spleens of NOD mice. Sorted MHC-II+ cells were purified from total lymphocytes. Cells were stained with FITC-labeled 15 mer peptide, anti-CD40, and B cell markers CD19 and CD21. MHC-II+ cells were gated for CD19+ and CD21+ and then 15-mer peptide versus CD40 antibody was measured. The results of this study are shown in Figure 5.

This study shows that a substantial majority, 90% of CD40+ T-cells, also bind the 15-mer peptide, thereby demonstrating that the 15-mer peptide is highly specific for CD40+ cells. It also shows that while 90% of B cells were CD40 positive, only 8% of B cells bound the 15-mer peptide.

Example 4

This example demonstrates the level of CD40 positive cells in the blood of type-I diabetic subjects and non-diabetic (control) subjects. 1 ml of whole blood was obtained from each individual and incubated with biotin- conjugated, 15-mer peptide. The cells were then exposed to horseradish peroxidase (HRP)-avidin, washed and the absorbance at 405 nm determined using a spectrophotometer. The results of this study are shown in Figure 6.

This study demonstrates that blood cells from patients having type-I diabetes had higher 15-mer peptide binding activity than cells from non-diabetic controls.

Example 5

This example demonstrates the level of insulin granulation observed in the pancreas of NOD mice treated with either the 15-mer peptide or a peptide from ovalbumin.

At the onset of diabetes, six NOD mice were injected with 100 ug/ml of the 15-mer peptide (SEQ ID NO:24), resulting in the reversal of hyperglycemia in 80% of the recipients. Six weeks after reversal of hyperglycemia, mice were sacrificed and the pancreas removed for analysis. The pancreas was fixed, sectioned and then stained using an aldehyde/fuschsin stain that allows detection of insulin granules. Granulation of the tissue was scored as follows: 4=completely granulated; 3=75% of islet granulated; 2= 50% of islet granulated, and peri-insulitis; 1=25% of islet granulated; 0= no insulin granules detected. The results of this analysis are shown in Figure 7.

This analysis demonstrates that the 15-mer peptide preserved insulin granules in the majority of the mice, and was significantly improved in peptide-reversed diabetic mice compared to diabetic mice that received an irrelevant peptide.

Example 6

This example demonstrates the effect of mutations in the 15-mer peptide on its ability to prevent the onset of diabetes. Fig. 8 provides results related to this Example 6.

Peptide were designed and produced as described in Example 1. Variant peptides were produced so that in each variant, a glycine was substituted for an amino acid corresponding to an amino acid in positions 1-9 of SEQ ID NO: 24, as follows:

Gly-1 G-L-Q-W-A-K-K-G-Y-Y-T-M-K-S-N (SEQ ID NO:28)

Gly-2 V-G-Q-W-A-K-K-G-Y-Y-T-M-K-S-N (SEQ ID NO:29)

Gly-3 V-L-G-W-A-K-K-G-Y-Y-T-M-K-S-N (SEQ ID NO: 30)

Gly-4 V-L-Q-G-A-K-K-G-Y-Y-T-M-K-S-N (SEQ ID NO: 31)

Gly-5 V-L-Q-W-G-K-K-G-Y-Y-T-M-K-S-N (SEQ ID NO:32) Gly-6 V-L-Q-W-A-G-K-G-Y-Y-T-M-K-S-N (SEQ ID NO:33)

Gly-7 V-L-Q-W-A-K-G-G-Y-Y-T-M-K-S-N (SEQ ID NO:34)

Gly-9 V-L-Q-W-A-K-K-G-G-Y-T-M-K-S-N (SEQ ID NO:35)

Gly-10 V-L-Q-W-A-K-K-G-Y-G-T-M-K-S-N (SEQ ID NO:36)

Gly-11 V-L-Q-W-A-K-K-G-Y-Y-G-M-K-S-N (SEQ ID NO:37)

Gly-12 V-L-Q-W-A-K-K-G-Y-Y-T-G-K-S-N (SEQ ID NO:38)

NOD mice were placed in groups of 10, and the mice in each group injected IV weekly with 25 ug of either wild-type (WT; Legend) peptide or a variant peptide (in PBS, ph 7.2) listed above. The development of diabetes was monitored by measuring blood glucose levels on a weekly basis. Mice were considered“diabetic” when blood glucose was 250 mg/dl or greater for 2 consecutive readings. Injections began at 6 weeks of age = pre-diabetes.

This example demonstrates that substitution of a glycine at any of positions 1-7, or 9-12, reduces the ability of the 15-mer peptide to inhibit the development of diabetes. It also shows that such mutations do not completely abolish the ability of the mutated 15-mer peptide to inhibit the development of diabetes.

Example 7

This example demonstrates that the same elevation of Th40 cell levels in the ApoE deficient mouse model of atherosclerosis is also notably elevated in human Type 1 Diabetes (T1D).

The peripheral blood was measured was measured for total count of CD3+CD4+CD40+ cell numbers in NOD, NOR (non-obese diabetic resistant), and BALB/c (control) mice as in FIG. 9. This was compared to the percentage of Th40 cells in peripheral blood in human subjects for control, diabetic/new onset, and long-term diabetic populations as in FIG. 10.

Example 8

Dog blood was obtained from a known diabetic dog. Peripheral blood mononuclear cells were isolated. Cells were stained with CD3, CD4 and CD40 and with the peptide of SEQ ID NO: 46. These results are shown in Fig. 11.

Example 9

Five diabetic dogs including 4 new onset and 1 long - term, (diagnosis greater than 3 years) were recruited into a test case study. The dogs were enrolled at Colorado State University

Veterinary Hospital (CSU-VH), Fort Collins, CO for the study. Each dog was assessed by a Board-Certified Veterinarian. Blood samples were collected from each dog and analyzed by chemistry panels to determine general health, as well as assessed by complete physical examination. The dogs were each confirmed diabetic, blood glucose ranging from 300 - 600 mg/dl but were generally in good health otherwise. One dog was diagnosed with hypertension, and two dogs had eye problems, partial blindness and the long-term dog was completely blind from diabetes.

Breeds were varied and sizes ranged from 8 to 27 kilos. Four of the dogs had been diagnosed for 3 months or less, 1 dog was diagnosed for 3.5 years. All dogs were receiving daily insulin treatments. The route of administration was IV infusion of SEQ ID NO. 13 over 30 minutes at 2 mg/kg on Days 0, 4, 8 and then weekly thereafter for a total of 8 weeks. The dogs were treated with the dog-sequence (SEQ ID NO: 13 - VLRWAPKGYYTISSN); acetate salt form; brought up in sterile PBS for injection; injection was done as a slow infusion over 30 minutes. All five dogs experienced positive outcomes while on treatment: reduction of blood and urine glucose, elimination of ketonuria in all dogs and in 2 of the dog’s urine glucose was eliminated. The dog diagnosed for 3.5 years experienced an improvement of time-in-range (percentage of a 24-hour period that blood glucose ranged between 80 and 160) from 10% to 80% for 5 consecutive days in the 5th week and achieved this for the last 3 test weeks. Typical responses in all five dogs was that 3 - 5 days post treatment improvements in blood glucose occurred. For example, blood glucose would move from 400 to 120, remain stable for 2 days but return to elevated levels. In all the treated dogs, glucose spikes were at lower levels, 300 mg/dl as a high, compared to > 500 mg/dl prior to treatment. In fact, prior to treatment, none of the dogs recorded blood glucose levels less than 350. During treatment dogs were at 200 mg/dl or lower the majority of the time and in many cases were at 150 mg/dl or less. There were no negative impacts on dogs WBC and RBC and all chemistry panel numbers were normal, as before.

Prior to treatment blood samples were analyzed for blood glucose levels and Th40 cell percentages in peripheral blood. These pre-treatment Th40 cell levels and blood glucose levels can be seen in FIG. 15, 16, and 17. In FIG. 15, controls are blood samples from test beagles used by CSU-VH and were all confirmed non-diabetic (i.e. not diabetic). Control samples demonstrate tht Th40 cells in“normal” dogs are at modestly low levels (<25%), as has been reported for mice and humans. On day 0, prior to the first treatment, blood was collected from leg vein and analyzed for Th40 cell percentages. Prior to treatment, all diabetic dogs demonstrated Th40 cells in peripheral blood at 50% or higher as in FIG. 15. Samples were analyzed for Th40 cells by flow cytometry. Furthermore, isolated blood was processed to obtain lymphocytes. Lymphocytes were stained with antibodies to recognize CD4 and CD40, which are defmitionally Th40 cells. The percentage of CD4+CD40+ as a subset of total CD4+ cells was determined and graphed. By day 6, following the third treatment, all dogs showed reduction in Th40 cell levels, by day 16 (5 th treatment) all dogs had Th40 cell percentages in the normal range. Dogs were treated through day 28. On day 35, one week after the last treatment, Th40 cell levels remained at or within a normal range. Treatment did not result in ablation, but only reduction in Th40 cell numbers. Three of the five dog subjects were re-examined two months later, Th40 cell levels remained reduced, but were beginning to return to higher numbers. None of the dogs experienced any adverse effects. Blood chemistry panels showed normal levels on most parameters, each improved from blood samples prior to treatment. Two examples of blood panels and results are provided in FIG. 19 and FIG. 20.

Blood glucose measurements were taken on diabetic dogs prior to, during, and post-peptide treatment. Data collected by a blood analysis and by continuous glucose monitoring were obtained. The continuous glucose monitoring readings were obtained periodically and the readings displayed are daily averages taken over a 2-week period. Male dog - subject 5 and female dog - subject 3 both demonstrated consistent high glucose readings prior to treatment, see FIG. 16 for results of male dog - subject 5; see FIG. 17 for results of female dog - subject 3. During the treatment period of 28 days, blood samples were taken and glucose monitoring data was recorded. The results are plotted and shown in FIG. 15. For this graph, daily averages were taken after day 6 and through day 28, which was the final treatment. Post-treatment blood glucose levels were monitored for up to 2 months.

FIG. 12 provides a graph of the blood glucose levels on four of the five dogs pre-treatment and post-treatment. Longer treatment may result in further glucose control. Using a multi column t- test, each dog experienced a significant p = 0.0042 blood glucose drop. Overall, all treatment reduced blood glucose significantly, p = 0.0040.

FIG. 13 provides a graph of glycosylated fructosamine serum level values of dog subjects, pre treatment and post-treatment. Glycosylated fructosamine is a measure of systemice inflammation. In human T1D, AIC is measured. Dog subject data for FIG. 13 were treated as described above. Prior to dosing with peptide, peripheral blood chemistry panels were performed and collected. Fructosamine levels were included. When systemic inflammation occurs, fructosamine levels are higher. After 8 weeks of treatment, fructosamine levels were measured again and compared to original numbers. The white square is pre-treatment value and black circle is the corresponding value post-treatment. Levels of fructosamine were reduced in each dog.

FIG. 14a, FIG. 14b, FIG. 14c, FIG. 14d, and FIG. 14e provide graphs of c-peptide

measurements during treatment for dog subjects 1-5. C-peptide is a measure of potential beta cell recovery. When beta cell islets produce insulin, c-peptide levels increase. Here, plasma was sampled pre- and post-treatment. Plasma was analyzed for c-peptide using commercially available test kits. This ELISA, enzyme-linked immune-sorbent assay, showed that in all cases c-peptide levels increased after treatment. This indicates that treatment affected and restored normal beta cell activity, proper production of insulin. FIG. 14a, FIG. 14b, FIG. 14c, FIG. 14d, and FIG. 14e provide the results of these c-peptide measurement studies.

Example 10

The percentage of Th40 cells in peripheral blood was studied in mouse, human and dog subjects. The results are shown in FIG. 18. Thus, FIG. 18 provides a graph of control and diabetic mouse, human and dog subjects. In this study, peripheral blood from non-diabetic dogs (n=12) was compared to diabetic dogs (n=9). All dogs were from the Colorado State University

Veterinary Teaching Hospital, where they were all examined and seen in clinic. The determination of Th40 is the same in dog, as for mouse and human: CD4+CD3+CD40+ with confirmed TCR expression.

Example 11

This example was performed in mice; however, it demonstrates that KGYY-15 (SEQ ID NO:

11) may be utilized to significantly reduce and/or reverse hyperglycemia in diabetic NOD mice. In this study, NOD mice, blood glucose between 250 - 400 mg/dl were treated with KGYY-15. Control mice were treated with MR1 an anti-CD 154 antibody or scrambled 15-mer peptide (SEQ ID NO: 17). KGYY-15 (SEQ ID NO: 11) reduced blood glucose in 57% of mice treated, the controls MR1 and scrambled 15-mer peptide had no effect. These results are provided in graph format shown in FIG. 21.

Example 12

FIG. 22 provides a graph showing the binding stability of KGYY-15 (SEQ ID NO: 11).

KGYY-15 bound to human or mouse CD4+CD40+ cells maintained in culture for 10 days.

KGYY15 was highly stable through 5 days, dropping off at 7 days, and again dropping off at 10 days. Cell death was the leading cause for binding loss. Example 13

FIG. 23 provides a graph demonstrating that SEQ ID NO: 11 regulates the inflammatory cytokine IFNy without impacting production of non-inflammatory cytokine IL-4. In this study, human T cells, isolated from peripheral blood of T1D subjects, were stimulated with autologous antigen presenting cells loaded with human islets in the absence/presence of KGYY15 (SEQ ID NO: 11) then intracellular IFNy and IL-4 were measured by flow cytometry. P<0.001 for IFNy.

Example 14

FIG. 24 provides a graph showing that different concentrations of peptide of SEQ ID NO:46 affects or influences levels of IL-2, IFNy, and IL-17A. In this study, CD3+CD4+CD40+ (Th40) splenic cells were purified from ApoE /_ with cardiovascular disease. The cells were cultured overnight in the absence/presence of varying concentrations of KGYY6 peptide. Treated cells demonstrated varying degrees of reduction in the inflammatory cytokines IL-2, IFNy and IL-17A compared to the untreated cells.

Example 15

FIG. 25 provides a graph showing the effect that treatment with SEQ ID NO: 13 had on the keytones as measured in the urine obtained from the dog test subjects described in Example 9. Keytones were measured in urine at the start of the study and approximately every 2 weeks thereafter. The dog subjects treated with SEQ ID NO: 13 demonstrated that treatment with SEQ ID NO: 13 resulted in all dog subjects reaching 0 keytone measurement by the end of the study.

From the foregoing, it is readily apparent that new and useful implementations of the methods have been herein described and illustrated which fulfill numerous desiderata in remarkably unexpected fashions. It is, of course, understood that such modifications, alterations and adaptations as may readily occur to the artisan confronted with this disclosure are intended within the spirit of this disclosure, which is limited only by the scope of the claims appended hereto.