METHOD FOR EX-VIVO EXPANSION OF REGULATORY T CELLS WITH ENHANCED SUPPRESSIVE FUNCTION FOR CLINICAL APPLICATION IN IMMUNE MEDIATED DISEASES

The invention provides methods for the ex-vivo expansion of CD4+CD25+ Tregs. The invention provides a method for producing ex vivo expanded Tregs that may be used to inhibit unwanted human immune responses against self-antigens or allergens. Additionally, the ex vivo expanded Tregs may provide treatment for inflammatory/autoimmune diseases.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 12/781,451, filed on May 17, 2010, which claims the benefit of U.S. Provisional Application No. 61/179,165, filed May 18, 2009. The entire disclosure of each application that is set forth in this Cross Reference to Related Applications section is hereby incorporated by reference.

Field of the Invention

The invention relates to methods for enhancing the suppressive function of regulatory T cells. In particular, the invention provides a method for ex-vivo expansion of regulatory T cells, which cells demonstrate enhanced suppressive characteristics.

Background of the Invention

Regulatory T cells, or Tregs, are known to be critical in maintaining a tolerance to self-antigens by suppressing the activation of the immune system. In person with inflammatory/autoimmune diseases, such as systemic lupus erythematous, multiple sclerosis, rheumatoid arthritis, asthma, ulcerative colitis, and Crohn's Disease, there is a functional defect or frequency decrease in the transcription factor Foxp3 expressing CD4+CD25+Tregs. In such autoimmune diseases, the person's immune system fails to recognize cells, or parts of cells as the person's own resulting in tissue destruction.

The therapeutic effects of administration of CD4+CD25+ Tregs has been shown in the treatment of a variety of animal disease models including, without limitation, rheumatoid arthritis, asthma, and graft-versus-host disease (“GVHD”). Use of Tregs in treatment applications is problematic because they are present as only a very small percentage, approximately 1 to 2%, of human peripheral blood mononuclear cells. Thus, methods of activating and expanding, or proliferating, Tregs ex-vivo have been developed for use in the treatment of certain diseases.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is a graph showing the frequency of CD4+Foxp3+ Tregs from the population of Example 1.

FIG. 2 is a graph showing the purity CD4+ Tregs of Example 2.

FIGS. 3A-3F are a graphs showing the Treg expansion of Example 3.

FIG. 4 is a graph showing the percentage of ex-vivo expanded cells of Example 3 that expressed Foxp3.

FIGS. 5A-5L are a graphs showing the inhibitory activity of the expanded Tregs of Example 4.

DETAILED DESCRIPTION OF THE INVENTION AND PREFERRED EMBODIMENTS

The invention provides methods for the ex-vivo expansion of CD4+CD25+ Tregs, compositions resulting from the method and methods for use of the compositions. The cells resulting from use of the method demonstrate enhanced suppressive activities compared to fresh Treg cells. The invention provides a method for producing ex vivo expanded Tregs that may be used to inhibit unwanted human immune responses against self-antigens or allergens. Additionally, the ex vivo expanded Tregs may provide treatment for inflammatory/automimmune diseases.

In one embodiment, the invention provides a methods comprising, consisting essentially of, and consisting of: a) obtaining a population of T cells from an individual; b) isolating and purifying from the T cell population a subpopulation of CD4+CD25+ Tregs; and c) expanding the Treg cells of the subpopulation, wherein the expanded Treg cells exhibit enhanced suppressive activity compared to a population of freshly purified, unexpanded Tregs from the individual.

It is a discovery of the invention that Tregs taken from a person with autoimmune/inflammatory diseases including, without limitation Systemic Lupus Erythematous (“SLE”), Multiple Sclerosis (“MS”), asthma, Rheumatoid Arthritis (“RA”), Crohn's Disease (“CD”), and Ulcerative Colitis (“UC), display enhanced suppressive activities to inhibit T cell proliferation when compared to freshly purified Tregs from the same person. It is a further discovery of the invention that the Tregs can be expanded 100 to 1,000 fold ex-vivo while having enhanced suppressive function.

In a first step of the method of the invention, a population of T cells is obtained. The T cells may be obtained from any suitable source including, without limitation, an individual's peripheral blood, thymus, lymph nodes, spleen or bone marrow. Preferably, the T cells are obtained from the peripheral blood of a person. More preferably, the T cells are obtained from an individual with an autoimmune/inflammatory diseases and most preferably from an individual with one or more of SLE, MS, asthma, RA, CD and UC whether the disease is in remission or during a period in which the disease is active. Most preferably, human CD4+CD25+ T regs are purified from whole blood units or leukpaks.

A subpopulation of CD4+CD25+ Tregs is then isolated from the T cell population using any convenient separation techniques based on Treg specific cell markers, including, without limitation, flow cytometry by any convenient method. For example, any number of kits for carrying out such isolation and purification are commercially available. The commercially available kits include, without limitation, Miltenyi Treg kit with Auotmacs, ClinMACS, and the like. Preferably, the isolation and purification is carried out until a population that is greater than 40% positive for Foxp3 and greater than 90% positive for CD4 is obtained.

The Treg subpopulation is then expanded ex-vivo using an autoantigen specific regulatory T cell stimulatory composition. Preferably, the composition antigen-specifically binds and activates the T cell receptor complex. More preferably, the expansion is carried out in the presence of effective amounts of a first and a second activator and a co-stimulator activator. By effective amount is meant an amount effective to stimulate the regulatory T cells to the degree desired.

The first activator is a TCR/CD3 activator that may be a multivalent antibody or ligand for TCR/CD3 including, without limitation, antigen non-specific activators such as an anti-CD antibody, and antigen-specific activators, such as an MHC-peptide multimer in which the peptide is an autoimmune/inflammatory disease associated peptide. Preferably, the TCR/CD3 activator is an anti-CD3 antibody.

The composition includes a second activator that is an additional suitable regulatory T cell stimulator. This component may include, without limitation an interleukin. Preferably IL-2 is used. The IL-2 typically is used in recombinant form and used in an amount of about 1,000 IU/ml.

A multivalent antibody or ligand specific for a TCR co-stimulator may be used as the co-stimulator activator. Preferably, the co-stimulator activator is CD28. The activator may be enhanced by use of one or more additives including, without limitation, rapamycin, a P13 kinase inhibitor, anit-IL6 and the like.

The TCR/CD3 activator and TCR co-stimulator activator preferably are immobilized on a three-dimensional solid surface, more preferably on a bead. Suitable beads are commercially available such as cell Expander Dynalbeads (Invitrogen). The Treg to bead ratio is preferably is 1:3. Optimal bead size will depend on a consideration of the size required to efficiently congregate the antibodies and typically is about 1 to 10 microns in diameter. The anti-CD3 and anti-CD28 antibody ratio on the surface of the bead may vary from about 1 to 20 or about 20 to 1.

The expansion is carried out to at least a 100 fold expansion, preferably to a greater than 1,000. The expansion will depend upon the stimulation and length of the culture. However, other variations may be introduced into the culture the longer the length of the culture and, thus, a culture time that permits obtaining the most cells without a substantial introduction of other unfavorable factors is preferred.

The invention will be clarified by consideration of the following, non-limiting examples.

EXAMPLES Example 1

Approximately 5 to 10 peripheral blood samples of 50 cc each were collected from 9 persons with SLE, 7 persons with RA, 7 persons with asthma, 10 persons with MS, and 7 persons with CD as well as from 11 persons with no autoimmune disease.

CD4+ Tregs were isolated and purified from the peripheral blood mononuclear cells (“PBMCs”) of the samples. The PBMCs were isolated from blood samples by density gradient centrifiguation with Ficoll Hypaque (Amersham). The CD4+CD25+ Tregs were purified from PBMCs by autoMACS using the human CD4+CD25+ regulatory T cell isolation kit (Miltenyi Biotec, Auburn, Calif.) according to the manufacturer's instructions. Briefly, CD4+ T cells were first negatively isolated from PBMCs by depleting non CD4 cells with the mixture of monoclonal antibodies against human CD8, CD14, CD16, CD19, CD36, CD56, CD123, TCRy/8 and CD235a. Human CD4+CD25+ Tregs were then positively isolated with anti-human CD25 antibody-conjugated microbeads from the enriched CD4+ T cell population. The frequency of Foxp3-expressing CD4+ Tregs was assessed using intracellular Foxp3 staining and analyzed by flow cytometry. The results are displayed in FIG. 1 below in which the percentage of CD4+Fox3+ Tregs in the CD4+ pool population from each person is shown.

Example 2

Approximately 5 to 10 peripheral blood samples of 50 cc each were collected from 9 persons with SLE, 7 persons with RA, 7 persons with asthma, 8 persons with MS, and 7 persons with CD as well as from 25 persons with no autoimmune disease.

CD4+ Tregs were purified from the PBMCs of the samples. Purification was carried out using as set forth in Example 1. The purity of purified CD4+ Tregs at day 0 was assessed using intracellular Foxp3 staining and analyzed by flow cytometry and the results are displayed on the graph in FIG. 2. Approximately 40 to 75% of the purified cells expressed Foxp3.

Example 3

Approximately 5 to 10 peripheral blood samples of 50 cc each were collected from persons with SLE (FIG. 3A), RA (FIG. 3C), asthma (FIG. 3B), MS (FIG. 3D), CD (FIG. 3E), and UC (FIG. 3F). CD4+ Tregs were purified from the PBMCs of the samples. Purification was carried out using as set forth in Example 1.

The CD4+Fox3+ Tregs were cultured in X-VIVO 15™ media (Cambrex Bio-Whitaker, East Rutherford, N.J.) supplemented with 10% pooled human AB serum (Cambrex) in the presence of 1000 IU/Ml of human rIL-2 (Proleukin). In addition, anti-CD3/ant-CD28 coated beads (Dynal, Oslo, Norway) were added at a 1:3 cell to bead ratio. After 3 weeks of culture at 37° C. in an incubator, the expanded Tregs reached over 100 to 1,000 fold expansion, or 1 billion cells, as shown in the graphs in FIG. 3. In FIG. 4 is a graph depicting that an average of 40 to 50% of the ex-vivo expanded cells expressed Foxp3 by intracellular staining at 2 weeks.

Example 4

The Tregs of Example 3 were evaluated at week 2 with standardized suppressive assays as follows. For anti-CD3 stimulated cultures, allogenic CD4+CD25+ human T cells (5×104 cells/well) from the same donor were used as responder cells, 1 mg/ml anti-CD3 (OKT3), and allogenic human dendritic cells (from one donor, 5×103 cells/well) and serially diluted expanded Tregs were put in 96-well plates in triplicate. For alloantigen stimulated cultures, CD4+CD25+ human Treg celss (5×104 cells/well) from one donor, 5×103 human dendritic cells from another donor, and serially diluted expanded Tregs were plated in triplicate in 96-well plates.

The results, shown in the graphs of FIGS. 5A and 5B (SLE), FIGS. 5C and 5D (asthma), FIGS. 5E and 5F (RA), FIGS. 5G and 5H (MS), FIGS. 5I and 5J (CD), and FIGS. 5K and 5L (UC), demonstrate that the ex vivo expanded human Tregs had potent inhibitory activity to inhibit T cell proliferation in in vitro functional assays. Additionally, the Tregs' inhibitory activity was enhanced as compared to that of freshly purified Tregs from the same persons. This suppressive activity may be used to inhibit unwanted human immune responses in autoimmune/inflammatory diseases and provide a clinical therapeutic potential for autoimmune/inflammatory diseases.

Claims

1-10. (canceled)

11. A method comprising: a) obtaining a population of T cells from an individual with asthma; b) isolating and purifying from the T cell population a subpopulation of CD4+CD25+ Tregs; and c) expanding the Treg cells of the subpopulation in the presence of effective amounts of a first and a second activator and a co-stimulator activator, wherein the expanded Treg cells exhibit enhanced suppressive activity compared to a population of freshly purified, unexpanded Tregs from the individual.

12. The method of claim 11, wherein the expansion is between about 100 to 1,000 fold.

13. The method of claim 11, wherein the expansion was carried out for a period of about 3 weeks.

14. The method of claim 11, wherein the first activator is an anti-CD3 antibody.

15. The method of claim 11, wherein the second activator is IL-2.

16. The method of claim 11, wherein the co-stimulator activator is CD28.

17. The method of claim 11, wherein the first activator is an anti-CD3 antibody, the second activator is IL-2 and the co-stimulator activator is CD28.

18. The method of claim 11, wherein the isolating and purifying are carried out until the subpopulation of CD4+CD25+, Tregs is greater than 40% positive for Foxp3 and greater than 90% for CD4.

19. A method of treating an individual with immune-mediated disease, the method comprising: a) obtaining a population of T cells from an individual with asthma; b) isolating and purifying from the T cell population a subpopulation of CD4+CD25+ Tregs; c) expanding the Treg cells of the subpopulation in the presence of effective amounts of a first and a second activator and a co-stimulator; and d) administering a portion of the CD4+CD25+ regulatory T cells to a human being treated for the immune-mediated disease.

20. The method of claim 19, wherein the first activator is an anti-CD3 antibody.

21. The method of claim 19, wherein the second activator is IL-2.

22. The method of claim 19, wherein the co-stimulator activator is CD28.

23. The method of claim 19, wherein the first activator is an anti-CD3 antibody, the second activator is IL-2 and the co-stimulator activator is CD28.

24. The method of claim 19, wherein the isolating and purifying are carried out until the subpopulation of CD4+CD25, Tregs is greater than 40% positive for Foxp3 and greater than 90% for CD4.

Patent History
Publication number: 20170298322
Type: Application
Filed: Apr 8, 2016
Publication Date: Oct 19, 2017
Inventors: Tinghua Cao (Malvern, PA), Li Li (Downington, PA)
Application Number: 15/094,467
Classifications
International Classification: C12N 5/0783 (20100101); A61K 35/17 (20060101); A61K 35/12 (20060101);