WO2012062697A1 - Combination therapy for type 1 diabetes - Google Patents

Combination therapy for type 1 diabetes Download PDF

Info

Publication number
WO2012062697A1
WO2012062697A1 PCT/EP2011/069531 EP2011069531W WO2012062697A1 WO 2012062697 A1 WO2012062697 A1 WO 2012062697A1 EP 2011069531 W EP2011069531 W EP 2011069531W WO 2012062697 A1 WO2012062697 A1 WO 2012062697A1
Authority
WO
WIPO (PCT)
Prior art keywords
gad65
vaccine
diabetes
mice
cells
Prior art date
Application number
PCT/EP2011/069531
Other languages
French (fr)
Inventor
Matthias Von Herrath
Philippe Pagni
Original Assignee
Novartis Ag
La Jolla Institute For Allergy & Immunology
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag, La Jolla Institute For Allergy & Immunology filed Critical Novartis Ag
Publication of WO2012062697A1 publication Critical patent/WO2012062697A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/245IL-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination

Definitions

  • the present invention relates to a combination treatment for Type 1 diabetes. More specifically, the invention relates to a method of treatment for Type 1 diabetes based on administration of an antibody and a vaccine to a patient in need thereof.
  • Type 1 diabetes is a devastating autoimmune condition in which self-reactive T cells attack pancreatic ⁇ cells that secrete insulin.
  • Interleukin (IL)-i p is a proinflammatory cytokine produced by several cell types, including pancreatic ⁇ cells when exposed to high glucose concentrations. Since 11-1 ⁇ can exert direct cytotoxic effects on ⁇ cells, one therapeutic avenue for treating T1 D would be to neutralize this cytokine temporarily. This could be combined with antigen specific approaches to maintain tolerance to ⁇ cell antigens. For example, recent studies in our laboratory showed increased efficacy using combination therapies with anti-CD3 and islet autoantigens such as insulin or glutamic acid decarboxylase of 65 kd (GAD65)-expressing plasmids.
  • GID65 glutamic acid decarboxylase of 65 kd
  • RIP-GP transgenic mice that turned diabetic upon LCMV infection were treated weekly with antibodies and at day 0 » 4 and 11 with GAD65 DNA.
  • Results show that anti-lL1 p monotherapy could only protect 25% ⁇ 1/4 ⁇ of mice from T1 D, but that combination-therapy with neutralizing IL- ⁇ ⁇ antibodies and GAD65 DNA could reverse disease in 7/9 animals (78%). Consistently with earlier studies in the lab using DNA vaccinations alone in recent-onset diabetic mice (Bresson et al.
  • the invention provides a method of treatment of a human subject suffering from or at risk of developing type 1 diabetes comprising administering to the subject an effective amount of a) an IL1 ⁇ inhibitory molecule and b) a tolerizing or suppressive vaccine against a type I diabetes autoantigen.
  • the invention provides a) an ⁇ L 1 ⁇ inhibitory molecule and b) a tolerizing or suppressive vaccine against a type I diabetes autoantigen for use in the treatment of a human subject suffering from or at risk of developing type 1 diabetes.
  • the vaccine is directed against, tolerizes against or suppresses an immune response against one or more autoantigens selected from the group consisting of: GAD65, GAD67, insulin, proinsulin, HSP60, IA-2, IA-2 ⁇ , carboxy-peptidase H, islet-cell expressed gangliosides, ZnT8 (a zinc transporter in the membrane of islet ⁇ cell secretory granules); and IGRP (islet-specific glucose-6-phosphate catalytic subunit related protein).
  • autoantigens selected from the group consisting of: GAD65, GAD67, insulin, proinsulin, HSP60, IA-2, IA-2 ⁇ , carboxy-peptidase H, islet-cell expressed gangliosides, ZnT8 (a zinc transporter in the membrane of islet ⁇ cell secretory granules); and IGRP (islet-specific glucose-6-phosphate catalytic subunit related protein).
  • the vaccine is directed against, tolerizes against or suppresses an immune response against one or more peptide fragments of the one or more autoantigens.
  • the vaccine is a protein vaccine.
  • the vaccine is a DNA vaccine.
  • the 111 ⁇ inhibitory molecule is an antibody directed against IL1 ⁇ .
  • the antibody is a monoclonal antibody selected from the group consisting of Canakinumab, Xoma 052, LY2189102.
  • the antibody is Canakinumab and the vaccine tolerizes against or suppresses an immune response against GAD65 or peptide fragments thereof.
  • FIG. 1 Mean blood glucose values (BGVs) in mg/dL in all groups when treatment was started, (B) Individual BGVs for mice whose value was over 400mg/dL at treatment start and (C) Diabetes incidence in recent-onset diabetic mice.
  • BGVs blood glucose values
  • Individual BGVs for mice whose value was over 400mg/dL at treatment start and
  • Diabetes incidence in recent-onset diabetic mice Female B6 mice expressing glycoprotein (GP) from lymphocytic choriomeningitis virus (LCMV) under the rat insulin promoter (RIP) were infected with 10 4 PFU LCMV Armstrong intraperitoneal ⁇ .
  • GP glycoprotein
  • LCMV lymphocytic choriomeningitis virus
  • RIP rat insulin promoter
  • Antibodies were administered intraperitoneally (10ug per gram body weight) once a week for 5 weeks and GAD65 DNA vaccine was given intramuscularly (100ug per leg) at day 0, 4 and 11.
  • mice were sacrificed and spleen, pancreatic lymph nodes (PLNs) as well as pancreata were collected and processed for analysis.
  • Figure 2 Frequencies of IFN- ⁇ and TNF-a producing GP33-specific CD8+ effector T cells in spleen and PLNs of treated mice. After 5-week treatment, spleens and PLNs were mashed onto 70um cell strainers to obtain single-cell suspensions. Cells were then plated in round bottom 96- well plates and left unstimulated, or stimulated for 4 to 5 hours with 5ug/mL (CD8 T-cell specific) GP33 peptide.
  • FIG. 3 Proportions of Foxp3-expressing Tregs in treated mice.
  • Spleen and PLN cells from treated mice were mashed against 70um cell strainers, resuspended, and prepared for further flow cytometry staining for CD4+CD25+Foxp3+ regulatory T cells (Tregs) using FoxpS staining kit (eBiosicence) according to the manufacturer's instructions.
  • Data were acquired on a LSRII cytometer (BD) and analyzed with FlowJo software (Tree Star). Data are expressed as % of gated CD4+ cells. Results from 2-3 pooled experiments with 2-6 mice per group are shown.
  • Figure 4 Preservation of insulin-producing ⁇ cells in the pancreas of treated mice. Shown is a picture representative of the size of islets counted and plotted in the lower panel. Magnification: 20X. Frozen sections (6um) were cut from pancreata of mice treated with the different regimens and left to dry overnight. Next day, sections were fixed with acetone and blocked with Tris- buffer/NaCl solution plus protease inhibitors and goat serum. Sections were stained with guinea pig anti-insuiin primary antibody (Dako), followed by staining with goat anti-guinea pig alkaline phosphatase (AP) antibody (Sigma). AP detection was performed using Vector Blue AP III (Vector) and sections were mounted with aqueous mounting solution (Lemer Lab.). Pictures were taken with a Nikon 80i fluorescence microscope.
  • Dako guinea pig anti-insuiin primary antibody
  • AP goat anti-guinea pig alkaline phosphatase
  • FIG. 5 CD4+ ,CD8+ and CD11b+ cell infiltration in mice treated with GAD65 DNA with anti- isotype or anti-IL- ⁇ antibodies.
  • Pancreas sections from treated mice were stained for insulin as described in Figure 4.
  • different stains for CD4, CD8 and CD11 b were performed using biotinylated primary antibodies (1/100, BD), followed by the addition of streptavidin coupled-horseradish peroxidase (1/1000, Vector) and developed using Nova Red Peroxidase substrate kit (Vector).
  • the extent of CD4+, CD8+ or CD11 b+ cells was counted for 16 to 111 islets from 2 to 5 mice per group.
  • ⁇ nti-IL1 ⁇ treatment can reverse the course of recent-onset T1D to a modest extent when used alone, but to a much greater extent when used in combination with GAD65 DNA vaccine.
  • Recent-onset diabetic RIP-LCMV mice were treated with the following regimens: monotherapy with anti-IL- ⁇ ⁇ antibody alone, anti-isotype antibody alone, combination therapy with anti-IL- ⁇ ⁇ antibody and GAD65-expressing DNA plasmid, or anti-isotype antibody and GAD65 DNA.
  • Figure 1A shows a similar average BGV when treatment was started in all groups, suggesting that randomization of mice was performed without any bias.
  • Figure 1 B demonstrates that mice whose BGV was over 400mg/di_ when treatment was initiated, could not revert to normoglycemia, regardless of the tested treatment.
  • GAD65 DNA vaccine therapy preserves insulin-producing pancreatic ⁇ cells, regadless of the antibody used in combination.
  • anti-IL-1 p/GAD65-treated mice presented in average more medium (6.71 vs 3) and small (8.57 vs 2.5) islets compared to anti-IL-1 ⁇ -treated mice. Altogether, these findings are consistent with the generally improved diabetes outcome observed in GAD65-treated mice with or without IL- ⁇ blockade.

Abstract

The present invention relates to a method of treatment of a human subject suffering from or at risk of developing type 1 diabetes comprising administering to the subject an effective amount of a) an IL1β inhibitory molecule and b) a tolerizing vaccine against a type I diabetes autoantigen.

Description

Combination therapy for Type 1 diabetes
The present invention relates to a combination treatment for Type 1 diabetes. More specifically, the invention relates to a method of treatment for Type 1 diabetes based on administration of an antibody and a vaccine to a patient in need thereof.
Type 1 diabetes (T1 D) is a devastating autoimmune condition in which self-reactive T cells attack pancreatic β cells that secrete insulin. Interleukin (IL)-i p is a proinflammatory cytokine produced by several cell types, including pancreatic β cells when exposed to high glucose concentrations. Since 11-1 β can exert direct cytotoxic effects on β cells, one therapeutic avenue for treating T1 D would be to neutralize this cytokine temporarily. This could be combined with antigen specific approaches to maintain tolerance to β cell antigens. For example, recent studies in our laboratory showed increased efficacy using combination therapies with anti-CD3 and islet autoantigens such as insulin or glutamic acid decarboxylase of 65 kd (GAD65)-expressing plasmids.
The inventors investigated whether treatment of recent-onset diabetic mice with anti-IL1 β antibody alone (provided by Novartis) or in combination with a GAD85 DNA vaccine could reverse disease development. During 5 weeks, RIP-GP transgenic mice that turned diabetic upon LCMV infection (RIP-LCMV model for type 1 diabetes) were treated weekly with antibodies and at day 0» 4 and 11 with GAD65 DNA. Results show that anti-lL1 p monotherapy could only protect 25% {1/4} of mice from T1 D, but that combination-therapy with neutralizing IL-Ι β antibodies and GAD65 DNA could reverse disease in 7/9 animals (78%). Consistently with earlier studies in the lab using DNA vaccinations alone in recent-onset diabetic mice (Bresson et al. JCI, 2006) GAD65 treatment alone achieved disease reversal in 3/6 animals (50%). Reversion of hyperglycemia was associated with preservation of insulin-producing islets and correlated in the spleen with decreased production of IFN-γ by GP33 peptide-specific ('autoreactive') CD8 effector T cells, which are required for disease in this diabetes model because they recognize the transgenically expressed GP protein on β cells. Reversion to normoglycemia was also reflected by increases in frequencies of Foxp3-expressing CD4 T regulatory cells in pancreatic lymph nodes, but no statistically significant difference was observed between GAD65 treatment with or without anti-IL-1 β antibodies. In these two groups, the extent of CD4+ and CD8+ cell infiltration was also found to be similar. However, mice treated with anti-IL1 β Ab + GAD65 DNA vaccine seemed to display less severe infiltration by CD1 1 b+ cells (presumably macrophages) compared to isotype Ab/GAD65-treated mice.
The present findings in the RIP-LCMV mouse model provide evidence that IL-Ι β blockade alone can only reverse diabetes to a certain extent, but that combination therapy with GAD65 DNA vaccines permits a marked improvement of clinical and immunological aspects of the disease. These data hold promise for the treatment of new-onset T1 D patients with combination therapies involving blockade of IL-1.
In one aspect the invention provides a method of treatment of a human subject suffering from or at risk of developing type 1 diabetes comprising administering to the subject an effective amount of a) an IL1 β inhibitory molecule and b) a tolerizing or suppressive vaccine against a type I diabetes autoantigen.
In a further aspect the invention provides a) an ΙL 1 β inhibitory molecule and b) a tolerizing or suppressive vaccine against a type I diabetes autoantigen for use in the treatment of a human subject suffering from or at risk of developing type 1 diabetes.
In one embodiment the vaccine is directed against, tolerizes against or suppresses an immune response against one or more autoantigens selected from the group consisting of: GAD65, GAD67, insulin, proinsulin, HSP60, IA-2, IA-2 β, carboxy-peptidase H, islet-cell expressed gangliosides, ZnT8 (a zinc transporter in the membrane of islet β cell secretory granules); and IGRP (islet-specific glucose-6-phosphate catalytic subunit related protein).
In one embodiment the vaccine is directed against, tolerizes against or suppresses an immune response against one or more peptide fragments of the one or more autoantigens.
In one embodiment the vaccine is a protein vaccine.
In one embodiment the vaccine is a DNA vaccine. In one preferred embodiment the 111 β inhibitory molecule is an antibody directed against IL1 β. In one embodiment the antibody is a monoclonal antibody selected from the group consisting of Canakinumab, Xoma 052, LY2189102.
In one specific embodiment, the antibody is Canakinumab and the vaccine tolerizes against or suppresses an immune response against GAD65 or peptide fragments thereof. Brief description of the Figures
Figure 1: (A) Mean blood glucose values (BGVs) in mg/dL in all groups when treatment was started, (B) Individual BGVs for mice whose value was over 400mg/dL at treatment start and (C) Diabetes incidence in recent-onset diabetic mice. Female B6 mice expressing glycoprotein (GP) from lymphocytic choriomeningitis virus (LCMV) under the rat insulin promoter (RIP) were infected with 104 PFU LCMV Armstrong intraperitoneal^. From day 7 post-infection, BGV was assessed for all the mice and upon BGVs above 250mg/dL, mice were included in one of the following treatment groups; anti-isotype antibody (black line, n=4), anti-IL-1 p antibody (red line, n=4), anti-isotype antibody plus GAD65 DNA vaccine (green line, n=6) or anti-IL-1 β antibody plus GAD65 DNA vaccine (blue line, n=9). Antibodies were administered intraperitoneally (10ug per gram body weight) once a week for 5 weeks and GAD65 DNA vaccine was given intramuscularly (100ug per leg) at day 0, 4 and 11. At the end of the treatment, mice were sacrificed and spleen, pancreatic lymph nodes (PLNs) as well as pancreata were collected and processed for analysis. Figure 2: Frequencies of IFN-γ and TNF-a producing GP33-specific CD8+ effector T cells in spleen and PLNs of treated mice. After 5-week treatment, spleens and PLNs were mashed onto 70um cell strainers to obtain single-cell suspensions. Cells were then plated in round bottom 96- well plates and left unstimulated, or stimulated for 4 to 5 hours with 5ug/mL (CD8 T-cell specific) GP33 peptide. PMA (50ng/mL) and ionomycin (1 ug/mL) were used as positive controls. In all conditions, brefeldin A (10ug/mL) was used to block protein secretion. Cells were then stained in surface with fluorochrome-coupled CD8 and CD4 antibodies (Biolegend), fixed and permeabilized using FoxpS staining kit (eBioscience), and later stained with fluorochrome- coupled IFN-γ and TNF-a antibodies (Biolegend). Data were acquired on a LSRII cytometer (BD) and analyzed with FlowJo software (Tree Star). Data are expressed as % of gated CD8+ cells. Results from 2-3 pooled experiments with 2-6 mice per group are shown.
Figure 3: Proportions of Foxp3-expressing Tregs in treated mice. Spleen and PLN cells from treated mice were mashed against 70um cell strainers, resuspended, and prepared for further flow cytometry staining for CD4+CD25+Foxp3+ regulatory T cells (Tregs) using FoxpS staining kit (eBiosicence) according to the manufacturer's instructions. Data were acquired on a LSRII cytometer (BD) and analyzed with FlowJo software (Tree Star). Data are expressed as % of gated CD4+ cells. Results from 2-3 pooled experiments with 2-6 mice per group are shown.
Figure 4: Preservation of insulin-producing β cells in the pancreas of treated mice. Shown is a picture representative of the size of islets counted and plotted in the lower panel. Magnification: 20X. Frozen sections (6um) were cut from pancreata of mice treated with the different regimens and left to dry overnight. Next day, sections were fixed with acetone and blocked with Tris- buffer/NaCl solution plus protease inhibitors and goat serum. Sections were stained with guinea pig anti-insuiin primary antibody (Dako), followed by staining with goat anti-guinea pig alkaline phosphatase (AP) antibody (Sigma). AP detection was performed using Vector Blue AP III (Vector) and sections were mounted with aqueous mounting solution (Lemer Lab.). Pictures were taken with a Nikon 80i fluorescence microscope.
Figure 5:. CD4+ ,CD8+ and CD11b+ cell infiltration in mice treated with GAD65 DNA with anti- isotype or anti-IL-Ιβ antibodies. Pancreas sections from treated mice were stained for insulin as described in Figure 4. In parallel, different stains for CD4, CD8 and CD11 b were performed using biotinylated primary antibodies (1/100, BD), followed by the addition of streptavidin coupled-horseradish peroxidase (1/1000, Vector) and developed using Nova Red Peroxidase substrate kit (Vector). The extent of CD4+, CD8+ or CD11 b+ cells was counted for 16 to 111 islets from 2 to 5 mice per group. Classification was performed as follows: No infiltration; mild infiltration when cells were found around the insulin-producing islet; heavy infiltration when cells were found within the insulin-producing islet. A 2-way ANOVA test concluded that differences the two groups were statistically significant with regard to CD1 1 b+ cell infiltration.
Experimental section
1. Αnti-IL1β treatment can reverse the course of recent-onset T1D to a modest extent when used alone, but to a much greater extent when used in combination with GAD65 DNA vaccine.
Recent-onset diabetic RIP-LCMV mice were treated with the following regimens: monotherapy with anti-IL-Ι β antibody alone, anti-isotype antibody alone, combination therapy with anti-IL-Ι β antibody and GAD65-expressing DNA plasmid, or anti-isotype antibody and GAD65 DNA. Figure 1A shows a similar average BGV when treatment was started in all groups, suggesting that randomization of mice was performed without any bias. Figure 1 B demonstrates that mice whose BGV was over 400mg/di_ when treatment was initiated, could not revert to normoglycemia, regardless of the tested treatment. However, when mice were treated with a starting BGV between 250 and 400mg/dL, we could observe that treatments with anti-IL-Ι β Ab provided a slightly protective effect in 25% (1/4) of mice while anti-isotype control Ab treatment protected none of the animals (0/4) (Figure 1 C). Moreover, GAD65 DNA vaccine monotherapy resulted in a 50% (3/6) reversion to normoglycemia upon 5 weeks of treatment, Interestingly, when treating diabetic animals with anti-IL-1 β Ab in combination with GAD65, 78% (7/9) of animals reverted to normoglycemia (Figure 1 C). These data suggest that an additive effect is provided by the concomitant action of IL-1 β neutralization and GAD65 DNA administrations.
2, GAD65 DNA vaccine given with either antibody leads decreased frequencies of GP33-specific IFN-y producing CD8 cells in the spleen, Cells from spleen and PLNs were isolated and restimuiated with LCMV GP33 peptide in order to evaluate CDS T-cell specific cytokine production. In comparison to isotype control antibody treatment, we noticed a tendency towards diminished IFN-γ secreting cells in the spleen of anti- IL-1 β-treated animals (Figure 2), The co-administration of GAD85 DNA together with either antibody correlated with decreased production of IFN-γ (and TNF-ct, to a lesser extent) by GP33- specific CD8 effector T cells in the spleen, but not PLNs.
3, The improved diabetes incidence upon combination therapy is not reflected by higher proportions of Foxp3-expressing Tregs.
We then assessed whether the increased success upon IL-1 p blockade in conjunction with GAD65 DNA could be explained by a greater presence of Foxp3-expressing Tregs in spleen and PLNs. We found that compared to single-antibody treatment, the addition of GAD65 DNA vaccination significantly increased the frequencies of CD4+CD25+Foxp3+ Tregs locally in PLNs. Nonetheless, no statistically significant difference could be seen between GAD treatment with or without anti-IL-1 β antibodies (Figure 3). Taken together, we can conclude that in comparison to GAD65 treatment alone, the improved benefit provided by IL-1 β blockade cannot be attributed to an increase in Foxp3-expressing Tregs.
4, GAD65 DNA vaccine therapy preserves insulin-producing pancreatic β cells, regadless of the antibody used in combination.
We further investigated whether the protection, or lack thereof, seen in the different groups of mice would correlate with the number of insulin-producing β cells. To do so, 6-um pancreata sections were cut and stained for insulin. We found that anti-isotype or anti-IL-1 β antibody- treated animals did not present large islets, as defined in Figure 4 (upper panel), while anti- isotype/GAD65- and anti-IL-1 p/GAD65-treated mice presented averages of 3.34 and 3.71 islets per section, respectively. Moreover, anti-isotype/GAD65-treated mice had in average higher numbers of medium (4 vs 1 ) and small (7.67 vs 1 ) islets compared to anti-isotype Ab-treated mice. Likewise, anti-IL-1 p/GAD65-treated mice presented in average more medium (6.71 vs 3) and small (8.57 vs 2.5) islets compared to anti-IL-1 β-treated mice. Altogether, these findings are consistent with the generally improved diabetes outcome observed in GAD65-treated mice with or without IL-Ιβ blockade.
5, IL'lfi blockade upon diabetes onset seems to participate in the reduction of pancreatic infiltration by CD11b+ macrophages, but not by CD4+ nor CD8+ T cells, 11-1 β can be produced by β cells, especially in conditions of hyperglycemia, but also by macrophages which can count among the different inflammatory cell infiltrates in diabetic mice and patients. Thus, we assessed by immunohistochemistry the extent of pancreatic infiltration by CD4+ and CD8+ T cells, but also CD11 b+ macrophages in anti-isotype/GAD65- and anti-IL- 1 p/GAD65-treated mice, In these two groups, we found that overall CD4+ and CD8+ cell infiltration was similar. However, mice treated with anti-IL1 p/GAD65 DNA vaccine seemed to display less severe infiltration by CD11 b+ macrophages compared to isotype Ab/GAD65-treated mice.
In sum, the results suggest that the protective effect of anti-IL-1 β therapy alone in recent-onset mouse model of diabetes is modest, which is consistent with the minor effect IL-1 R deficiency had on diabetes development in the RIP-LCMV model (Barrai et al. J. Autoimmunity 2006). When performed alone, the induction of an antigen-specific tolerance using GAD65 DNA vaccination resulted in 50% reversal from hyperglycemia, which is in line with previous studies (Bresson et al., JCI 2006). However, combining the neutralization of IL-Ι β with GAD65 DNA administration for a period of 5 weeks significantly improved the reversal of recent-onset diabetes in transgenic RIP-GP B6 mice. Compared to either antibody treatment, the addition of GAD65 DNA vaccine resulted in (i) diminished IFN-y production by GP33-specific CDS T cells in the spleen, (ii) higher proportions of Foxp3-expressing Tregs in PLNs and (iii) higher numbers of preserved insulin-producing β cells. !Ι_-1 β blockade together with GAD65 DNA vaccination resulted in a reduced infiltration of CD1 1 b+ macrophages, but not CD4 and CDS T cells, in the pancreas of treated mice. These data confer a potential explanation for the improved clinical outcome observed upon this combination therapy (78% reversion) in comparison to separate administrations of either ΙΙ_-1 β blockade (25% reversion) or GAD65 DNA vaccination (50% reversion). The perspective that the efficacy of an anti-inflammatory drug such as anti-IL-1 β can be improved by combination with antigen specific approaches known to have no to minimal systemic side effects is clinically promising.

Claims

Claims
1 ) A method of treatment of a human subject suffering from or at risk of developing type 1 diabetes comprising administering to the subject an effective amount of a) an I L1 β inhibitory molecule and b) a toierizing vaccine against a type I diabetes autoantigen. 2) The method according to claim 1 , wherein the toierizing vaccine is directed against one or more autoantigens or fragments thereof, selected from the group consisting of; GAD65, GAD67, insulin, proinsulin, HSP60, IA-2, IA-2 β, carboxy-peptidase H, ZnT8, IGRP, and islet-cell expressed gangliosides.
3} The method according to claim 1 or 2, wherein the toierizing vaccine is a protein vaccine. 4) The method according to any of the preceding claims, wherein the toierizing vaccine is a DNA vaccine.
5) The method according to any of the preceding claims, wherein the 1L1 β inhibitory molecule is a monoclonal antibody directed against IL1 β.
6) The method according to any of the preceding claims, wherein the antibody is selected from the group consisting of Canakinumab, Xoma 052, LY2189102.
PCT/EP2011/069531 2010-11-08 2011-11-07 Combination therapy for type 1 diabetes WO2012062697A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US41109110P 2010-11-08 2010-11-08
US61/411,091 2010-11-08

Publications (1)

Publication Number Publication Date
WO2012062697A1 true WO2012062697A1 (en) 2012-05-18

Family

ID=44999750

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/069531 WO2012062697A1 (en) 2010-11-08 2011-11-07 Combination therapy for type 1 diabetes

Country Status (1)

Country Link
WO (1) WO2012062697A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017189483A1 (en) * 2016-04-25 2017-11-02 The Johns Hopkins University Znt8 assays for drug development and pharmaceutical compositions
WO2018083111A1 (en) * 2016-11-01 2018-05-11 Novo Nordisk A/S Tolerogenic dna vaccine
JP2019532659A (en) * 2016-11-01 2019-11-14 ノヴォ ノルディスク アー/エス Tolerance-inducing DNA vaccine
EP3760224A3 (en) * 2014-06-04 2021-03-31 Diamyd Medical AB Novel combinations for antigen based therapy
US11040943B2 (en) 2016-11-28 2021-06-22 Lb Pharmaceuticals Inc. Psychotropic agents and uses thereof
US11279745B2 (en) 2019-04-26 2022-03-22 Novo Nordisk A/S Tolerogenic DNA vaccine
US11892457B2 (en) 2017-07-12 2024-02-06 The Johns Hopkins University Proteoliposome-based ZnT8 self-antigen for type 1 diabetes diagnosis

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010028273A1 (en) * 2008-09-05 2010-03-11 Xoma Technology Ltd. Methods for improvement of beta cell function

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010028273A1 (en) * 2008-09-05 2010-03-11 Xoma Technology Ltd. Methods for improvement of beta cell function

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BARRAL ET AL., J. AUTOIMMUNITY, 2006
BRESSON D ET AL: "Moving towards efficient therapies in type 1 diabetes: to combine or not to combine?", AUTOIMMUNITY REVIEWS APR 2007 LNKD- PUBMED:17412305, vol. 6, no. 5, April 2007 (2007-04-01), pages 315 - 322, XP022015535, ISSN: 1568-9972 *
BRESSON DAMIEN ET AL: "Genetic-induced variations in the GAD65 T-cell repertoire governs efficacy of anti-CD3/GAD65 combination therapy in new-onset type 1 diabetes.", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY FEB 2010 LNKD- PUBMED:19690518, vol. 18, no. 2, February 2010 (2010-02-01), pages 307 - 316, XP002667930, ISSN: 1525-0024 *
BRESSON ET AL., JCI, 2006
HERRATH VON M G ET AL: "DNA VACCINATION TO TREAT AUTOIMMUNE DIABETES", ANNALS OF MEDICINE, TAYLOR & FRANCIS A B, SE, vol. 32, no. 5, 1 July 2000 (2000-07-01), pages 285 - 292, XP008056367, ISSN: 0785-3890 *
VON HERRATH MATTHIAS: "Combination therapies for type 1 diabetes: why not now?", IMMUNOTHERAPY MAY 2010 LNKD- PUBMED:20635894, vol. 2, no. 3, May 2010 (2010-05-01), pages 289 - 291, XP002667929, ISSN: 1750-7448 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL273265B2 (en) * 2014-06-04 2023-06-01 Diamyd Medical Ab Novel combinations for antigen based therapy
EP3760224A3 (en) * 2014-06-04 2021-03-31 Diamyd Medical AB Novel combinations for antigen based therapy
US11016085B2 (en) 2016-04-25 2021-05-25 The Johns Hopkins University ZNT8 assays for drug development and pharmaceutical compositions
US11841363B2 (en) 2016-04-25 2023-12-12 The Johns Hopkins University ZnT8 assays for drug development and pharmaceutical compositions
WO2017189483A1 (en) * 2016-04-25 2017-11-02 The Johns Hopkins University Znt8 assays for drug development and pharmaceutical compositions
JP2020062057A (en) * 2016-11-01 2020-04-23 ノヴォ ノルディスク アー/エス Tolerogenic dna vaccine
EP3799882A1 (en) * 2016-11-01 2021-04-07 Novo Nordisk A/S Tolerogenic dna vaccine
IL266237B (en) * 2016-11-01 2022-08-01 Novo Nordisk As Tolerogenic dna vaccine
US11466278B2 (en) 2016-11-01 2022-10-11 Novo Nordisk A/S Temperature based plasmid regulation system
JP2019532659A (en) * 2016-11-01 2019-11-14 ノヴォ ノルディスク アー/エス Tolerance-inducing DNA vaccine
WO2018083111A1 (en) * 2016-11-01 2018-05-11 Novo Nordisk A/S Tolerogenic dna vaccine
US11040943B2 (en) 2016-11-28 2021-06-22 Lb Pharmaceuticals Inc. Psychotropic agents and uses thereof
US11713295B2 (en) 2016-11-28 2023-08-01 Lb Pharmaceuticals Inc. Psychotropic agents and uses thereof
US11892457B2 (en) 2017-07-12 2024-02-06 The Johns Hopkins University Proteoliposome-based ZnT8 self-antigen for type 1 diabetes diagnosis
US11279745B2 (en) 2019-04-26 2022-03-22 Novo Nordisk A/S Tolerogenic DNA vaccine

Similar Documents

Publication Publication Date Title
Robert et al. Oral delivery of glutamic acid decarboxylase (GAD)-65 and IL10 by Lactococcus lactis reverses diabetes in recent-onset NOD mice
WO2012062697A1 (en) Combination therapy for type 1 diabetes
US11286303B2 (en) CTLA4 fusion proteins for the treatment of diabetes
US20230158109A1 (en) Use of cd83 in combination therapies
JP2007520566A (en) Anti-CD3 and antigen-specific immunotherapy for autoimmune therapy
RU2448729C2 (en) Compositions causing specific response of cytotoxic t-lymphocytes, including lymph-ablative compound and molecule which contains antigen sequences and is targeted at specialised antigen-presenting cells
JPH08508036A (en) Methods and compositions for delaying or preventing the onset of autoimmune disease
WO2011135544A2 (en) Methods and compositions for treating hepatitis with anti-cd3 immune molecule therapy
US20210177967A1 (en) Apoptotic cell-mediated induction of antigen specific regulatory t-cells for the therapy of autoimmune diseases in animals and humans
US20060165687A1 (en) Vaccine adjuvant
Puentes et al. Immune modulation and prevention of autoimmune disease by repeated sequences from parasites linked to self antigens
US20080085261A1 (en) Vaccine Adjuvant
Li et al. Mucosal tolerance induction in autoimmune myocarditis and myocardial infarction
US20070190078A1 (en) Filamentous haemagglutinin in the treatment and/or prophylaxis of immune-mediated disorders
Pritchard Novel therapeutic approaches to Guillain-Barré syndrome
AU2008238215B2 (en) Ig-pConsensus gene vaccination protects from antibody-dependent immune pathology in autoimmune disease
JP6969790B2 (en) Multipeptide composition
Kim et al. Immunotherapeutic restoration in HIV-infected individuals
Ferrera et al. New therapies in SLE
Li et al. Immunotherapy for Type 1 Diabetes–Preclinical and Clinical Trials
Singh et al. Immunomodulation in human and experimental uveitis: recent advances
WO2021222851A1 (en) Combinations of viral proteins, peptide sequences, epitopes, and methods and uses thereof
Lane et al. Immunologic Approaches to the Therapy of Patients with HIV Infection
Cava et al. New Therapies in SLE
Fulton Intestinal and systemic cytotoxic T lymphocyte and humoral immune responses to oral and parenteral reovirus infection

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11784972

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11784972

Country of ref document: EP

Kind code of ref document: A1