Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Diabetologia ; 67(1): 27-41, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37782353

ABSTRACT

AIMS/HYPOTHESIS: We hypothesised that islet beta cell antigen presentation in the gut along with a tolerising cytokine would lead to antigen-specific tolerance in type 1 diabetes. We evaluated this in a parallel open-label Phase 1b study using oral AG019, food-grade Lactococcus lactis bacteria genetically modified to express human proinsulin and human IL-10, as a monotherapy and in a parallel, randomised, double-blind Phase 2a study using AG019 in combination with teplizumab. METHODS: Adults (18-42 years) and adolescents (12-17 years) with type 1 diabetes diagnosed within 150 days were enrolled, with documented evidence of at least one autoantibody and a stimulated peak C-peptide level >0.2 nmol/l. Participants were allocated to interventions using interactive response technology. We treated 42 people aged 12-42 years with recent-onset type 1 diabetes, 24 with Phase 1b monotherapy (open-label) and 18 with Phase 2a combination therapy. In the Phase 2a study, after treatment of the first two open-label participants, all people involved were blinded to group assignment, except for the Data Safety Monitoring Board members and the unblinded statistician. The primary endpoint was safety and tolerability based on the incidence of treatment-emergent adverse events, collected up to 6 months post treatment initiation. The secondary endpoints were pharmacokinetics, based on AG019 detection in blood and faeces, and pharmacodynamic activity. Metabolic and immune endpoints included stimulated C-peptide levels during a mixed meal tolerance test, HbA1c levels, insulin use, and antigen-specific CD4+ and CD8+ T cell responses using an activation-induced marker assay and pooled tetramers, respectively. RESULTS: Data from 24 Phase 1b participants and 18 Phase 2a participants were analysed. No serious adverse events were reported and none of the participants discontinued AG019 due to treatment-emergent adverse events. No systemic exposure to AG019 bacteria, proinsulin or human IL-10 was demonstrated. In AG019 monotherapy-treated adults, metabolic variables were stabilised up to 6 months (C-peptide, insulin use) or 12 months (HbA1c) post treatment initiation. In participants treated with AG019/teplizumab combination therapy, all measured metabolic variables stabilised or improved up to 12 months and CD8+ T cells with a partially exhausted phenotype were significantly increased at 6 months. Circulating preproinsulin-specific CD4+ and CD8+ T cells were detected before and after treatment, with a reduction in the frequency of preproinsulin-specific CD8+ T cells after treatment with monotherapy or combination therapy. CONCLUSIONS/INTERPRETATION: Oral delivery of AG019 was well tolerated and safe as monotherapy and in combination with teplizumab. AG019 was not shown to interfere with the safety profile of teplizumab and may have additional biological effects, including changes in preproinsulin-specific T cells. These preliminary data support continuing studies with this agent alone and in combination with teplizumab or other systemic immunotherapies in type 1 diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT03751007, EudraCT 2017-002871-24 FUNDING: This study was funded by Precigen ActoBio.


Subject(s)
Diabetes Mellitus, Type 1 , Adult , Adolescent , Humans , Interleukin-10 , C-Peptide , CD8-Positive T-Lymphocytes/metabolism , Proinsulin , Double-Blind Method
2.
Front Immunol ; 11: 1103, 2020.
Article in English | MEDLINE | ID: mdl-32582188

ABSTRACT

A combination treatment (CT) of proinsulin and IL-10 orally delivered via genetically modified Lactococcus lactis bacteria combined with low-dose anti-CD3 (aCD3) therapy successfully restores glucose homeostasis in newly diagnosed non-obese diabetic (NOD) mice. Tolerance is accompanied by the accumulation of Foxp3+ regulatory T cells (Tregs) in the pancreas. To test the potential of this therapy outside the window of acute diabetes diagnosis, we substituted autoimmune diabetic mice, with disease duration varying between 4 and 53 days, with syngeneic islets at the time of therapy initiation. Untreated islet recipients consistently showed disease recurrence after 8.2 ± 0.7 days, while 32% of aCD3-treated and 48% of CT-treated mice remained normoglycemic until 6 weeks after therapy initiation (P < 0.001 vs. untreated controls for both treatments, P < 0.05 CT vs. aCD3 therapy). However, mice that were diabetic for more than 2 weeks before treatment initiation were less efficient at maintaining normoglycemia than those treated within 2 weeks of diabetes diagnosis, particularly in the aCD3-treated group. The complete elimination of endogenous beta cell mass with alloxan at the time of diabetes diagnosis pointed toward the significance of continuous feeding of the islet antigen proinsulin at the time of aCD3 therapy for treatment success. The CT providing proinsulin protected 69% of mice, compared to 33% when an irrelevant antigen (ovalbumin) was combined with aCD3 therapy, or to 27% with aCD3 therapy alone. Sustained tolerance was accompanied with a reduction of IGRP+CD8+ autoreactive T cells and an increase in insulin-reactive (InsB12-20 or InsB13-2) Foxp3+CD4+ Tregs, with a specific accumulation of Foxp3+ Tregs around the insulin-containing islet grafts after CT with proinsulin. The combination of proinsulin and IL-10 via oral Lactococcus lactis with low-dose aCD3 therapy can restore tolerance to beta cells in autoimmune diabetic mice, also when therapy is started outside the window of acute diabetes diagnosis, providing persistence of insulin-containing islets or prolonged beta cell function.


Subject(s)
CD3 Complex/antagonists & inhibitors , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/drug effects , Interleukin-10/administration & dosage , Proinsulin/administration & dosage , Animals , Diabetes Mellitus, Experimental/immunology , Genetic Vectors , Humans , Lactococcus lactis , Mice , Mice, Inbred NOD , Self Tolerance/drug effects , Self Tolerance/immunology
3.
Diabetes ; 63(8): 2876-87, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24677716

ABSTRACT

Growing insight into the pathogenesis of type 1 diabetes (T1D) and numerous studies in preclinical models highlight the potential of antigen-specific approaches to restore tolerance efficiently and safely. Oral administration of protein antigens is a preferred method for tolerance induction, but degradation during gastrointestinal passage can impede such protein-based therapies, reducing their efficacy and making them cost-ineffective. To overcome these limitations, we generated a tolerogenic bacterial delivery technology based on live Lactococcus lactis (LL) bacteria for controlled secretion of the T1D autoantigen GAD65370-575 and the anti-inflammatory cytokine interleukin-10 in the gut. In combination with short-course low-dose anti-CD3, this treatment stabilized insulitis, preserved functional ß-cell mass, and restored normoglycemia in recent-onset NOD mice, even when hyperglycemia was severe at diagnosis. Combination therapy did not eliminate pathogenic effector T cells, but increased the presence of functional CD4(+)Foxp3(+)CD25(+) regulatory T cells. These preclinical data indicate a great therapeutic potential of orally administered autoantigen-secreting LL for tolerance induction in T1D.


Subject(s)
Autoantigens/pharmacology , Diabetes Mellitus/immunology , Glutamate Decarboxylase/pharmacology , Interleukin-10/metabolism , Peptide Fragments/pharmacology , Administration, Oral , Aging , Animals , Autoantigens/administration & dosage , Autoantigens/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Glutamate Decarboxylase/administration & dosage , Interleukin-10/genetics , Lactococcus lactis , Mice , Mice, Inbred NOD , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , T-Lymphocytes, Regulatory/drug effects
4.
Cell Host Microbe ; 11(4): 387-96, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22520466

ABSTRACT

The intestinal microbiota has been linked to inflammatory bowel diseases (IBD), and oral treatment with specific bacteria can ameliorate IBD. One bacterial mixture, VSL#3, containing Lactobacillus, Bifidobacterium, and Streptococcus, was clinically shown to reduce inflammation in IBD patients and normalize intestinal levels of IP-10, a lymphocyte-recruiting chemokine, in a murine colitis model. We identified Lactobacillus paracasei prtP-encoded lactocepin as a protease that selectively degrades secreted, cell-associated, and tissue-distributed IP-10, resulting in significantly reduced lymphocyte recruitment after intraperitoneal injection in an ileitis model. A human Lactobacillus casei isolate was also found to encode lactocepin and degrade IP-10. L. casei feeding studies in a murine colitis model (T cell transferred Rag2(-/-) mice) revealed that a prtP-disruption mutant was significantly less potent in reducing IP-10 levels, T cell infiltration and inflammation in cecal tissue compared to the isogenic wild-type strain. Thus, lactocepin-based therapies may be effective treatments for chemokine-mediated diseases like IBD.


Subject(s)
Chemokine CXCL10/metabolism , Colitis/metabolism , Lacticaseibacillus casei/enzymology , Lactobacillus/enzymology , Serine Endopeptidases/metabolism , Amino Acid Sequence , Animals , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Colitis/immunology , Colitis/microbiology , Colitis/therapy , Disease Models, Animal , Female , Humans , Lactobacillus/immunology , Lacticaseibacillus casei/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Probiotics/therapeutic use , Protein Transport , Proteolysis , Serine Endopeptidases/immunology
5.
J Clin Invest ; 122(5): 1717-25, 2012 May.
Article in English | MEDLINE | ID: mdl-22484814

ABSTRACT

Current interventions for arresting autoimmune diabetes have yet to strike the balance between sufficient efficacy, minimal side effects, and lack of generalized immunosuppression. Introduction of antigen via the gut represents an appealing method for induction of antigen-specific tolerance. Here, we developed a strategy for tolerance restoration using mucosal delivery in mice of biologically contained Lactococcus lactis genetically modified to secrete the whole proinsulin autoantigen along with the immunomodulatory cytokine IL-10. We show that combination therapy with low-dose systemic anti-CD3 stably reverted diabetes in NOD mice and increased frequencies of local Tregs, which not only accumulated in the pancreatic islets, but also suppressed immune response in an autoantigen-specific way. Cured mice remained responsive to disease-unrelated antigens, which argues against excessive immunosuppression. Application of this therapeutic tool achieved gut mucosal delivery of a diabetes-relevant autoantigen and a biologically active immunomodulatory cytokine, IL-10, and, when combined with a low dose of systemic anti-CD3, was well tolerated and induced autoantigen-specific long-term tolerance, allowing reversal of established autoimmune diabetes. Therefore, we believe this method could be an effective treatment strategy for type 1 diabetes in humans.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Immune Tolerance , Lactococcus lactis/genetics , Animals , Autoantigens/biosynthesis , Autoantigens/genetics , CD3 Complex/immunology , Cell Count , Cell Proliferation , Combined Modality Therapy , Diabetes Mellitus, Type 1/immunology , Humans , Hypoglycemic Agents/therapeutic use , Immunologic Factors/therapeutic use , Immunosuppression Therapy , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-10/metabolism , Intestinal Mucosa , Lactococcus lactis/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Proinsulin/biosynthesis , Proinsulin/genetics , Proinsulin/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/physiology
6.
Front Biosci (Landmark Ed) ; 14(13): 4825-35, 2009 06 01.
Article in English | MEDLINE | ID: mdl-19482589

ABSTRACT

The taxonomically diverse lactic acid bacteria (LAB) are unified by their capability to produce lactic acid from carbohydrates by fermentation. The LAB Lactococcus (L.) lactis has been characterized into great detail and is increasingly used as a production host for heterologous proteins. L. lactis is a non-pathogenic and non-colonizing LAB species and can be efficiently engineered to produce proteins of viral, bacterial or eukaryotic origin, both intra- or extracellularly. Importantly, orally formulated L. lactis strains (ActoBiotics), engineered to synthesize and secrete therapeutic peptides and proteins in the gastrointestinal tract, are already in advanced stages of preclinical and clinical development. This review focuses on the genetic engineering of LAB in general and L. lactis in specific to secrete high-quality, correctly processed, bioactive molecules derived from a eukaryotic background. The therapeutic applications of these genetically modified strains are discussed, as well as the need for a sound environmental containment strategy, and a detailed review is presented on Lactococcus strains engineered to produce specific antigens, antibodies, cytokines and trefoil factors, with special regards to immunomodulation.


Subject(s)
Immunologic Factors/biosynthesis , Immunologic Factors/genetics , Lactococcus/genetics , Lactococcus/immunology , Animals , Antibodies/genetics , Antibody Formation , Bacterial Vaccines/biosynthesis , Bacterial Vaccines/genetics , Containment of Biohazards , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/therapy , Lactococcus lactis/genetics , Lactococcus lactis/immunology , Probiotics/therapeutic use , Protein Engineering , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Th1 Cells/immunology , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL