1
|
Yedke NG, Arthur R, Kumar P. Bacillus calmette gaurine vaccine ameliorates the neurotoxicity of quinolinic acid in rats via the modulation of antioxidant, inflammatory and apoptotic markers. J Chem Neuroanat 2023; 131:102287. [PMID: 37172828 DOI: 10.1016/j.jchemneu.2023.102287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/06/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
A mutation in the Huntingtin gene causes 'Huntington's disease, which presents as a motor and behavioral impairment. Due to the limited drug therapy for this disease, scientists are constantly searching for newer and alternative drugs that may either retard or prevent the progress of the disease. This study aims to explore the neuroprotective potential of Bacillus Calmette Gaurine (BCG) vaccine against quinolinic acid-induced (QA) neurotoxicity in rats. QA (200 nmol/2 µl, i.s) was injected bilaterally into the rat striatum, after which a single dose of BCG (2 × 10^7, cfu) was given to the rats. Animals were assessed for behavioral parameters on the 14th and 21st days. On the 22nd day, animals were sacrificed, brains were harvested, and striatum was separated to evaluate biochemical, inflammatory, and apoptotic mediators. Histopathological studies were performed using Hematoxyline and Eosin staining to assess neuronal morphology. BCG treatment reversed motor abnormalities, reduced oxidative stress and neuroinflammatory markers, apoptotic mediators and striatal lesions induced by QA treatment. In conclusion, treat' 'ing rats with BCG vaccine (2 × 10^7, cfu) mitigated the quinolinic acid-induced Huntington's disease-like symptoms. Hence, BCG vaccine (2 ×10^7, cfu) could be used as an adjuvant in managing HD.
Collapse
Affiliation(s)
- Narhari Gangaram Yedke
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda 151001, Punjab, India; Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
2
|
Ludvigsson J. Glutamic acid decarboxylase immunotherapy for type 1 diabetes. Curr Opin Endocrinol Diabetes Obes 2022; 29:361-369. [PMID: 35776501 DOI: 10.1097/med.0000000000000748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW To describe recent development of an autoantigen (GAD) treatment towards well tolerated and efficacious precision medicine in type 1 diabetes. RECENT FINDINGS Although subcutaneous GAD-alum treatment failed to reach primary endpoint in a phase III trial, metanalyses showed a 97% probability of efficacy, and clear efficacy in patients carrying Hyman Leucoycte Antigen (HLA) DR3DQ2. Efforts have been made to improve efficacy by trying combination therapies with vitamin D + Ibuprofen resp vitamin D + Etanercept (TNF-α inhibition), without any breakthrough until the administration of GAD-alum was changed from subcutaneous to intralymphatic. With a very small dose of GAD-alum (4 μg) given into an inguinal lymph three times with 1 month interval, the efficacy in patients with HLADR3DQ2 has been impressive, with significantly better beta cell preservation than patients who got placebo in a double-blind randomized trial, and clinical efficacy with more patients in partial remission (IDAA1c < 9) and larger proportion of patients with CGM-measured blood glucose Time In Range (TIR), significantly correlated to the C-peptide values. The treatment has been easy for patients and healthcare without treatment-related risk or adverse events. SUMMARY Intralymphatic GAD-alum treatment in type 1 diabetes patients carrying HLA DR3DQ2 seems to be an attractive immune intervention.
Collapse
Affiliation(s)
- Johnny Ludvigsson
- Crown Princess Victoria Children's Hospital and Division of Pediatrics, Departmentt of Biomedical and Clinical Sciences, Linköping University, Sweden
| |
Collapse
|
3
|
Designing Personalized Antigen-Specific Immunotherapies for Autoimmune Diseases-The Case for Using Ignored Target Cell Antigen Determinants. Cells 2022; 11:cells11071081. [PMID: 35406645 PMCID: PMC8997884 DOI: 10.3390/cells11071081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
We have proposed that antigen-specific immunotherapies (ASIs) for autoimmune diseases could be enhanced by administering target cell antigen epitopes (determinants) that are immunogenic but ignored by autoreactive T cells because these determinants may have large pools of naïve cognate T cells available for priming towards regulatory responses. Here, we identified an immunogenic preproinsulin determinant (PPIL4-20) that was ignored by autoimmune responses in type 1 diabetes (T1D)-prone NOD mice. The size of the PPIL4-20-specific splenic naive T cell pool gradually increased from 2–12 weeks in age and remained stable thereafter, while that of the major target determinant insulin B-chain9-23 decreased greatly after 12 weeks in age, presumably due to recruitment into the autoimmune response. In 15–16 week old mice, insulin B-chain9-23/alum immunization induced modest-low level of splenic T cell IL-10 and IL-4 responses, little or no spreading of these responses, and boosted IFNγ responses to itself and other autoantigens. In contrast, PPIL4-20/alum treatment induced robust IL-10 and IL-4 responses, which spread to other autoantigens and increased the frequency of splenic IL-10-secreting Treg and Tr-1-like cells, without boosting IFNγ responses to ß-cell autoantigens. In newly diabetic NOD mice, PPIL4-20, but not insulin B-chain9-23 administered intraperitoneally (with alum) or intradermally (as soluble antigen) supplemented with oral GABA induced long-term disease remission. We discuss the potential of personalized ASIs that are based on an individual’s naïve autoantigen-reactive T cell pools and the use of HLA-appropriate ignored autoantigen determinants to safely enhance the efficacy of ASIs.
Collapse
|
4
|
Sun J, Shi J, Li J, Wu M, Li Y, Jia S, Ma C, Wang X, Li Z, Hu N, Hu Y. The Effect of Immunosuppressive Adjuvant Kynurenine on Type 1 Diabetes Vaccine. Front Immunol 2021; 12:681328. [PMID: 34305913 PMCID: PMC8293994 DOI: 10.3389/fimmu.2021.681328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/14/2021] [Indexed: 01/13/2023] Open
Abstract
Inducing antigen-specific tolerance is a promising treatment for preventing or reversing Type 1 diabetes (T1D). In contrast to a vaccine that induces immune responses against pathogens, a tolerogenic vaccine can suppress immunity against antigens causing diseases by administrating a mixture of self-antigens with an adjuvant that decreases the strength of antigen-specific response. Kynurenine (Kyn) is an endogenous substance that can inhibit the natural killer cell and T cell proliferation and promote the differentiation of naïve T cells into regulatory T cells (Tregs). In this study, we evaluated the efficacy of Kyn as a novel suppressive adjuvant. Kyn was co-immunized with GAD65 phage vaccine to induce Treg cells and tolerogenic responses for the prevention of T1D in NOD mouse model. Mice were subcutaneously immunized two times with 1011 Pfu (100μL,1012 Pfu/ml) GAD65 phage vaccine doses mixed with 200 μg of Kyn. Serum antibodies and cytokines were detected by ELISA and electrochemiluminescence, respectively. Flow cytometry assay was used to analyze DC and Treg. MTS was used for the analysis of spleen lymphocyte proliferation. RNA sequencing was used to investigate mRNA and miRNA expression profiles in spleen lymphocytes. Compared to GAD65 phage vaccine alone, co-immunization of Kyn and GAD65 phage vaccine resulted in the prevention of hyperglycemia in 60% of mice for at least one month. Further, Kyn enhances GAD65-specific Th2-mediated immune responses; regulates the Th1/Th2 imbalance and increases the secretion of Th2 cytokines and the number of CD4+CD25+Foxp3+T cells; suppresses DC maturation and GAD65-specific T lymphocyte proliferation. Moreover, we integrated Kyn related miRNA and mRNA expression profiles obtained from the spleen lymphocyte RNA-sequencing which was stimulated by Kyn in vitro. These data provide an important basis for understanding the mechanisms underlying Kyn as an immunosuppressive adjuvant which regulated the immune response. These findings suggest that Kyn can serve as an effective suppressive adjuvant candidate for Type 1 diabetes vaccines.
Collapse
Affiliation(s)
- Jing Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Jiandong Shi
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Jianfang Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Meini Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yanhan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Sengquan Jia
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Chunli Ma
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Kunming Medical University, Kunming, China
| | - Xinyi Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Zhiyuan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Kunming Medical University, Kunming, China
| | - Ningzhu Hu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| | - Yunzhang Hu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
| |
Collapse
|
5
|
Liu J, Ting JP, Al-Azzam S, Ding Y, Afshar S. Therapeutic Advances in Diabetes, Autoimmune, and Neurological Diseases. Int J Mol Sci 2021; 22:ijms22062805. [PMID: 33802091 PMCID: PMC8001105 DOI: 10.3390/ijms22062805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023] Open
Abstract
Since 2015, 170 small molecules, 60 antibody-based entities, 12 peptides, and 15 gene- or cell-therapies have been approved by FDA for diverse disease indications. Recent advancement in medicine is facilitated by identification of new targets and mechanisms of actions, advancement in discovery and development platforms, and the emergence of novel technologies. Early disease detection, precision intervention, and personalized treatments have revolutionized patient care in the last decade. In this review, we provide a comprehensive overview of current and emerging therapeutic modalities developed in the recent years. We focus on nine diseases in three major therapeutics areas, diabetes, autoimmune, and neurological disorders. The pathogenesis of each disease at physiological and molecular levels is discussed and recently approved drugs as well as drugs in the clinic are presented.
Collapse
Affiliation(s)
- Jinsha Liu
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Joey Paolo Ting
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Shams Al-Azzam
- Professional Scientific Services, Eurofins Lancaster Laboratories, Lancaster, PA 17605, USA;
| | - Yun Ding
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Sepideh Afshar
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
- Correspondence:
| |
Collapse
|
6
|
Boddul SV, Sharma RK, Dubnovitsky A, Raposo B, Gerstner C, Shen Y, Iyer VS, Kasza Z, Kwok WW, Winkler AR, Klareskog L, Malmström V, Bettini M, Wermeling F. In vitro and ex vitro functional characterization of human HLA-DRB1∗04 restricted T cell receptors. J Transl Autoimmun 2021; 4:100087. [PMID: 33768201 PMCID: PMC7980064 DOI: 10.1016/j.jtauto.2021.100087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Recent advances in single-cell sequencing technologies enable the generation of large-scale data sets of paired TCR sequences from patients with autoimmune disease. Methods to validate and characterize patient-derived TCR data are needed, as well as relevant model systems that can support the development of antigen-specific tolerance inducing drugs. We have generated a pipeline to allow streamlined generation of 'artificial' T cells in a robust and reasonably high throughput manner for in vitro and in vivo studies of antigen-specific and patient-derived immune responses. Hereby chimeric (mouse-human) TCR alpha and beta constructs are re-expressed in three different formats for further studies: (i) transiently in HEK cells for peptide-HLA tetramer validation experiments, (ii) stably in the TCR-negative 58 T cell line for functional readouts such as IL-2 production and NFAT-signaling, and lastly (iii) in human HLA-transgenic mice for studies of autoimmune disease and therapeutic interventions. As a proof of concept, we have used human HLA-DRB1∗04:01 restricted TCR sequences specific for a type I diabetes-associated GAD peptide, and an influenza-derived HA peptide. We show that the same chimeric TCR constructs can be used in each of the described assays facilitating sequential validation and prioritization steps leading to humanized animal models.
Collapse
Key Words
- APC, antigen presenting cells
- BM, bone marrow
- Ca2+, calcium
- Cell lines
- GAD, glutamic acid decarboxylase
- GFP, green fluorescent protein
- GWAS, Genome-wide association studies
- HA, Influenza hemagglutinin
- HLA
- HLA, Human leukocyte antigen
- HSCs, hematopoietic stem cells
- Humanized animal models
- MHC, major histocompatibility complex
- NFAT, Nuclear factor of activated T-cells
- RA, Rheumatoid arthritis
- RAG, Recombination-activating genes
- T1D, Type-1 diabetes
- TCR
- TCR, T cell receptor
- TCRa, TCR alpha
- TCRb, TCR beta
- TMR, HLA tetramer
- Tolerance
- hCD4, human CD4
- hTCR, human TCR
Collapse
Affiliation(s)
- Sanjaykumar V Boddul
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Ravi Kumar Sharma
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Anatoly Dubnovitsky
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden.,Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Bruno Raposo
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Christina Gerstner
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Yunbing Shen
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Vaishnavi Srinivasan Iyer
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden.,School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore
| | - Zsolt Kasza
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - William W Kwok
- Translational Research Program, BRI at Virginia Mason, Seattle, WA, USA
| | - Aaron R Winkler
- Department of Inflammation and Immunology, Pfizer Inc., Cambridge, MA, USA
| | - Lars Klareskog
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Maria Bettini
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Fredrik Wermeling
- Division of Rheumatology, Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
7
|
Ludvigsson J. Autoantigen Treatment in Type 1 Diabetes: Unsolved Questions on How to Select Autoantigen and Administration Route. Int J Mol Sci 2020; 21:E1598. [PMID: 32111075 PMCID: PMC7084272 DOI: 10.3390/ijms21051598] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/11/2022] Open
Abstract
Autoantigen treatment has been tried for the prevention of type 1 diabetes (T1D) and to preserve residual beta-cell function in patients with a recent onset of the disease. In experimental animal models, efficacy was good, but was insufficient in human subjects. Besides the possible minor efficacy of peroral insulin in high-risk individuals to prevent T1D, autoantigen prevention trials have failed. Other studies on autoantigen prevention and intervention at diagnosis are ongoing. One problem is to select autoantigen/s; others are dose and route. Oral administration may be improved by using different vehicles. Proinsulin peptide therapy in patients with T1D has shown possible minor efficacy. In patients with newly diagnosed T1D, subcutaneous injection of glutamic acid decarboxylase (GAD) bound to alum hydroxide (GAD-alum) can likely preserve beta-cell function, but the therapeutic effect needs to be improved. Intra-lymphatic administration may be a better alternative than subcutaneous administration, and combination therapy might improve efficacy. This review elucidates some actual problems of autoantigen therapy in the prevention and/or early intervention of type 1 diabetes.
Collapse
Affiliation(s)
- Johnny Ludvigsson
- Crown Princess Victoria Children´s Hospital and Div of Pediatrics, Dept of Biomedical and Clinical Sciences, Lnköping university, SE 58185 Linköping, Sweden
| |
Collapse
|
8
|
Cabello-Olmo M, Araña M, Radichev I, Smith P, Huarte E, Barajas M. New Insights into Immunotherapy Strategies for Treating Autoimmune Diabetes. Int J Mol Sci 2019; 20:ijms20194789. [PMID: 31561568 PMCID: PMC6801436 DOI: 10.3390/ijms20194789] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/18/2019] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes mellitus (T1D) is an autoimmune illness that affects millions of patients worldwide. The main characteristic of this disease is the destruction of pancreatic insulin-producing beta cells that occurs due to the aberrant activation of different immune effector cells. Currently, T1D is treated by lifelong administration of novel versions of insulin that have been developed recently; however, new approaches that could address the underlying mechanisms responsible for beta cell destruction have been extensively investigated. The strategies based on immunotherapies have recently been incorporated into a panel of existing treatments for T1D, in order to block T-cell responses against beta cell antigens that are very common during the onset and development of T1D. However, a complete preservation of beta cell mass as well as insulin independency is still elusive. As a result, there is no existing T1D targeted immunotherapy able to replace standard insulin administration. Presently, a number of novel therapy strategies are pursuing the goals of beta cell protection and normoglycemia. In the present review we explore the current state of immunotherapy in T1D by highlighting the most important studies in this field, and envision novel strategies that could be used to treat T1D in the future.
Collapse
Affiliation(s)
- Miriam Cabello-Olmo
- Biochemistry Area, Health Science Department, Faculty of Health Sciences, Public University of Navarra, 31008 Pamplona, Spain.
| | - Miriam Araña
- Biochemistry Area, Health Science Department, Faculty of Health Sciences, Public University of Navarra, 31008 Pamplona, Spain.
| | - Ilian Radichev
- Diabetes research group at Sanford Research, Sioux Falls, SD 57104, USA.
| | - Paul Smith
- Incyte Corporation, Wilmington, DE 19803, USA.
| | | | - Miguel Barajas
- Biochemistry Area, Health Science Department, Faculty of Health Sciences, Public University of Navarra, 31008 Pamplona, Spain.
| |
Collapse
|
9
|
Yong J, Tian J, Dang H, Wu TT, Atkinson MA, Sun R, Kaufman DL. Increased risk for T cell autoreactivity to ß-cell antigens in the mice expressing the A vy obesity-associated gene. Sci Rep 2019; 9:4269. [PMID: 30862859 PMCID: PMC6414670 DOI: 10.1038/s41598-019-38905-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 01/14/2019] [Indexed: 12/17/2022] Open
Abstract
There has been considerable debate as to whether obesity can act as an accelerator of type 1 diabetes (T1D). We assessed this possibility using transgenic mice (MIP-TF mice) whose ß-cells express enhanced green fluorescent protein (EGFP). Infecting these mice with EGFP-expressing murine herpes virus-68 (MHV68-EGFP) caused occasional transient elevation in their blood glucose, peri-insulitis, and Th1 responses to EGFP which did not spread to other ß-cell antigens. We hypothesized that obesity-related systemic inflammation and ß-cell stress could exacerbate the MHV68-EGFP-induced ß-cell autoreactivity. We crossed MIP-TF mice with Avy mice which develop obesity and provide models of metabolic disease alongside early stage T2D. Unlike their MIP-TF littermates, MHV68-EGFP-infected Avy/MIP-TF mice developed moderate intra-insulitis and transient hyperglycemia. MHV68-EGFP infection induced a more pronounced intra-insulitis in older, more obese, Avy/MIP-TF mice. Moreover, in MHV68-EGFP-infected Avy/MIP-TF mice, Th1 reactivity spread from EGFP to other ß-cell antigens. Thus, the spreading of autoreactivity among ß-cell antigens corresponded with the transition from peri-insulitis to intra-insulitis and occurred in obese Avy/MIP-TF mice but not lean MIP-TF mice. These observations are consistent with the notion that obesity-associated systemic inflammation and ß-cell stress lowers the threshold necessary for T cell autoreactivity to spread from EGFP to other ß-cell autoantigens.
Collapse
Affiliation(s)
- Jing Yong
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095-1735, United States.,Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, 92037, United States
| | - Jide Tian
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095-1735, United States
| | - Hoa Dang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095-1735, United States
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095-1735, United States
| | - Mark A Atkinson
- Departments of Pathology and Paediatrics, University of Florida Diabetes Institute, Gainesville, FL, 32610, United States
| | - Ren Sun
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095-1735, United States
| | - Daniel L Kaufman
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095-1735, United States.
| |
Collapse
|
10
|
Elding Larsson H, Lundgren M, Jonsdottir B, Cuthbertson D, Krischer J. Safety and efficacy of autoantigen-specific therapy with 2 doses of alum-formulated glutamate decarboxylase in children with multiple islet autoantibodies and risk for type 1 diabetes: A randomized clinical trial. Pediatr Diabetes 2018; 19:410-419. [PMID: 29171140 DOI: 10.1111/pedi.12611] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/18/2017] [Accepted: 10/22/2017] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Treatments have failed to delay or stop the autoimmune process, preceding onset of type 1 diabetes. We investigated if autoantigen-specific treatment with alum-formulated glutamate decarboxylase (GAD-Alum) was safe and affected progression to type 1 diabetes in children with islet autoimmunity. METHODS In an investigator-initiated, double-blind, placebo-controlled clinical trial, non-diabetic children aged 4 to 17.9 years with autoantibodies to glutamate decarboxylase (GADA) and at least one of insulinoma-associated protein 2, insulin or zinc-transporter 8, were randomized, stratified by 2 or ≥3 islet autoantibodies, to 2 injections of 20 μg GAD-Alum or placebo, 30 days apart. Main outcome was safety, investigated by adverse events, hematology, chemistry, thyroid and celiac autoimmunity and titers of islet autoantibodies, and efficacy, investigated by cumulative incidence of diabetes onset over 5-year follow-up. Secondary variables: change in first-phase insulin release (FPIR) after intravenous glucose tolerance tests, fasting, 120 minutes and Area under the curve (AUC) C-peptide and p-glucose after oral glucose tolerance tests and HbA1c. RESULTS Fifty children (median age: 5.2) were assigned 1:1 to GAD-Alum or placebo, all receiving full treatment and included in the analyses. GAD-Alum did not affect any safety parameter, while GADA titers increased (P = .001). Time to clinical diagnosis was not affected by treatment (hazard ratio, HR = 0.77, P = .574) in the full population or in the separate stratum groups. Treatment did not affect any of the secondary variables. CONCLUSIONS GAD-Alum as a subcutaneous prime and boost injection was safe in prediabetic young children but did not affect progression to type 1 diabetes. The safety of GAD-Alum should prove useful in future prevention studies.
Collapse
Affiliation(s)
- Helena Elding Larsson
- Department of Clinical Sciences/CRC, Skåne University Hospital, Lund University, Lund, Sweden
| | - Markus Lundgren
- Department of Clinical Sciences/CRC, Skåne University Hospital, Lund University, Lund, Sweden
| | - Berglind Jonsdottir
- Department of Clinical Sciences/CRC, Skåne University Hospital, Lund University, Lund, Sweden
| | - David Cuthbertson
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Jeffrey Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | | |
Collapse
|
11
|
Immature Dendritic Cell Therapy Confers Durable Immune Modulation in an Antigen-Dependent and Antigen-Independent Manner in Nonobese Diabetic Mice. J Immunol Res 2018; 2018:5463879. [PMID: 29651443 PMCID: PMC5832131 DOI: 10.1155/2018/5463879] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/19/2017] [Accepted: 11/27/2017] [Indexed: 01/25/2023] Open
Abstract
Dendritic cell (DC) immunotherapy has been effective for prevention of type 1 diabetes (T1D) in NOD mice but fails to protect if initiated after active autoimmunity. As autoreactivity expands inter- and intramolecularly during disease progression, we investigated whether DCs unpulsed or pulsed with β cell antigenic dominant determinants (DD), subdominant determinants (SD), and ignored determinants (ID) could prevent T1D in mice with advanced insulitis. We found that diabetes was significantly delayed by DC therapy. Of interest, DCs pulsed with SD or ID appeared to provide better protection. T lymphocytes from DC-treated mice acquired spontaneous proliferating capability during in vitro culture, which could be largely eliminated by IL-2 neutralizing antibodies. This trend maintained even 29 weeks after discontinuing DC therapy and appeared antigen-independent. Furthermore, CD4+Foxp3+ T regulatory cells (Tregs) from DC-treated mice proliferated more actively in vitro compared to the controls, and Tregs from DC-treated mice showed significantly enhanced immunosuppressive activities in contrast to those from the controls. Our study demonstrates that DC therapy leads to long-lasting immunomodulatory effects in an antigen-dependent and antigen-independent manner and provides evidence for peptide-based intervention during a clinically relevant window to guide DC-based immunotherapy for autoimmune diabetes.
Collapse
|
12
|
Hull CM, Peakman M, Tree TIM. Regulatory T cell dysfunction in type 1 diabetes: what's broken and how can we fix it? Diabetologia 2017; 60:1839-1850. [PMID: 28770318 PMCID: PMC6448885 DOI: 10.1007/s00125-017-4377-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/17/2017] [Indexed: 01/07/2023]
Abstract
Type 1 diabetes is an autoimmune disease characterised by the destruction of insulin producing beta cells in the pancreas. Whilst it remains unclear what the original triggering factors for this destruction are, observations from the natural history of human type 1 diabetes, including incidence rates in twins, suggest that the disease results from a combination of genetic and environmental factors. Whilst many different immune cells have been implicated, including members of the innate and adaptive immune systems, a view has emerged over the past 10 years that beta cell damage is mediated by the combined actions of CD4+ and CD8+ T cells with specificity for islet autoantigens. In health, these potentially pathogenic T cells are held in check by multiple regulatory mechanisms, known collectively as 'immunological tolerance'. This raises the question as to whether type 1 diabetes develops, at least in part, as a result of a defect in one or more of these control mechanisms. Immunological tolerance includes both central mechanisms (purging of the T cell repertoire of high-affinity autoreactive T cells in the thymus) and peripheral mechanisms, a major component of which is the action of a specialised subpopulation of T cells, known as regulatory T cells (Tregs). In this review, we highlight the evidence suggesting that a reduction in the functional capacity of different Treg populations contributes to disease development in type 1 diabetes. We also address current controversies regarding the putative causes of this defect and discuss strategies to correct it as a means to reduce or prevent islet destruction in a clinical setting.
Collapse
Affiliation(s)
- Caroline M Hull
- Programme of Infection and Immunity, Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, Borough Wing, Guy's Hospital, London, SE1 9RT, UK.
| | - Mark Peakman
- Programme of Infection and Immunity, Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Timothy I M Tree
- Programme of Infection and Immunity, Department of Immunobiology, Faculty of Life Sciences and Medicine, King's College London, Borough Wing, Guy's Hospital, London, SE1 9RT, UK.
- NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK.
| |
Collapse
|
13
|
Affiliation(s)
- Johnny Ludvigsson
- Divsion of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping,Sweden
| |
Collapse
|
14
|
Beam CA, MacCallum C, Herold KC, Wherrett DK, Palmer J, Ludvigsson J. GAD vaccine reduces insulin loss in recently diagnosed type 1 diabetes: findings from a Bayesian meta-analysis. Diabetologia 2017; 60:43-49. [PMID: 27704166 PMCID: PMC5856235 DOI: 10.1007/s00125-016-4122-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/31/2016] [Indexed: 01/13/2023]
Abstract
AIMS/HYPOTHESIS GAD is a major target of the autoimmune response that occurs in type 1 diabetes mellitus. Randomised controlled clinical trials of a GAD + alum vaccine in human participants have so far given conflicting results. METHODS In this study, we sought to see whether a clearer answer to the question of whether GAD65 has an effect on C-peptide could be reached by combining individual-level data from the randomised controlled trials using Bayesian meta-analysis to estimate the probability of a positive biological effect (a reduction in C-peptide loss compared with placebo approximately 1 year after the GAD vaccine). RESULTS We estimate that there is a 98% probability that 20 μg GAD with alum administered twice yields a positive biological effect. The effect is probably a 15-20% reduction in the loss of C-peptide at approximately 1 year after treatment. This translates to an annual expected loss of between -0.250 and -0.235 pmol/ml in treated patients compared with an expected 2 h AUC loss of -0.294 pmol/ml at 1 year for untreated newly diagnosed patients. CONCLUSIONS/INTERPRETATION The biological effect of this vaccination should be developed further in order to reach clinically desirable reductions in insulin loss in patients recently diagnosed with type 1 diabetes.
Collapse
Affiliation(s)
- Craig A Beam
- Division of Epidemiology and Biostatistics, Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D. School of Medicine, 1000 Oakland Drive, Kalamazoo, MI, 49008-8071, USA.
| | - Colleen MacCallum
- Division of Epidemiology and Biostatistics, Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D. School of Medicine, 1000 Oakland Drive, Kalamazoo, MI, 49008-8071, USA
| | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, USA
| | - Diane K Wherrett
- Department of Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Jerry Palmer
- University of Washington and VA Puget Sound Health Care System, Seattle, WA, USA
| | - Johnny Ludvigsson
- Divison of Pediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | |
Collapse
|
15
|
Liu X, Zhang S, Li X, Zheng P, Hu F, Zhou Z. Vaccination with a co-expression DNA plasmid containing GAD65 fragment gene and IL-10 gene induces regulatory CD4(+) T cells that prevent experimental autoimmune diabetes. Diabetes Metab Res Rev 2016; 32:522-33. [PMID: 26797873 DOI: 10.1002/dmrr.2780] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 12/11/2015] [Accepted: 01/15/2016] [Indexed: 01/26/2023]
Abstract
BACKGROUND The non-obese diabetic (NOD) mouse is a commonly used animal model for studying type 1 diabetes (T1D). The aims of our study were to explore the diabetes-preventive effect in NOD mice and the potential mechanisms of an optimized co-expression DNA vaccine containing GAD65 fragment gene with the IL-10 gene (SGAD65190-315 /IL-10). METHODS Female NOD mice at the age of 3-4 weeks old were randomly divided into two groups and received intra-muscular injection of either blank pBudCE4.l vector (n = 34) or pBudCE4.l carrying the SGAD65190-315 /IL-10 (n = 32). The incidence of diabetes was monitored up to 30 weeks of age. The severity of insulitis, apoptosis rate of β cells and relevant mechanisms were examined. RESULTS Administration with SGAD65190-315 /IL-10 blocked the onset of autoimmune diabetes in NOD mice, significantly suppressed islet inflammation, inhibited the apoptosis of islet β cells, induced immune tolerance to autoantigen GAD65 and proinsulin and shifted the Th1/Th2 balance towards Th2. More importantly, the frequencies of CD4(+) CD25(+) Foxp3(+) regulatory T cells (Tregs) in the spleen and pancreatic lymph nodes in vaccine-immunized mice were significantly increased, and these Tregs were GAD65-reactive. In addition, Treg depletion by anti-CD25 mAb administration abolished the protective effects of SGAD65190-315 /IL-10 on diabetes and insulitis. Moreover, depletion of CD4(+) CD25(+) T cells using magnetic-activated cell sorting impaired the protective effect of SGAD65190-315 /IL-10 vaccination on adoptive transfer of diabetes. CONCLUSIONS Our data suggested that SGAD65190-315 /IL-10 DNA vaccine had protective effects on T1D by upregulating autoantigen-reactive Tregs. Our findings may provide a novel preventive therapy for T1D. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Xinyuan Liu
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Song Zhang
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Xia Li
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Peilin Zheng
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Fang Hu
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, Second Xiangya Hospital, Central South University, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China, 410011
| |
Collapse
|
16
|
Davis IC, Randell J, Davis SN. Immunotherapies currently in development for the treatment of type 1 diabetes. Expert Opin Investig Drugs 2016; 24:1331-41. [PMID: 26364507 DOI: 10.1517/13543784.2015.1075973] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Type I diabetes (T1DM) is an autoimmune disorder that affects the pancreas' ability to produce insulin. While T1DM can be managed using insulin therapy, patients face financial burden, serious complications and premature mortality, from the disease. Efforts have sought to define and ultimately suppress the underlying autoimmune attack that results in T1DM. AREAS COVERED The authors lay out promising immunosuppressive and immunomodulating drugs currently in development for T1DM and outline options for future immune treatment for the disorder. There have been several pharmacological strategies to combat the immune attack which will serve as the organization for this review: antigen-specific therapies; monoclonal antibodies; fusion proteins; alternate Treg affectors. EXPERT OPINION Immunosuppression and immunomodulation studies in T1DM demonstrated differing levels of slowing the progression of the immune attack; however, no single therapeutic approach provides a lasting halt of the immune attack and remission of the disease. The immunosuppressants (teplizumab, rituximab and abatacept) show promise in slowing the T1DM progressions for a specific subpopulation of T1DM patients, but this approach appears temporary and has the potential for unwanted side affects. Combination therapies may have the greatest chance of achieving durable cessation of the T1DM autoimmune attack.
Collapse
Affiliation(s)
- Ian C Davis
- a 1 University of Maryland School of Medicine , 3805 Greenway, Baltimore, MD 21218, USA
| | | | - Stephen N Davis
- c 3 University of Maryland School of Medicine , 22 South Greene St. N3W42, Baltimore, MD 21201, USA +1 41 0328 2488 ; +1 41 0328 8688 ;
| |
Collapse
|
17
|
Krishnamurthy B, Selck C, Chee J, Jhala G, Kay TWH. Analysis of antigen specific T cells in diabetes - Lessons from pre-clinical studies and early clinical trials. J Autoimmun 2016; 71:35-43. [PMID: 27083395 DOI: 10.1016/j.jaut.2016.03.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 01/06/2023]
Abstract
Antigen-specific immune tolerance promises to provide safe and effective therapies to prevent type 1 diabetes (T1D). Antigen-specific therapy requires two components: well-defined, clinically relevant autoantigens; and safe approaches to inducing tolerance in T cells specific for these antigens. Proinsulin is a critical autoantigen in both NOD mice, based on knockout mouse studies and induction of immune tolerance to proinsulin preventing disease whereas most antigens cannot, and also in human T1D based on proinsulin-specific T cells being found in the islets of affected individuals and the early appearance of insulin autoantibodies. Effective antigen-specific therapies that prevent T1D in humans have not yet been developed although doubt remains about the best molecular form of the antigen, the dose and the route of administration. Preclinical studies suggest that antigen specific therapy is most useful when administered before onset of autoimmunity but this time-window has not been tested in humans until the recent "pre-point" study. There may be a 'window of opportunity' during the neonatal period when 'vaccine' like administration of proinsulin for a short period may be sufficient to prevent diabetes. After the onset of autoimmunity, naive antigen-specific T cells have differentiated into antigen-experienced memory cells and the immune responses have spread to multiple antigens. Induction of tolerance at this stage becomes more difficult although recent studies have suggested generation of antigen-specific TR1 cells can inhibit memory T cells. Preclinical studies are required to identify additional 'help' that is required to induce tolerance to memory T cells and develop protocols for effective therapy in individuals with established autoimmunity.
Collapse
Affiliation(s)
- Balasubramanian Krishnamurthy
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia
| | - Claudia Selck
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia
| | - Jonathan Chee
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia
| | - Guarang Jhala
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia
| | - Thomas W H Kay
- St. Vincent's Institute, 41 Victoria Parade, Fitzroy, 3065, Victoria, Australia; The University of Melbourne Department of Medicine, St Vincent's Hospital, Fitzroy, 3065, Victoria, Australia.
| |
Collapse
|
18
|
Abstract
Type 1 diabetes (T1D) results from a chronic and selective destruction of insulin-secreting β-cells within the islets of Langerhans of the pancreas by autoreactive CD4(+) and CD8(+) T lymphocytes. The use of animal models of T1D was instrumental for deciphering the steps of the autoimmune process leading to T1D. The non-obese diabetic (NOD) mouse and the bio-breeding (BB) rat spontaneously develop the disease similar to the human pathology in terms of the immune responses triggering autoimmune diabetes and of the genetic and environmental factors influencing disease susceptibility. The generation of genetically modified models allowed refining our understanding of the etiology and the pathogenesis of the disease. In the present review, we provide an overview of the experimental models generated and used to gain knowledge on the molecular and cellular mechanisms underlying the breakdown of self-tolerance in T1D and the progression of the autoimmune response. Immunotherapeutic interventions designed in these animal models and translated into the clinical arena in T1D patients will also be discussed.
Collapse
|
19
|
Bergamin CS, Dib SA. Enterovirus and type 1 diabetes: What is the matter? World J Diabetes 2015; 6:828-839. [PMID: 26131324 PMCID: PMC4478578 DOI: 10.4239/wjd.v6.i6.828] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/30/2015] [Accepted: 04/09/2015] [Indexed: 02/05/2023] Open
Abstract
A complex interaction of genetic and environmental factors can trigger the immune-mediated mechanism responsible for type 1 diabetes mellitus (T1DM) establishment. Environmental factors may initiate and possibly sustain, accelerate, or retard damage to β-cells. The role of environmental factors in this process has been exhaustive studied and viruses are among the most probable ones, especially enteroviruses. Improvements in enterovirus detection methods and randomized studies with patient follow-up have confirmed the importance of human enterovirus in the pathogenesis of T1DM. The genetic risk of T1DM and particular innate and acquired immune responses to enterovirus infection contribute to a tolerance to T1DM-related autoantigens. However, the frequency, mechanisms, and pathways of virally induced autoimmunity and β-cell destruction in T1DM remain to be determined. It is difficult to investigate the role of enterovirus infection in T1DM because of several concomitant mechanisms by which the virus damages pancreatic β-cells, which, consequently, may lead to T1DM establishment. Advances in molecular and genomic studies may facilitate the identification of pathways at earlier stages of autoimmunity when preventive and therapeutic approaches may be more effective.
Collapse
|
20
|
Fousteri G, Jofra T, Di Fonte R, Battaglia M. Combination of an Antigen-Specific Therapy and an Immunomodulatory Treatment to Simultaneous Block Recurrent Autoimmunity and Alloreactivity in Non-Obese Diabetic Mice. PLoS One 2015; 10:e0127631. [PMID: 26080071 PMCID: PMC4469694 DOI: 10.1371/journal.pone.0127631] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/16/2015] [Indexed: 12/12/2022] Open
Abstract
Restoration of endogenous insulin production by islet transplantation is considered a curative option for patients with type 1 diabetes. However, recurrent autoimmunity and alloreactivity cause graft rejection hindering successful transplantation. Here we tested whether transplant tolerance to allogeneic islets could be achieved in non-obese diabetic (NOD) mice by simultaneously tackling autoimmunity via antigen-specific immunization, and alloreactivity via granulocyte colony stimulating factor (G-CSF) and rapamycin (RAPA) treatment. Immunization with insB9-23 peptide alone or in combination with two islet peptides (IGRP206-214 and GAD524-543) in incomplete Freund’s adjuvant (IFA) were tested for promoting syngeneic pancreatic islet engraftment in spontaneously diabetic NOD mice. Treatment with G-CSF/RAPA alone or in combination with insB9-23/IFA was examined for promoting allogeneic islet engraftment in the same mouse model. InsB9-23/IFA immunization significantly prolonged syngeneic pancreatic islet survival in NOD mice by a mechanism that necessitated the presence of CD4+CD25+ T regulatory (Treg) cells, while combination of three islet epitopes was less efficacious in controlling recurrent autoimmunity. G-CSF/RAPA treatment was unable to reverse T1D or control recurrent autoimmunity but significantly prolonged islet allograft survival in NOD mice. Blockade of interleukin-10 (IL-10) during G-CSF/RAPA treatment resulted in allograft rejection suggesting that IL-10-producing cells were fundamental to achieve transplant tolerance. G-CSF/RAPA treatment combined with insB9-23/IFA did not further increase the survival of allogeneic islets. Thus, insB9-23/IFA immunization controls recurrent autoimmunity and G-CSF/RAPA treatment limits alloreactivity, however their combination does not further promote allogeneic pancreatic islet engraftment in NOD mice.
Collapse
Affiliation(s)
- Georgia Fousteri
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
- * E-mail: (GF); (MB)
| | - Tatiana Jofra
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
| | - Roberta Di Fonte
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
| | - Manuela Battaglia
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Via Olgettina 58, Milan, Italy
- * E-mail: (GF); (MB)
| |
Collapse
|
21
|
Schroder PM, Khattar M, Baum CE, Miyahara Y, Chen W, Vyas R, Muralidharan S, Mierzejewska B, Stepkowski SM. PD-1-dependent restoration of self-tolerance in the NOD mouse model of diabetes after transient anti-TCRβ mAb therapy. Diabetologia 2015; 58:1309-18. [PMID: 25794782 DOI: 10.1007/s00125-015-3564-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/03/2015] [Indexed: 01/07/2023]
Abstract
AIMS/HYPOTHESIS T cells play a major role in the pathogenesis of type 1 diabetes, and there is great interest in developing curative immunotherapies targeting these cells. In this study, a monoclonal antibody (mAb) targeting the T cell receptor β-chain (TCRβ) was investigated for its ability to prevent and reverse disease in mouse models of diabetes. METHODS RIP-OVA(hi) (C57BL/6-Tg(Ins2-OVA)59Wehi/WehiJ) mice adoptively transferred with ovalbumin-specific T cells (an induced model of diabetes) and NOD mice (a spontaneous model of diabetes) were used to test anti-TCRβ mAb therapy as a means of preventing and reversing type 1 diabetes. RESULTS A single dose of anti-TCRβ completely prevented disease in RIP-OVA(hi) mice without inducing the release of inflammatory cytokines. Transient anti-TCRβ therapy prevented diabetes in 90% of NOD mice and reversed the disease after its onset in 73% of NOD mice. Long after the remission of type 1 diabetes, the anti-TCRβ treated mice were able to reject BALB/c skin allografts with normal kinetics while maintaining normoglycaemia. Treatment did not cause significant reductions in lymphocyte numbers in the spleen or pancreatic lymph nodes, but did result in a decreased percentage of chemokine receptor 9 (CCR9) positive, CD8(+) T cells. Notably, anti-TCRβ therapy increased the expression of programmed death 1 (PD-1) on the surface of the T cells; PD-1 expression is important for maintaining anti-TCRβ-induced self-tolerance, as type 1 diabetes recurs in mice following a blockade of PD-1 signalling. CONCLUSIONS/INTERPRETATION Anti-TCRβ mAb is a safe and effective immunotherapy that results in reduced numbers of CCR9(+) T cells, an increased expression of PD-1 on T cells and the restoration of self-tolerance in NOD mice.
Collapse
MESH Headings
- Allografts
- Animals
- Antibodies, Monoclonal/pharmacology
- Blood Glucose/analysis
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cytokines/blood
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Female
- Glucose Tolerance Test
- Inflammation
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Programmed Cell Death 1 Receptor/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, CCR/metabolism
Collapse
Affiliation(s)
- Paul M Schroder
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine, 3000 Arlington Avenue, HEB 263A, Toledo, OH, 43614, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Bluestone JA, Bour-Jordan H, Cheng M, Anderson M. T cells in the control of organ-specific autoimmunity. J Clin Invest 2015; 125:2250-60. [PMID: 25985270 DOI: 10.1172/jci78089] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Immune tolerance is critical to the avoidance of unwarranted immune responses against self antigens. Multiple, non-redundant checkpoints are in place to prevent such potentially deleterious autoimmune responses while preserving immunity integral to the fight against foreign pathogens. Nevertheless, a large and growing segment of the population is developing autoimmune diseases. Deciphering cellular and molecular pathways of immune tolerance is an important goal, with the expectation that understanding these pathways will lead to new clinical advances in the treatment of these devastating diseases. The vast majority of autoimmune diseases develop as a consequence of complex mechanisms that depend on genetic, epigenetic, molecular, cellular, and environmental elements and result in alterations in many different checkpoints of tolerance and ultimately in the breakdown of immune tolerance. The manifestations of this breakdown are harmful inflammatory responses in peripheral tissues driven by innate immunity and self antigen-specific pathogenic T and B cells. T cells play a central role in the regulation and initiation of these responses. In this Review we summarize our current understanding of the mechanisms involved in these fundamental checkpoints, the pathways that are defective in autoimmune diseases, and the therapeutic strategies being developed with the goal of restoring immune tolerance.
Collapse
|
23
|
Abstract
Studies over the past 35 years in the nonobese diabetic (NOD) mouse have shown that a number of agents can prevent or even reverse type 1 diabetes mellitus (T1DM); however, these successes have not been replicated in human clinical trials. Although some of these interventions have delayed disease onset or progression in subsets of participants, none have resulted in a complete cure. Even in the most robust responders, the treatments do not permanently preserve insulin secretion or stimulate the proliferation of β cells, as has been observed in mice. The shortfalls of translating NOD mouse studies into the clinic questions the value of using this model in preclinical studies. In this Perspectives, we suggest how immunological and genetic differences between NOD mice and humans might contribute to the differential outcomes and suggest ways in which the mouse model might be modified or applied as a tool to develop treatments and improve understanding of clinical trial outcomes.
Collapse
Affiliation(s)
- James C Reed
- Department of Immunobiology, 300 George Street, #353E, New Haven, CT 06520, USA
| | - Kevan C Herold
- Department of Immunobiology, Department of Internal Medicine, Yale University, 300 George Street, #353E, New Haven, CT 06520, USA
| |
Collapse
|
24
|
Smilek DE, Ehlers MR, Nepom GT. Restoring the balance: immunotherapeutic combinations for autoimmune disease. Dis Model Mech 2014; 7:503-13. [PMID: 24795433 PMCID: PMC4007402 DOI: 10.1242/dmm.015099] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Autoimmunity occurs when T cells, B cells or both are inappropriately activated, resulting in damage to one or more organ systems. Normally, high-affinity self-reactive T and B cells are eliminated in the thymus and bone marrow through a process known as central immune tolerance. However, low-affinity self-reactive T and B cells escape central tolerance and enter the blood and tissues, where they are kept in check by complex and non-redundant peripheral tolerance mechanisms. Dysfunction or imbalance of the immune system can lead to autoimmunity, and thus elucidation of normal tolerance mechanisms has led to identification of therapeutic targets for treating autoimmune disease. In the past 15 years, a number of disease-modifying monoclonal antibodies and genetically engineered biologic agents targeting the immune system have been approved, notably for the treatment of rheumatoid arthritis, inflammatory bowel disease and psoriasis. Although these agents represent a major advance, effective therapy for other autoimmune conditions, such as type 1 diabetes, remain elusive and will likely require intervention aimed at multiple components of the immune system. To this end, approaches that manipulate cells ex vivo and harness their complex behaviors are being tested in preclinical and clinical settings. In addition, approved biologic agents are being examined in combination with one another and with cell-based therapies. Substantial development and regulatory hurdles must be overcome in order to successfully combine immunotherapeutic biologic agents. Nevertheless, such combinations might ultimately be necessary to control autoimmune disease manifestations and restore the tolerant state.
Collapse
Affiliation(s)
- Dawn E Smilek
- The Immune Tolerance Network, 185 Berry Street #3515, San Francisco, CA 94107, USA
| | | | | |
Collapse
|
25
|
Abstract
Autoimmune diseases are common chronic disorders that not only have a major impact on the quality of life but are also potentially life-threatening. Treatment modalities that are currently favored have conferred significant clinical benefits, but they may have considerable side effects. An optimal treatment strategy for autoimmune disease would specifically target disease-associated antigens and limit systemic side effects. Similar to allergen-specific immunotherapy for allergic rhinitis, antigen-specific immunotherapy for autoimmune disease aims to induce immune deviation and promote tolerance to specific antigens. In this review, we present the current status of studies and clinical trials in both human and animal hosts that use antigen-based immunotherapy for autoimmune disease.
Collapse
Affiliation(s)
- Darren Lowell Hirsch
- Division of Allergy and Immunology, North Shore-Long Island Jewish Health System/Hofstra North Shore-LIJ School of Medicine, New Hyde Park, NY, USA
| | - Punita Ponda
- Division of Allergy and Immunology, North Shore-Long Island Jewish Health System/Hofstra North Shore-LIJ School of Medicine, New Hyde Park, NY, USA
| |
Collapse
|
26
|
Tian J, Dang H, Nguyen AV, Chen Z, Kaufman DL. Combined therapy with GABA and proinsulin/alum acts synergistically to restore long-term normoglycemia by modulating T-cell autoimmunity and promoting β-cell replication in newly diabetic NOD mice. Diabetes 2014; 63:3128-34. [PMID: 25146474 PMCID: PMC4141368 DOI: 10.2337/db13-1385] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Antigen-based therapies (ABTs) fail to restore normoglycemia in newly diabetic NOD mice, perhaps because too few β-cells remain by the time that ABT-induced regulatory responses arise and spread. We hypothesized that combining a fast-acting anti-inflammatory agent with an ABT could limit pathogenic responses while ABT-induced regulatory responses arose and spread. γ-Aminobutyric acid (GABA) administration can inhibit inflammation, enhance regulatory T-cell (Treg) responses, and promote β-cell replication in mice. We examined the effect of combining a prototypic ABT, proinsulin/alum, with GABA treatment in newly diabetic NOD mice. Proinsulin/alum monotherapy failed to correct hyperglycemia, while GABA monotherapy restored normoglycemia for a short period. Combined treatment restored normoglycemia in the long term with apparent permanent remission in some mice. Proinsulin/alum monotherapy induced interleukin (IL)-4- and IL-10-secreting T-cell responses that spread to other β-cell autoantigens. GABA monotherapy induced moderate IL-10 (but not IL-4) responses to β-cell autoantigens. Combined treatment synergistically reduced spontaneous type 1 T-helper cell responses to autoantigens, ABT-induced IL-4 and humoral responses, and insulitis, but enhanced IL-10 and Treg responses and promoted β-cell replication in the islets. Thus, combining ABT with GABA can inhibit pathogenic T-cell responses, induce Treg responses, promote β-cell replication, and effectively restore normoglycemia in newly diabetic NOD mice. Since these treatments appear safe for humans, they hold promise for type 1 diabetes intervention.
Collapse
Affiliation(s)
- Jide Tian
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| | - Hoa Dang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| | - An Viet Nguyen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| | - Zheying Chen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| | - Daniel L Kaufman
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
27
|
Bruni A, Gala-Lopez B, Pepper AR, Abualhassan NS, Shapiro AMJ. Islet cell transplantation for the treatment of type 1 diabetes: recent advances and future challenges. Diabetes Metab Syndr Obes 2014; 7:211-23. [PMID: 25018643 PMCID: PMC4075233 DOI: 10.2147/dmso.s50789] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Islet transplantation is a well-established therapeutic treatment for a subset of patients with complicated type I diabetes mellitus. Prior to the Edmonton Protocol, only 9% of the 267 islet transplant recipients since 1999 were insulin independent for >1 year. In 2000, the Edmonton group reported the achievement of insulin independence in seven consecutive patients, which in a collaborative team effort propagated expansion of clinical islet transplantation centers worldwide in an effort to ameliorate the consequences of this disease. To date, clinical islet transplantation has established improved success with insulin independence rates up to 5 years post-transplant with minimal complications. In spite of marked clinical success, donor availability and selection, engraftment, and side effects of immunosuppression remain as existing obstacles to be addressed to further improve this therapy. Clinical trials to improve engraftment, the availability of insulin-producing cell sources, as well as alternative transplant sites are currently under investigation to expand treatment. With ongoing experimental and clinical studies, islet transplantation continues to be an exciting and attractive therapy to treat type I diabetes mellitus with the prospect of shifting from a treatment for some to a cure for all.
Collapse
Affiliation(s)
- Anthony Bruni
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Boris Gala-Lopez
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Andrew R Pepper
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nasser S Abualhassan
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - AM James Shapiro
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
28
|
Abstract
Type 1 diabetes mellitus (T1DM) is the result of autoimmune destruction of pancreatic β cells in genetically predisposed individuals with impaired immune regulation. The insufficiency in the modulation of immune attacks on the β cells might be partly due to genetic causes; indeed, several of the genetic variants that predispose individuals to T1DM have functional features of impaired immune regulation. Whilst defects in immune regulation in patients with T1DM have been identified, many patients seem to have immune regulatory capacities that are indistinguishable from those of healthy individuals. Insight into the regulation of islet autoimmunity might enable us to restore immune imbalances with therapeutic interventions. In this Review, we discuss the current knowledge on immune regulation and dysfunction in humans that is the basis of tissue-specific immune regulation as an alternative to generalized immune suppression.
Collapse
Affiliation(s)
- Bart O Roep
- Leiden University Medical Center, Department of Immunohaematology & Blood Transfusion, P. O. Box 9600, NL-2300 RC Leiden, Netherlands
| | - Timothy I M Tree
- Department of Immunobiology, King's College London, School of Medicine, London SE1 9RT, UK
| |
Collapse
|
29
|
Abstract
This paper reviews the presentation of peptides by major histocompatibility complex (MHC) class II molecules in the autoimmune diabetes of the nonobese diabetic (NOD) mouse. Islets of Langerhans contain antigen-presenting cells that capture the proteins and peptides of the beta cells' secretory granules. Peptides bound to I-A(g7), the unique MHC class II molecule of NOD mice, are presented in islets and in pancreatic lymph nodes. The various beta cell-derived peptides interact with selected CD4 T cells to cause inflammation and beta cell demise. Many autoreactive T cells are found in NOD mice, but not all have a major role in the initiation of the autoimmune process. I emphasize here the evidence pointing to insulin autoreactivity as a seminal component in the diabetogenic process.
Collapse
Affiliation(s)
- Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| |
Collapse
|
30
|
Wan X, Guloglu FB, VanMorlan AM, Rowland LM, Zaghouani S, Cascio JA, Dhakal M, Hoeman CM, Zaghouani H. Recovery from overt type 1 diabetes ensues when immune tolerance and β-cell formation are coupled with regeneration of endothelial cells in the pancreatic islets. Diabetes 2013; 62:2879-89. [PMID: 23715620 PMCID: PMC3717841 DOI: 10.2337/db12-1281] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immune modulation of pancreatic inflammation induces recovery from type 1 diabetes (T1D), but remission was not durable, perhaps because of an inability to sustain the formation and function of new pancreatic β-cells. We have previously shown that Ig-GAD2, carrying GAD 206-220 peptide, induced in hyperglycemic mice immune modulation that was able to control pancreatic inflammation, stimulate β-cell regeneration, and prevent T1D progression. Herein, we show that the same Ig-GAD2 regimen given to mice with overt T1D was unable to reverse the course of disease despite eradication of Th1 and Th17 cells from the pancreas. However, the regimen was able to sustain recovery from T1D when Ig-GAD2 was accompanied with transfer of bone marrow (BM) cells from healthy donors. Interestingly, alongside immune modulation, there was concomitant formation of new β-cells and endothelial cells (ECs) in the pancreas. The new β-cells were of host origin while the donor BM cells gave rise to the ECs. Moreover, transfer of purified BM endothelial progenitors instead of whole BM cells sustained both β-cell and EC formation and reversal of diabetes. Thus, overcoming T1D requires both immune modulation and repair of the islet vascular niche to preserve newly formed β-cells.
Collapse
Affiliation(s)
- Xiaoxiao Wan
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - F. Betul Guloglu
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Amie M. VanMorlan
- Department of Child Health, University of Missouri School of Medicine, Columbia, Missouri
| | - Linda M. Rowland
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Sarah Zaghouani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Jason A. Cascio
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Mermagya Dhakal
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Christine M. Hoeman
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
| | - Habib Zaghouani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri
- Department of Child Health, University of Missouri School of Medicine, Columbia, Missouri
- Department of Neurology, University of Missouri School of Medicine, Columbia, Missouri
- Corresponding author: Habib Zaghouani,
| |
Collapse
|
31
|
Schneider DA, Kretowicz AM, von Herrath MG. Emerging immune therapies in type 1 diabetes and pancreatic islet transplantation. Diabetes Obes Metab 2013. [PMID: 23194064 DOI: 10.1111/dom.12046] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In type 1 diabetes (T1D) the immune system attacks insulin-producing pancreatic β-cells. Unfortunately, our ability to curb this pathogenic autoimmune response in a disease- and organ-specific manner is still very limited due to the inchoate understanding of the exact nature and the kinetics of the immunological pathomechanisms that lead to T1D. None of the clinical immune interventions thus far, which focused primarily on new-onset disease, were successful in producing lasting remission or curbing recurrent autoimmunity. However, these studies do provide us access to a tremendous amount of clinical data and specimens, which will aid us in revising our therapeutical approaches and defining the highly needed paradigm shift in T1D immunotherapy. Analysing the foundation and the results of the most current T1D immunotherapeutic trials, this article gives an outlook for future directions of the field.
Collapse
Affiliation(s)
- D A Schneider
- Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | | | | |
Collapse
|
32
|
Boettler T, Pagni PP, Jaffe R, Cheng Y, Zerhouni P, von Herrath M. The clinical and immunological significance of GAD-specific autoantibody and T-cell responses in type 1 diabetes. J Autoimmun 2013; 44:40-8. [PMID: 23770292 DOI: 10.1016/j.jaut.2013.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/20/2013] [Accepted: 05/22/2013] [Indexed: 01/12/2023]
Abstract
Antigen-specific interventions are desirable approaches in Type 1 Diabetes (T1D) as they can alter islet-specific autoimmunity without systemic side effects. Glutamic acid decarboxylase of 65 kDa (GAD65) is a major autoantigen in type 1 diabetes (T1D) and GAD-specific autoimmunity is a common feature of T1D in humans but also in mouse models of the disease. In humans, administration of the GAD65 protein in an alum formulation has been shown to reduce C-peptide decline in recently diagnosed patients, however, these observations were not confirmed in subsequent phase II/III clinical trials. As GAD-based immune interventions in different formulations have successfully been employed to prevent the establishment of T1D in mouse models of T1D, we sought to analyze the efficacy of GAD-alum treatment and the effects on the GAD-specific immune response in two different mouse models of T1D. Consistent with the latest clinical trials, mice treated with GAD-alum were not protected from diabetes, although GAD-alum induced a GAD-specific Th2-deviated immune response in transgenic rat insulin promoter-glycoprotein (RIP-GP) mice. These observations underline the importance of a thorough, preclinical evaluation of potential drugs before the initiation of clinical trials.
Collapse
Affiliation(s)
- Tobias Boettler
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
33
|
Herold KC, Vignali DAA, Cooke A, Bluestone JA. Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat Rev Immunol 2013; 13:243-56. [PMID: 23524461 PMCID: PMC4172461 DOI: 10.1038/nri3422] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Type 1 diabetes (T1D) remains an important health problem, particularly in western countries, where the incidence has been increasing in younger children. In 1986, Eisenbarth described T1D as a chronic autoimmune disease. Work over the past three-and-a-half decades has identified many of the genetic, immunological and environmental factors that are involved in the disease and have led to hypotheses concerning its pathogenesis. Clinical trials have been conducted to test these hypotheses but have had mixed results. Here, we discuss the findings that have led to our current concepts of the disease mechanisms involved in T1D and the clinical studies promoted by these studies. The findings from preclinical and clinical studies support the original proposed model for how T1D develops but have also suggested that this disease is more complex than was originally thought and will require broader treatment approaches.
Collapse
Affiliation(s)
- Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, Connecticut 06520, USA.
| | | | | | | |
Collapse
|
34
|
CHEN WENHAO, XIE AINI, CHAN LAWRENCE. Mechanistic basis of immunotherapies for type 1 diabetes mellitus. Transl Res 2013; 161:217-29. [PMID: 23348026 PMCID: PMC3602320 DOI: 10.1016/j.trsl.2012.12.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/07/2012] [Accepted: 12/28/2012] [Indexed: 01/10/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease for which there is no cure. The pancreatic beta cells are the source of insulin that keeps blood glucose normal. When susceptible individuals develop T1D, their beta cells are destroyed by autoimmune T lymphocytes and no longer produce insulin. T1D patients therefore depend on daily insulin injections for survival. Gene therapy in T1D aims at the induction of new islets to replace those that have been destroyed by autoimmunity. A major goal of T1D research is to restore functional beta cell mass while eliminating diabetogenic T cells in the hope of achieving insulin independence. Multiple therapeutic strategies for the generation of new beta cells have been under intense investigations. However, newly formed beta cells would be immediately destroyed by diabetogenic T cells. Therefore, successful islet induction therapy must be supported by potent immunotherapy that will protect the newly formed beta cells. Herein, we will summarize the current information on immunotherapies that aim at modifying T cell response to beta cells. We will first outline the immune mechanisms that underlie T1D development and progression and review the scientific background and rationale for specific modes of immunotherapy. Numerous clinical trials using antigen-specific strategies and immune-modifying drugs have been published, though most have proved too toxic or have failed to provide long-term beta cell protection. To develop an effective immunotherapy, there must be a continued effort on defining the molecular basis that underlies T cell response to pancreatic islet antigens in T1D.
Collapse
Affiliation(s)
- WENHAO CHEN
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine and Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA 77030
| | - AINI XIE
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine and Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA 77030
| | - LAWRENCE CHAN
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine and Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA 77030
| |
Collapse
|
35
|
|
36
|
Gastrointestinal Tract and Endocrine System. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
37
|
Cabrera SM, Rigby MR, Mirmira RG. Targeting regulatory T cells in the treatment of type 1 diabetes mellitus. Curr Mol Med 2012; 12:1261-72. [PMID: 22709273 PMCID: PMC3709459 DOI: 10.2174/156652412803833634] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 02/20/2012] [Accepted: 02/21/2012] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is a T cell-mediated autoimmune disease resulting in islet β cell destruction, hypoinsulinemia, and severely altered glucose homeostasis. T1DM has classically been attributed to the pathogenic actions of auto-reactive effector T cells(Teffs) on the β cell. Recent literature now suggests that a failure of a second T cell subtype, known as regulatory T cells (Tregs), plays a critical role in the development of T1DM. During immune homeostasis, Tregs counterbalance the actions of autoreactive Teff cells, thereby participating in peripheral tolerance. An imbalance in the activity between Teff and Tregs may be crucial in the breakdown of peripheral tolerance, leading to the development of T1DM. In this review, we summarize our current understanding of Treg function in health and in T1DM, and examine the effect of experimental therapies for T1DM on Treg cell number and function in both mice and humans.
Collapse
Affiliation(s)
- Susanne M. Cabrera
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Mark R. Rigby
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Raghavendra G. Mirmira
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
38
|
Abstract
A classic understanding of the interplay between B and T cell components of the immune system that drive autoimmunity, where B cells provide an effector function, is represented by systemic lupus erythematosus (SLE), an autoimmune condition characterised by the production of auto-antibodies. In SLE, CD4+T cells provide cognate help to self-reactive B cells, which in turn produce pathogenic auto-antibodies (1). Thus, B cells act as effectors by producing auto-antibody aided by T cell help such that B and T cell interactions are unidirectional. However, this paradigm of B and T cell interactions is challenged by new clinical data demonstrating that B cell depletion is effective for T cell mediated autoimmune diseases including type I diabetes mellitus (T1D) (2), rheumatoid arthritis (3), and multiple sclerosis (4). These clinical data indicate a model whereby B cells can influence the developing autoimmune T cell response, and therefore act as effectors, in ways that extend beyond the production of autoantibody (5). In this review by largely focusing on type I diabetes we will develop a hypothesis that bi-directional B and T interactions control the course of autoimmunity.
Collapse
Affiliation(s)
- Eliana Mariño
- Centre of Immunology and Inflammation, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia
| | | |
Collapse
|
39
|
Abstract
Type 1 diabetes is an autoimmune disease that gradually destructs insulin-producing beta cells. Over the years, clinicians' knowledge regarding the immunopathogenesis of this disease has greatly increased. Immunotherapies that can change the course of immune-mediated destruction and preserve and possibly regenerate the pancreatic beta cells seem to be promising in preclinical trials but so far have been unsuccessful in human studies. This article reviews the important immune interventions for type 1 diabetes that have been tried so far targeting the different stages of disease development and provides an insight into what the future might hold.
Collapse
Affiliation(s)
- Smita Gupta
- Diabetes and Endocrinology Consultants, 8435 Clearvista Place, Suite 101 Indianapolis, IN 46256, USA.
| |
Collapse
|
40
|
Tooley JE, Waldron-Lynch F, Herold KC. New and future immunomodulatory therapy in type 1 diabetes. Trends Mol Med 2012; 18:173-81. [PMID: 22342807 DOI: 10.1016/j.molmed.2012.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 01/01/2012] [Accepted: 01/02/2012] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes is a common autoimmune disease that affects millions of people worldwide and has an incidence that is increasing at a striking rate, especially in young children. It results from the targeted self-destruction of the insulin-secreting β cells of the pancreas and requires lifelong insulin treatment. The effects of chronic hyperglycemia - the result of insulin deficiency - include secondary endorgan complications. Over the past two decades our increased understanding of the pathogenesis of this disease has led to the development of new immunomodulatory treatments. None have yet received regulatory approval, but this report highlights recent progress in this area.
Collapse
Affiliation(s)
- James E Tooley
- Department of Immunobiology, Yale University, New Haven, CT 06511, USA
| | | | | |
Collapse
|
41
|
Guan Y, Zhang M, Li Y, Cao W, Ji M, Liu Y. Vaccination with IA-2 autoantigen can prevent late prediabetic nonobese diabetic mice from developing diabetes mellitus. Diabetes Res Clin Pract 2012; 95:93-7. [PMID: 22004942 DOI: 10.1016/j.diabres.2011.09.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Revised: 09/15/2011] [Accepted: 09/20/2011] [Indexed: 11/23/2022]
Abstract
DNA vaccine can be applied to deliver genes into cells to induce tolerance of some autoimmune diseases. This study aimed to evaluate the effect of DNA vaccine in preventing late prediabetic nonobese diabetic (NOD) mice from developing autoimmune diabetes mellitus. The cDNA of human IA2 was recombined to be used as the DNA vaccine. Plasmid IL-4/MCP-1 was co-administrated as the DNA adjuvant. 49 10-11-week-old NOD mice were grouped into four groups: the control group (n=10), IA-2 vaccine group (n=17), IL-4/MCP-1 vaccine group (n=8) and IA-2 plus IL-4/MCP-1 vaccine group (n=14) by intramuscularly injected with 50 μg plasmid in each quadriceps muscle. Glucose levels in the groups were detected every 1-2 weeks. Insulitis was evaluated with hematoxylin and eosin-stained pancreatic sections. CD4(+) CD25(+)and CD8(+) T lymphocytes were measured with flow cytometry. The results showed that in 10-11-week-old female NOD mice, vaccination with IA2 or in combination with IL-4/MCP-1 delayed the onset of disease compared with the control group (p<0.05). Our results suggest that the DNA vaccine IA2 can prevent NOD mouse from developing autoimmune diabetes and this efficiency is related to the immune status of the recipients. Our findings offer new insights into the immune system and suggest novel methods of type 1 diabetes prevention.
Collapse
Affiliation(s)
- Yueyan Guan
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun 130041, China
| | | | | | | | | | | |
Collapse
|
42
|
Hinke SA. Inverse vaccination with islet autoantigens to halt progression of autoimmune diabetes. Drug Dev Res 2011. [DOI: 10.1002/ddr.20488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
43
|
Tian J, Dang H, Kaufman DL. Combining antigen-based therapy with GABA treatment synergistically prolongs survival of transplanted ß-cells in diabetic NOD mice. PLoS One 2011; 6:e25337. [PMID: 21966502 PMCID: PMC3178649 DOI: 10.1371/journal.pone.0025337] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 09/01/2011] [Indexed: 01/13/2023] Open
Abstract
Antigen-based therapies (ABTs) very effectively prevent the development of type 1 diabetes (T1D) when given to young nonobese diabetic (NOD) mice, however, they have little or no ability to reverse hyperglycemia in newly diabetic NOD mice. More importantly, ABTs have not yet demonstrated an ability to effectively preserve residual ß-cells in individuals newly diagnosed with type 1 diabetes (T1D). Accordingly, there is great interest in identifying new treatments that can be combined with ABTs to safely protect ß-cells in diabetic animals. The activation of γ-aminobutyric acid (GABA) receptors (GABA-Rs) on immune cells has been shown to prevent T1D, experimental autoimmune encephalomyelitis (EAE) and rheumatoid arthritis in mouse models. Based on GABA's ability to inhibit different autoimmune diseases and its safety profile, we tested whether the combination of ABT with GABA treatment could prolong the survival of transplanted ß-cells in newly diabetic NOD mice. Newly diabetic NOD mice were untreated, or given GAD/alum (20 or 100 µg) and placed on plain drinking water, or water containing GABA (2 or 6 mg/ml). Twenty-eight days later, they received syngenic pancreas grafts and were monitored for the recurrence of hyperglycemia. Hyperglycemia reoccurred in the recipients given plain water, GAD monotherapy, GABA monotherapy, GAD (20 µg)+GABA (2 mg/ml), GAD (20 µg)+GABA (6 mg/ml) and GAD (100 µg)+GABA (6 mg/ml) about 1, 2-3, 3, 2-3, 3-8 and 10-11 weeks post-transplantation, respectively. Thus, combined GABA and ABT treatment had a synergistic effect in a dose-dependent fashion. These findings suggest that co-treatment with GABA (or other GABA-R agonists) may provide a new strategy to safely enhance the efficacy of other therapeutics designed to prevent or reverse T1D, as well as other T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Jide Tian
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hoa Dang
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Daniel L. Kaufman
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
Tian J, Yong J, Dang H, Kaufman DL. Oral GABA treatment downregulates inflammatory responses in a mouse model of rheumatoid arthritis. Autoimmunity 2011; 44:465-70. [PMID: 21604972 PMCID: PMC5787624 DOI: 10.3109/08916934.2011.571223] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Current treatments for rheumatoid arthritis (RA) have long-term side effects such that new treatments are needed that can safely help manage the disease. There is a growing appreciation that GABA receptors (GABA-Rs) on immune cells provide new targets that can be used to modulate immune cell activity. Here, we show for the first time that activation of peripheral GABA-Rs can inhibit the development of disease in the collagen-induced arthritis (CIA) mouse model of RA. Mice that received oral GABA had a reduced incidence of CIA, and those mice that did develop CIA had milder symptoms. T cells from GABA-treated mice displayed reduced proliferative responses to collagen and their APC had a reduced ability to promote the proliferation of collagen-reactive T cells. Thus, GABA downregulated both T-cell autoimmunity and APC activity. Collagen-reactive T cells from GABA-treated mice displayed reduced recall responses in the presence of GABA ex vivo, indicating that GABA consumption did not desensitize these cells to GABA. GABA-treated mice had reduced collagen-reactive IgG2a, but not IgG1 antibodies, consistent with reduced Th1 help. The levels of serum anti-collagen IgG2a antibodies were correlated significantly with the CIA disease scores of individual mice. Our results suggest that activation of peripheral GABA-Rs may provide a new modality to modulate T cell, B cell, and APC activity and help ameliorate RA and other inflammatory diseases.
Collapse
Affiliation(s)
- Jide Tian
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095-1735, USA
| | | | | | | |
Collapse
|
45
|
Wherrett DK, Bundy B, Becker DJ, DiMeglio LA, Gitelman SE, Goland R, Gottlieb PA, Greenbaum CJ, Herold KC, Marks JB, Monzavi R, Moran A, Orban T, Palmer JP, Raskin P, Rodriguez H, Schatz D, Wilson DM, Krischer JP, Skyler JS, Type 1 Diabetes TrialNet GAD Study Group. Antigen-based therapy with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset type 1 diabetes: a randomised double-blind trial. Lancet 2011; 378:319-27. [PMID: 21714999 PMCID: PMC3580128 DOI: 10.1016/s0140-6736(11)60895-7] [Citation(s) in RCA: 286] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Glutamic acid decarboxylase (GAD) is a major target of the autoimmune response that occurs in type 1 diabetes mellitus. In animal models of autoimmunity, treatment with a target antigen can modulate aggressive autoimmunity. We aimed to assess whether immunisation with GAD formulated with aluminum hydroxide (GAD-alum) would preserve insulin production in recent-onset type 1 diabetes. METHODS Patients aged 3-45 years who had been diagnosed with type 1 diabetes for less than 100 days were enrolled from 15 sites in the USA and Canada, and randomly assigned to receive one of three treatments: three injections of 20 μg GAD-alum, two injections of 20 μg GAD-alum and one of alum, or 3 injections of alum. Injections were given subcutaneously at baseline, 4 weeks later, and 8 weeks after the second injection. The randomisation sequence was computer generated at the TrialNet coordinating centre. Patients and study personnel were masked to treatment assignment. The primary outcome was the baseline-adjusted geometric mean area under the curve (AUC) of serum C-peptide during the first 2 h of a 4-h mixed meal tolerance test at 1 year. Secondary outcomes included changes in glycated haemoglobin A(1c) (HbA(1c)) and insulin dose, and safety. Analysis included all randomised patients with known measurements. This trial is registered with ClinicalTrials.gov, number NCT00529399. FINDINGS 145 patients were enrolled and treated with GAD-alum (n=48), GAD-alum plus alum (n=49), or alum (n=48). At 1 year, the 2-h AUC of C-peptide, adjusted for age, sex, and baseline C-peptide value, was 0·412 nmol/L (95% CI 0·349-0·478) in the GAD-alum group, 0·382 nmol/L (0·322-0·446) in the GAD-alum plus alum group, and 0·413 nmol/L (0·351-0·477) in the alum group. The ratio of the population mean of the adjusted geometric mean 2-h AUC of C-peptide was 0·998 (95% CI 0·779-1·22; p=0·98) for GAD-alum versus alum, and 0·926 (0·720-1·13; p=0·50) for GAD-alum plus alum versus alum. HbA(1c), insulin use, and the occurrence and severity of adverse events did not differ between groups. INTERPRETATION Antigen-based immunotherapy therapy with two or three doses of subcutaneous GAD-alum across 4-12 weeks does not alter the course of loss of insulin secretion during 1 year in patients with recently diagnosed type 1 diabetes. Although antigen-based therapy is a highly desirable treatment and is effective in animal models, translation to human autoimmune disease remains a challenge. FUNDING US National Institutes of Health.
Collapse
Affiliation(s)
- Diane K Wherrett
- Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
Collaborators
Jay S Skyler, Mark Anderson, Katarzyna Bourcier, Dorothy Becker, Penelope Bingley, Janice Blum, Emanuele Bosi, Jane Buckner, H Peter Chase, Michael Clare-Salzler, Peter Colman, Linda DiMeglio, George S Eisenbarth, C Garrison Fathman, Stephen Gitelman, Robin Goland, Peter Gottlieb, Gilman Grave, Carla Greenbaum, Leonard Harrison, Kevan Herold, Richard Insel, Jeffrey P Krischer, Jeffrey Mahon, Jennifer Marks, Antoinette Moran, Jerry P Palmer, Mark Peakman, Alberto Pugliese, Philip Raskin, Maria Grazia Roncarolo, William Russell, Peter Savage, Desmond Schatz, Robert Sherwin, Mark Siegelman, Olli Simell, James Thomas, Massimo Trucco, John Wagner, Diane Wherrett, Darrell M Wilson, William Winter, Judith Fradkin, Ellen Leschek, Lisa Spain, Christophe Benoist, Jeffrey Bluestone, David Brown, Catherine Cowie, Bernard Hering, Stanley Jordan, Francine R Kaufman, John M Lachin, Kirsti Nanto-Salonen, Gerald Nepom, Tihamer Orban, Robertson Parkman, Mark Pescovitz, John Peyman, John Ridge, Henry Rodriguez, Anette Ziegler, Jay S Skyler, Katarzyna Bourcier, Carla J Greenbaum, Jeffrey P Krischer, Ellen Leschek, Lisa Rafkin, Peter Savage, Lisa Spain, Catherine Cowie, Mary Foulkes, Heidi Krause-Steinrauf, John M Lachin, Saul Malozowski, John Peyman, John Ridge, Stephanie J Zafonte, Jay S Skyler, Carla J Greenbaum, Norma S Kenyon, Lisa Rafkin, Irene Santiago, Jay M Sosenko, Jeffrey P Krischer, Brian Bundy, Joy Ramiro, Michael Abbondondolo, Timothy Adams, Persida Alies, Franz Badias, Craig Beam, Matthew Boonstra, David Boulware, David Cuthbertson, Christopher Eberhard, Julie Ford, Jinin Ginem, Heather Guillette, Brian Hays, Martha Henry, Pat Law, Cristin Linton, Shu Liu, Jennifer Lloyd, Aqesha Luvon Ritzie, Sarah Muller, Ryan O'Donnell, Yazandra Parrimon, Kate Paulus, Jennifer Pilger, Amy Roberts, Kelly Sadler, Amanda Terry, Margaret Wootten, Ping Xu, Kenneth Young, Monica Bassi, Doug Freeman, Moriah Granger, Michelle Kieffer, Lavanya Nallamshetty, Audrey Shor, John M Lachin, Mary Foulkes, Pamela Harding, Heidi Krause-Steinrauf, Susan McDonough, Paula F McGee, Kimberly Owens Hess, Donna Phoebus, Scott Quinlan, Erica Raiden, Judith Fradkin, Ellen Leschek, Peter Savage, Lisa Spain, Emily Blumberg, Jonathan Braun, Lori Laffel, Ali Naji, Jorn Nerup, Trevor Orchard, Anastasios Tsiatis, Robert Veatch, Dennis Wallace, Ake Lernmark, Bernard Lo, Herman Mitchell, Michael Steffes, Bernard Zinman, Brett Loechelt, Lindsey Baden, Michael Green, Adriana Weinberg, Sunanda Babu, George S Eisenbarth, Santica Marcovina, Jerry P Palmer, Adriana Weinberg, William Winter, Liping Yu, Diane Wherett, Penelope Bingley, Roberta Cook, Jeffrey Mahon, Jerry Palmer, Carla Greenbaum, Jeffrey Krischer, Ellen Leschek, Brett Loechelt, Lisa Rafkin, Jay S Skyler, Audrey Shor, Lisa Spain, John M Lachin, Heidi Krause-Steinrauf, Paula F McGee, Carla Greenbaum, Jennifer Bollyky, Srinath Sanda, David Tridgell, Marli McCulloch-Olson, Heather Vendettuoli, Deborah Hefty, Mary Ramey, Christine Webber, Kristen Kuhns, Nicole Hilderman, Angela Dove, Marissa Hammond, Jani Klein, Emily Batts, Roshanak Monzavi, Mary Halvorson, Meredith Bock, Lynda Fisher, Debra Jeandron, Jamie Wood, Robin Goland, Ellen Greenberg, Mary Pat Gallagher, Jeniece Trast, Mary Chan, Kelly Smith, Henry Rodriguez, Mark Pescovitz, Linda DiMeglio, Sheryl Lynch, Maria Nicholson, Martha Mendez, Jennifer Terrell, Tihamer Orban, Joseph Wolfsdorf, Alyne Ricker, Heyam Jalahej, Debbie Conboy, Hui Zhang, Steve Fay, Darrell M Wilson, Bruce A Buckingham, Tandy Aye, Trudy Esrey, Adriana Soto, Jennifer Perry, Bonita Baker, Barbara Berry, Stephen E Gitelman, Stephen M Rosenthal, Mark Anderson, Saleh Adi, Kathleen Breen, Celia Hamilton, Peter Gottlieb, Aaron Michaels, H Peter Chase, Susan Ulmer, Sandy Barry, Lisa Meyers, Debbie Lehr, Libbie Tuthill, Laurie Weiner, Desmond Schatz, Michael Haller, Michael Clare-Salzler, Roberta Cook, Diane Mancini, Annie Abraham, Elena Hicks, Gloria Cole, Jennifer B Marks, Alberto Pugliese, Della Matheson, Carlos Blaschke, Luz Arazo, Mario Cisneros, Brenda Acosta, Antoinette Moran, Brandon Nathan, John Wagner, Theresa Albright-Fischer, Jennifer Smith, Anne Street, Janice Leschyshyn, Chris Kwong, Dorothy Becker, Frederico Toledo, Ingrid Libman, Karen Riley, Kelli Delallo, Kym Smith, Diane Gwynn, Gyna Wohlers, Nursen Gurtunca, Philip Raskin, Perrin White, Bryan Dickson, Soumya Adhikari, Mark Siegelman, Marilyn Alford, Nenita Torres, Tauri Harden, Lourdes Pruneda, Erica Cordova, Renee Davis, Stefani Fernandez, Jamie Arthur, Diane Wherrett, Roze Kovalakovska, Lesley A Eisel, Brenda Ahenkorah, Mithula Sriskandarajah, Mary-Jo Ricci, Kevan C Herold, Laurie Feldman, Robert Sherwin,
Collapse
|
46
|
Current state of type 1 diabetes immunotherapy: incremental advances, huge leaps, or more of the same? Clin Dev Immunol 2011; 2011:432016. [PMID: 21785616 PMCID: PMC3139873 DOI: 10.1155/2011/432016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 04/28/2011] [Indexed: 01/09/2023]
Abstract
Thus far, none of the preclinically successful and promising immunomodulatory agents for type 1 diabetes mellitus (T1DM) has conferred stable, long-term insulin independence to diabetic patients. The majority of these immunomodulators are humanised antibodies that target immune cells or cytokines. These as well as fusion proteins and inhibitor proteins all share varying adverse event occurrence and severity. Other approaches have included intact putative autoantigens or autoantigen peptides. Considerable logistical outlays have been deployed to develop and to translate humanised antibodies targeting immune cells, cytokines, and cytokine receptors to the clinic. Very recent phase III trials with the leading agent, a humanised anti-CD3 antibody, call into question whether further development of these biologics represents a step forward or more of the same. Combination therapies of one or more of these humanised antibodies are also being considered, and they face identical, if not more serious, impediments and safety issues. This paper will highlight the preclinical successes and the excitement generated by phase II trials while offering alternative possibilities and new translational avenues that can be explored given the very recent disappointment in leading agents in more advanced clinical trials.
Collapse
|
47
|
Abstract
The clinical onset of type 1 diabetes or autoimmune diabetes occurs after a prodrome of islet autoimmunity. The warning signals for the ensuing loss of pancreatic islet beta cells are autoantibodies against insulin, GAD65, IA-2 and ZnT8, alone or in combinations. Autoantibodies against, for example, insulin alone have only a minor risk of type 1 diabetes. However, progression to clinical onset is increased by the induction of multiple islet autoantibodies. At the time of clinical onset, insulitis may be manifest, which seems to reduce the efficacy of immunosuppression. Autoantigen-specific immunotherapy with alum-formulated GAD65 (Diamyd(®)) shows promise to reduce the loss of beta-cell function after the clinical onset of type 1 diabetes. The mechanisms are unclear but may involve the induction of T regulatory cells, which may suppress islet autoantigen reactivity. Past and ongoing clinical trials have been safe. Future clinical trials, perhaps as combination autoantigen-specific immunotherapy, may increase the efficacy in preventing the clinical onset in subjects with islet autoantibodies or preserve residual beta-cell function in patients newly diagnosed with type 1 diabetes.
Collapse
Affiliation(s)
- Helena Elding Larsson
- Department of Clinical Sciences, Lund University/CRC, Skåne Uni0076ersity Hospital SUS, SE-20502 Malmö, Sweden
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/CRC, Skåne Uni0076ersity Hospital SUS, SE-20502 Malmö, Sweden
| |
Collapse
|
48
|
Affiliation(s)
- F Susan Wong
- Center for Diabetes and Endocrine Sciences, Cardiff UniversitySchool of Medicine, Cardiff, U.K.
| |
Collapse
|
49
|
Ludvigsson J, Hjorth M, Chéramy M, Axelsson S, Pihl M, Forsander G, Nilsson NÖ, Samuelsson BO, Wood T, Aman J, Ortqvist E, Casas R. Extended evaluation of the safety and efficacy of GAD treatment of children and adolescents with recent-onset type 1 diabetes: a randomised controlled trial. Diabetologia 2011; 54:634-40. [PMID: 21116604 DOI: 10.1007/s00125-010-1988-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 10/29/2010] [Indexed: 01/12/2023]
Abstract
AIMS/HYPOTHESIS The aim of this study was to investigate the safety and efficacy of alum formulated glutamic acid decarboxylase GAD(65) (GAD-alum) treatment of children and adolescents with type 1 diabetes after 4 years of follow-up. METHODS Seventy children and adolescents aged 10-18 years with recent onset type 1 diabetes participated in a phase II, double-blind, randomised placebo-controlled clinical trial. Patients identified as possible participants attended one of eight clinics in Sweden to receive information about the study and for an eligibility check, including a medical history. Participants were randomised to one of the two treatment groups and received either a subcutaneous injection of 20 μg of GAD-alum or placebo at baseline and 1 month later. The study was blinded to participants and investigators until month 30. The study was unblinded at 15 months to the sponsor and statistician in order to evaluate the data. At follow-up after 30 months there was a significant preservation of residual insulin secretion, as measured by C-peptide, in the group receiving GAD-alum compared with placebo. This was particularly evident in patients with <6 months disease duration at baseline. There were no treatment-related serious adverse events. We have now followed these patients for 4 years. Overall, 59 patients, 29 who had been treated with GAD-alum and 30 who had received placebo, gave their informed consent. RESULTS One patient in each treatment group experienced an episode of keto-acidosis between months 30 and 48. There were no treatment-related adverse events. The primary efficacy endpoint was the change in fasting C-peptide concentration from baseline to 15 months after the prime injection for all participants per protocol set. In the GAD-alum group fasting C-peptide was 0.332 ± 0.032 nmol/l at day 1 and 0.215 ± 0.031 nmol/l at month 15. The corresponding figures for the placebo group were 0.354 ± 0.039 and 0.184 ± 0.033 nmol/l, respectively. The decline in fasting C-peptide levels between day 1 and month 1, was smaller in the GAD-alum group than the placebo group. The difference between the treatment groups was not statistically significant. In those patients who were treated within 6 months of diabetes diagnosis, fasting C-peptide had decreased significantly less in the GAD-alum group than in the placebo-treated group after 4 years. CONCLUSION/INTERPRETATION Four years after treatment with GAD-alum, children and adolescents with recent-onset type 1 diabetes continue to show no adverse events and possibly to show clinically relevant preservation of C-peptide. TRIAL REGISTRATION ClinicalTrials.gov NCT00435981 FUNDING The study was funded by The Swedish Research Council K2008-55X-20652-01-3, Barndiabetesfonden (The Swedish Child Diabetes Foundation), the Research Council of Southeast Sweden, and an unrestricted grant from Diamyd Medical AB.
Collapse
Affiliation(s)
- J Ludvigsson
- Division of Pediatrics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, SE-58185, Linköping, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rivas EI, Driver JP, Garabatos N, Presa M, Mora C, Rodriguez F, Serreze DV, Stratmann T. Targeting of a T cell agonist peptide to lysosomes by DNA vaccination induces tolerance in the nonobese diabetic mouse. THE JOURNAL OF IMMUNOLOGY 2011; 186:4078-87. [PMID: 21346228 DOI: 10.4049/jimmunol.0902395] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD4 T cells are crucial effectors in the pathology of type 1 diabetes (T1D). Successful therapeutic interventions for prevention and cure of T1D in humans are still elusive. Recent research efforts have focused on the manipulation of T cells by treatment with DNA. In this paper, we studied the effects of a DNA treatment strategy designed to target antigenic peptides to the lysosomal compartment on a monospecific T cell population termed 2.5mi(+) T cells that shares reactivity with the diabetogenic T cell clone BDC-2.5 in the NOD mouse. MHC class II tetramer analysis showed that repeated administrations were necessary to expand 2.5mi(+) T cells in vivo. This expansion was independent of Ag presentation by B cells. A single peptide epitope was sufficient to induce protection against T1D, which was not due to Ag-specific T cell anergy. Typical Th2 cytokines such as IL-10 or IL-4 were undetectable in 2.5mi(+) T cells, arguing against a mechanism of immune deviation. Instead, the expanded 2.5mi(+) T cell population produced IFN-γ similar to 2.5mi(+) T cells from naive mice. Protection against T1D by DNA treatment was completely lost in NOD.CD28(-/-) mice which are largely deficient of natural regulatory T cells (Treg). Although Ag-specific Foxp3(+) Treg did not expand in response to DNA treatment, diabetes onset was delayed in Treg-reconstituted and DNA-treated NOD.SCID mice. These observations provide evidence for a Treg-mediated protective mechanism that is independent of the expansion or de novo generation of Ag-specific Treg.
Collapse
Affiliation(s)
- Elisa I Rivas
- Department of Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|