1
|
Wong JM, Pepper AR. Status of islet transplantation and innovations to sustainable outcomes: novel sites, cell sources, and drug delivery strategies. FRONTIERS IN TRANSPLANTATION 2024; 3:1485444. [PMID: 39553396 PMCID: PMC11565603 DOI: 10.3389/frtra.2024.1485444] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024]
Abstract
Islet transplantation (ITx) is an effective means to restore physiologic glycemic regulation in those living with type 1 diabetes; however, there are a handful of barriers that prevent the broad application of this functionally curative procedure. The restricted cell supply, requisite for life-long toxic immunosuppression, and significant immediate and gradual graft attrition limits the procedure to only those living with brittle diabetes. While intraportal ITx is the primary clinical site, portal vein-specific factors including low oxygen tension and the instant blood-mediated inflammatory reaction are detrimental to initial engraftment and long-term function. These factors among others prevent the procedure from granting recipients long-term insulin independence. Herein, we provide an overview of the status and limitations of ITx, and novel innovations that address the shortcomings presented. Despite the marked progress highlighted in the review from as early as the initial islet tissue transplantation in 1893, ongoing efforts to improve the procedure efficacy and success are also explored. Progress in identifying unlimited cell sources, more favourable transplant sites, and novel drug delivery strategies all work to broaden ITx application and reduce adverse outcomes. Exploring combination of these approaches may uncover synergies that can further advance the field of ITx in providing sustainable functional cures. Finally, the potential of biomaterial strategies to facilitate immune evasion and local immune modulation are featured and may underpin successful application in alternative transplant sites.
Collapse
Affiliation(s)
| | - Andrew R. Pepper
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
2
|
Chakraborty R, Mukherjee AK, Bala A. Breakthroughs in road mapping IL-35 mediated immunotherapy for type-1 and autoimmune diabetes mellitus. Cytokine 2024; 181:156692. [PMID: 38986251 DOI: 10.1016/j.cyto.2024.156692] [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: 04/05/2024] [Revised: 05/22/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
IL-35 is a recently discovered protein made up of IL-12α and IL-27β chains. It is encoded by IL12A and EBI3 genes. Interest in researching IL-35 has significantly increased in recent years, as evidenced by numerous scientific publications. Diabetes is on the rise globally, causing more illness and death in developing countries. The International Diabetes Federation (IDF) reports that diabetes is increasingly affecting children and teenagers, with varying rates across different regions. Therefore, scientists seek new diabetes treatments despite the growth of drug research. Recent research aims to emphasize IL-35 as a critical regulator of diabetes, especially type 1 and autoimmune diabetes. This review provides an overview of recent research on IL-35 and its link to diabetes and its associated complications. Studies suggest that IL-35 can offer protection against type-1 diabetes and autoimmune diabetes by regulating macrophage polarization, T-cell-related cytokines, and regulatory B cells (Bregs). This review will hopefully assist biomedical scientists in exploring the potential role of IL-35-mediated immunotherapy in treating diabetes. However, further research is necessary to determine the exact mechanism and plan clinical trials.
Collapse
Affiliation(s)
- Ratul Chakraborty
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati 781035, Assam, India; Academy of Scientific and Innovative Research (AcSIR), AcSIR (an Indian Institute of National Importance), Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201002, India
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati 781035, Assam, India
| | - Asis Bala
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Vigyan Path, Guwahati 781035, Assam, India; Academy of Scientific and Innovative Research (AcSIR), AcSIR (an Indian Institute of National Importance), Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
3
|
Saikia M, Lassi ZS, McCall AL. Editorial: Developing strategies to improve diabetes management in college-going young adults. Front Endocrinol (Lausanne) 2024; 15:1402133. [PMID: 38660515 PMCID: PMC11039824 DOI: 10.3389/fendo.2024.1402133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Affiliation(s)
- Mridusmita Saikia
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Zohra S. Lassi
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Anthony L. McCall
- University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
4
|
Jung S, Ben Nasr M, Bahmani B, Usuelli V, Zhao J, Sabiu G, Seelam AJ, Naini SM, Balasubramanian HB, Park Y, Li X, Khalefa SA, Kasinath V, Williams MD, Rachid O, Haik Y, Tsokos GC, Wasserfall CH, Atkinson MA, Bromberg JS, Tao W, Fiorina P, Abdi R. Nanotargeted Delivery of Immune Therapeutics in Type 1 Diabetes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300812. [PMID: 37357903 PMCID: PMC10629472 DOI: 10.1002/adma.202300812] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/14/2023] [Indexed: 06/27/2023]
Abstract
Immune therapeutics holds great promise in the treatment of type 1 diabetes (T1D). Nonetheless, their progress is hampered by limited efficacy, equipoise, or issues of safety. To address this, a novel and specific nanodelivery platform for T1D that targets high endothelial venules (HEVs) presented in the pancreatic lymph nodes (PLNs) and pancreas is developed. Data indicate that the pancreata of nonobese diabetic (NOD) mice and patients with T1D are unique in their expression of newly formed HEVs. Anti-CD3 mAb is encapsulated in poly(lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles (NPs), the surfaces of which are conjugated with MECA79 mAb that recognizes HEVs. Targeted delivery of these NPs improves accumulation of anti-CD3 mAb in both the PLNs and pancreata of NOD mice. Treatment of hyperglycemic NOD mice with MECA79-anti-CD3-NPs results in significant reversal of T1D compared to those that are untreated, treated with empty NPs, or provided free anti-CD3. This effect is associated with a significant reduction of T effector cell populations in the PLNs and a decreased production of pro-inflammatory cytokine in the mice treated with MECA79-anti-CD3-NPs. In summary, HEV-targeted therapeutics may be used as a means by which immune therapeutics can be delivered to PLNs and pancreata to suppress autoimmune diabetes effectively.
Collapse
Affiliation(s)
- Sungwook Jung
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Moufida Ben Nasr
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
| | - Baharak Bahmani
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Vera Usuelli
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
| | - Jing Zhao
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gianmarco Sabiu
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Andy Joe Seelam
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Said Movahedi Naini
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hari Baskar Balasubramanian
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
| | - Youngrong Park
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaofei Li
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Salma Ayman Khalefa
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
| | - Vivek Kasinath
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - MacKenzie D Williams
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Ousama Rachid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, 2713, Doha, Qatar
| | - Yousef Haik
- Department of Mechanical and Nuclear Engineering, University of Sharjah, 27272, Sharjah, UAE
| | - George C Tsokos
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
- Department of Pediatrics, University of Florida, Gainesville, FL, 32610, USA
| | - Jonathan S Bromberg
- Departments of Surgery and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wei Tao
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Paolo Fiorina
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, 20157, Milan, Italy
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Reza Abdi
- Transplantation Research Center and Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
5
|
Martin A, Mick GJ, Choat HM, Lunsford AA, Tse HM, McGwin GG, McCormick KL. A randomized trial of oral gamma aminobutyric acid (GABA) or the combination of GABA with glutamic acid decarboxylase (GAD) on pancreatic islet endocrine function in children with newly diagnosed type 1 diabetes. Nat Commun 2022; 13:7928. [PMID: 36566274 PMCID: PMC9790014 DOI: 10.1038/s41467-022-35544-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/06/2022] [Indexed: 12/25/2022] Open
Abstract
Gamma aminobutyric acid(GABA) is synthesized by glutamate decarboxylase(GAD) in β-cells. Regarding Type 1 diabetes(T1D), animal/islet-cell studies found that GABA promotes insulin secretion, inhibits α-cell glucagon and dampens immune inflammation, while GAD immunization may also preserve β-cells. We evaluated the safety and efficacy of oral GABA alone, or combination GABA with GAD, on the preservation of residual insulin secretion in recent-onset T1D. Herein we report a single-center, double-blind, one-year, randomized trial in 97 children conducted March 2015 to June 2019(NCT02002130). Using a 2:1 treatment:placebo ratio, interventions included oral GABA twice-daily(n = 41), or oral GABA plus two-doses GAD-alum(n = 25), versus placebo(n = 31). The primary outcome, preservation of fasting/meal-stimulated c-peptide, was not attained. Of the secondary outcomes, the combination GABA/GAD reduced fasting and meal-stimulated serum glucagon, while the safety/tolerability of GABA was confirmed. There were no clinically significant differences in glycemic control or diabetes antibody titers. Given the low GABA dose for this pediatric trial, future investigations using higher-dose or long-acting GABA formulations, either alone or with GAD-alum, could be considered, although GABA alone or in combination with GAD-alum did nor preserve beta-cell function in this trial.
Collapse
Affiliation(s)
- Alexandra Martin
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gail J Mick
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Heather M Choat
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alison A Lunsford
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gerald G McGwin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth L McCormick
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
6
|
Chen X, Affinati AH, Lee Y, Turcu AF, Henry NL, Schiopu E, Qin A, Othus M, Clauw D, Ramnath N, Zhao L. Immune Checkpoint Inhibitors and Risk of Type 1 Diabetes. Diabetes Care 2022; 45:1170-1176. [PMID: 35238899 PMCID: PMC9174959 DOI: 10.2337/dc21-2213] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/05/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Type 1 diabetes mellitus (T1DM) is a rare, irreversible immune-related adverse event reported in patients receiving treatment with immune checkpoint inhibitors (ICI). However, clinical risk factors for ICI-induced T1DM (ICI-T1DM) and its impact on survival in patients remain unknown. RESEARCH DESIGN AND METHODS We used Optum's Clinformatics Data Mart database for assessment of the incidence and characteristics of T1DM in a large de-identified cohort of patients treated with ICI between 2017 and 2020. We applied Fine-Gray and cause-specific hazard models to study associations between patient/treatment characteristics and ICI-T1DM and applied the Cox model with ICI-T1DM as a time-varying covariate to assess the impact of ICI-T1DM on survival. RESULTS ICI-T1DM was observed in 261 of 30,337 (0.86%) patients. Dual use of antibodies to cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed cell death 1 (PD-1) or programmed cell death ligand 1 (PD-L1) was associated with increasing risk of ICI-T1DM (hazard ratio [HR] 1.62; 95% CI 1.15-2.26) vs. anti-PD-L1 or anti-PD-1 alone. Younger age (HR 1.19 for every 5-year decrease; 95% CI 1.13-1.25) and preexisting non-T1DM diabetes (HR 4.48; 95% CI 3.45-5.83) were also associated with higher risk of ICI-T1DM. Conversely, prior use of immunosuppressive medications (HR 0.57; 95% CI 0.34-0.95) was associated with lower incidence of ICI-T1DM, but part of its protective effect may be due to the increased mortality rate. Development of ICI-T1DM does not seem to significantly impact patient survival. CONCLUSIONS The risk of ICI-T1DM is associated with the type of ICI therapy, patient age, and preexisting non-T1DM diabetes. These data may help guide risk assessment and screening practices for patients during ICI therapy.
Collapse
Affiliation(s)
- Xuan Chen
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Alison H. Affinati
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI
| | - Yungchun Lee
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Adina F. Turcu
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI
| | - Norah Lynn Henry
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Elena Schiopu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Angel Qin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Megan Othus
- Biostatistics and Biomathematics Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Ann Arbor, MI
| | - Dan Clauw
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI
| | - Nithya Ramnath
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
7
|
Xhonneux LP, Knight O, Lernmark Å, Bonifacio E, Hagopian WA, Rewers MJ, She JX, Toppari J, Parikh H, Smith KGC, Ziegler AG, Akolkar B, Krischer JP, McKinney EF. Transcriptional networks in at-risk individuals identify signatures of type 1 diabetes progression. Sci Transl Med 2021; 13:eabd5666. [PMID: 33790023 PMCID: PMC8447843 DOI: 10.1126/scitranslmed.abd5666] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/24/2020] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes (T1D) is a disease of insulin deficiency that results from autoimmune destruction of pancreatic islet β cells. The exact cause of T1D remains unknown, although asymptomatic islet autoimmunity lasting from weeks to years before diagnosis raises the possibility of intervention before the onset of clinical disease. The number, type, and titer of islet autoantibodies are associated with long-term disease risk but do not cause disease, and robust early predictors of individual progression to T1D onset remain elusive. The Environmental Determinants of Diabetes in the Young (TEDDY) consortium is a prospective cohort study aiming to determine genetic and environmental interactions causing T1D. Here, we analyzed longitudinal blood transcriptomes of 2013 samples from 400 individuals in the TEDDY study before both T1D and islet autoimmunity. We identified and interpreted age-associated gene expression changes in healthy infancy and age-independent changes tracking with progression to both T1D and islet autoimmunity, beginning before other evidence of islet autoimmunity was present. We combined multivariate longitudinal data in a Bayesian joint model to predict individual risk of T1D onset and validated the association of a natural killer cell signature with progression and the model's predictive performance on an additional 356 samples from 56 individuals in the independent Type 1 Diabetes Prediction and Prevention study. Together, our results indicate that T1D is characterized by early and longitudinal changes in gene expression, informing the immunopathology of disease progression and facilitating prediction of its course.
Collapse
Affiliation(s)
- Louis-Pascal Xhonneux
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Oliver Knight
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/CRC Skåne University Hospital Malmo, Jan Waldenströms gata 35, Malmö, Sweden
| | - Ezio Bonifacio
- Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - William A Hagopian
- Pacific Northwest Research Institute, 720 Broadway, Seattle, WA 98122, USA
| | - Marian J Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, 1775 Aurora Ct, Aurora, CO 80045, USA
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd., Augusta, GA 30912, USA
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, FI-20014 Turun Lyliopisto, Finland
| | - Hemang Parikh
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, and Klinikum rechts der Isar, Technische, Universität München, Forschergruppe Diabetes e.V., Arcisstraße 21, 80333 München, Germany
| | - Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, 9000 Rockville Pike Bethesda, MD 20892, USA
| | - Jeffrey P Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Eoin F McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
- Cambridge Centre for Artificial Intelligence in Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Effect of gamma aminobutyric acid (GABA) or GABA with glutamic acid decarboxylase (GAD) on the progression of type 1 diabetes mellitus in children: Trial design and methodology. Contemp Clin Trials 2019; 82:93-100. [DOI: 10.1016/j.cct.2019.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/06/2019] [Accepted: 06/19/2019] [Indexed: 11/18/2022]
|
9
|
Current practices and future outlook on the integration of biomarkers in the drug development process. Bioanalysis 2017; 9:1827-1837. [PMID: 29120222 DOI: 10.4155/bio-2017-0155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Over the last decade, there has been broad incorporation of translational biomarkers into the early drug development process to predict safety concerns, measure target engagement and monitor disease progression. One goal of translational biomarkers is to create a cycle whereby preclinical readouts influence candidate selection and subsequent clinical data are fed back into research to facilitate better decision making. Successes have been limited and not as broad in scope as desired. Collaborations between industry and regulators have increased the number of qualified biomarkers; but the process is lengthy and expensive. A high level overview of translational biomarkers as well as a discussion of some of the successes and failures encountered in development is discussed here.
Collapse
|
10
|
Lam HV, Nguyen DT, Nguyen CD. Sibling method increases risk assessment estimates for type 1 diabetes. PLoS One 2017; 12:e0176341. [PMID: 28510587 PMCID: PMC5433695 DOI: 10.1371/journal.pone.0176341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/10/2017] [Indexed: 12/29/2022] Open
Abstract
We presented a risk assessment model to distinguish between type 1 diabetes (T1D) affected and unaffected siblings using only three single nucleotide polymorphism (SNP) genotypes. In addition we calculated the heritability from genome-wide identity-by-descent (IBD) sharing between full siblings. We analyzed 1,253 pairs of affected individuals and their unaffected siblings (750 pairs from a discovery set and 503 pairs from a validation set) from the T1D Genetics Consortium (T1DGC), applying a logistic regression to analyze the area under the receiver operator characteristic (ROC) curve (AUC). To calculate the heritability of T1D we used the Haseman-Elston regression analysis of the squared difference between the phenotypes of the pairs of siblings on the estimate of their genome-wide IBD proportion. The model with only 3 SNPs achieving an AUC of 0.75 in both datasets outperformed the model using the presence of the high-risk DR3/4 HLA genotype, namely AUC of 0.60. The heritability on the liability scale of T1D was approximately from 0.53 to 0.92, close to the results obtained from twin studies, ranging from 0.4 to 0.88.
Collapse
Affiliation(s)
- Hoang V. Lam
- Department of Endocrinology, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Dat T. Nguyen
- Department of Science and Technology, Hoa Sen University, Ho Chi Minh City, Vietnam
| | - Cao D. Nguyen
- Department of Business and Information System, Economics University, Ho Chi Minh City, Vietnam
- Clinical Analysis and Modelling—Department of Health, Western Australia, Australia
- * E-mail:
| |
Collapse
|
11
|
Abstract
Treatments for autoimmune diseases including type 1 diabetes (T1D) are aimed at resetting the immune system, especially its adaptive arm. The innate immune system is often ignored in the design of novel immune-based therapies. There is increasing evidence for multiple natural killer (NK) subpopulations, but their role is poorly understood in autoimmunity and likely is contributing to the controversial role reported for NKs. In this review, we will summarize NK subsets and their roles in tolerance, autoimmune diabetes, and immunotherapy.
Collapse
Affiliation(s)
- Chris Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Allison L Bayer
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
12
|
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
|
13
|
Abdi R, Moore R, Sakai S, Donnelly CB, Mounayar M, Sackstein R. HCELL Expression on Murine MSC Licenses Pancreatotropism and Confers Durable Reversal of Autoimmune Diabetes in NOD Mice. Stem Cells 2015; 33:1523-31. [PMID: 25641589 PMCID: PMC4447299 DOI: 10.1002/stem.1948] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/18/2014] [Indexed: 12/19/2022]
Abstract
Type 1 diabetes (T1D) is an immune-mediated disease resulting in destruction of insulin-producing pancreatic beta cells. Mesenchymal stem cells (MSCs) possess potent immunomodulatory properties, garnering increasing attention as cellular therapy for T1D and other immunologic diseases. However, MSCs generally lack homing molecules, hindering their colonization at inflammatory sites following intravenous (IV) administration. Here, we analyzed whether enforced E-selectin ligand expression on murine MSCs could impact their effect in reversing hyperglycemia in nonobese diabetic (NOD) mice. Although murine MSCs natively do not express the E-selectin-binding determinant sialyl Lewis(x) (sLe(x) ), we found that fucosyltransferase-mediated α(1,3)-exofucosylation of murine MSCs resulted in sLe(x) display uniquely on cell surface CD44 thereby creating hematopoietic cell E-/L-selectin ligand (HCELL), the E-selectin-binding glycoform of CD44. Following IV infusion into diabetic NOD mice, allogeneic HCELL(+) MSCs showed threefold greater peri-islet infiltrates compared to buffer-treated (i.e., HCELL(-) ) MSCs, with distribution in proximity to E-selectin-expressing microvessels. Exofucosylation had no effect on MSC immunosuppressive capacity in in vitro assays; however, although engraftment was temporary for both HCELL(+) and HCELL(-) MSCs, administration of HCELL(+) MSCs resulted in durable reversal of hyperglycemia, whereas only transient reversal was observed following administration of HCELL(-) MSCs. Notably, exofucosylation of MSCs generated from CD44(-/-) mice induced prominent membrane expression of sLe(x) , but IV administration of these MSCs into hyperglycemic NOD mice showed no enhanced pancreatotropism or reversal of hyperglycemia. These findings provide evidence that glycan engineering to enforce HCELL expression boosts trafficking of infused MSCs to pancreatic islets of NOD mice and substantially improves their efficacy in reversing autoimmune diabetes. Stem Cells 2013;33:1523-1531.
Collapse
Affiliation(s)
- Reza Abdi
- Department of Medicine, Renal Division & Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Robert Moore
- Department of Medicine, Renal Division & Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Shinobu Sakai
- Department of Dermatology, Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115
| | - Conor B. Donnelly
- Department of Dermatology, Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115
| | - Marwan Mounayar
- Department of Medicine, Renal Division & Transplantation Research Center, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Robert Sackstein
- Department of Dermatology, Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA 02115
- Department of Medicine, Brigham and Women’s Hospital, Boston, Harvard Medical School, MA 02115
| |
Collapse
|
14
|
Pagni PP, Bresson D, Rodriguez-Calvo T, Bel Hani A, Manenkova Y, Amirian N, Blaszczak A, Faton S, Sachithanantham S, von Herrath MG. Combination therapy with an anti-IL-1β antibody and GAD65 DNA vaccine can reverse recent-onset diabetes in the RIP-GP mouse model. Diabetes 2014; 63:2015-25. [PMID: 24520125 PMCID: PMC4030110 DOI: 10.2337/db13-1257] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Type 1 diabetes is thought to be an autoimmune condition in which self-reactive T cells attack insulin-secreting pancreatic β-cells. As a proinflammatory cytokine produced by β-cells or macrophages, interleukin-1β (IL-1β) represents a potential therapeutic target in diabetes. We reasoned IL-1β blockade could be combined with islet antigen-specific approaches involving GAD of 65 kDa (GAD65)-expressing plasmids, as previously shown in combination therapies (CTs) with anti-CD3. Thus, we investigated whether anti-IL-1β antibody alone or combined with GAD65 vaccine could reverse diabetes development in a virus-induced mouse model. Given alone, anti-IL-1β had no effect on diabetes, while GAD65 plasmid resulted in 33% disease reversal after a 5-week observation. However, CTs cured 53% of animals and prevented worsening of glycemic control in nonprotected individuals for up to 12 weeks. While the GAD65 vaccine arm of the CT was associated with increased forkhead box p3(+) regulatory T-cell frequency in pancreatic lymph nodes, islet infiltration by CD11b(+/high) cells was less frequent upon CT, and its extent correlated with treatment success or failure. Altogether, our CTs provided prolonged improvement of clinical and immunological features. Despite unsuccessful clinical trials using anti-IL-1β monotherapy, these data hold promise for treatment of type 1 diabetic patients with IL-1β blockade combined with antigen-specific vaccines.
Collapse
Affiliation(s)
- Philippe P Pagni
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Damien Bresson
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | | | - Amira Bel Hani
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Yulia Manenkova
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Natalie Amirian
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Alecia Blaszczak
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Sina Faton
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | | | | |
Collapse
|
15
|
Preventive effects of andrographolide on the development of diabetes in autoimmune diabetic NOD mice by inducing immune tolerance. Int Immunopharmacol 2013; 16:451-6. [DOI: 10.1016/j.intimp.2013.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 05/01/2013] [Accepted: 05/01/2013] [Indexed: 01/26/2023]
|
16
|
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
|
17
|
Giannoukakis N, Trucco M. Dendritic cell therapy for Type 1 diabetes suppression. Immunotherapy 2013; 4:1063-74. [PMID: 23148758 DOI: 10.2217/imt.12.76] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
While dendritic cell-based therapy is a clinical reality for human malignancies, until now, some conceptual concerns have served to delay its consideration to treat human autoimmune diseases, even in light of almost two decades' worth of overwhelmingly supportive preclinical animal studies. This article provides an overview of the development of dendritic cell-based therapy for Type 1 diabetes mellitus, given that this is the best-studied autoimmune disorder and that there is a good understanding of the underlying immunology. This article also highlights data from the authors' pioneering Phase I clinical trial with tolerogenic dendritic cells, which hopes to motivate the clinical translation of other dendritic cell-based approaches, to one or more carefully selected Type 1 diabetic patient populations.
Collapse
Affiliation(s)
- Nick Giannoukakis
- Department of Pathology, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, Rangos Research Center, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | | |
Collapse
|
18
|
Diabetes Mellitus: New Challenges and Innovative Therapies. NEW STRATEGIES TO ADVANCE PRE/DIABETES CARE: INTEGRATIVE APPROACH BY PPPM 2013; 3. [PMCID: PMC7120768 DOI: 10.1007/978-94-007-5971-8_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Diabetes is a common chronic disease affecting an estimated 285 million adults worldwide. The rising incidence of diabetes, metabolic syndrome, and subsequent vascular diseases is a major public health problem in industrialized countries. This chapter summarizes current pharmacological approaches to treat diabetes mellitus and focuses on novel therapies for diabetes mellitus that are under development. There is great potential for developing a new generation of therapeutics that offer better control of diabetes, its co-morbidities and its complications. Preclinical results are discussed for new approaches including AMPK activation, the FGF21 target, cell therapy approaches, adiponectin mimetics and novel insulin formulations. Gene-based therapies are among the most promising emerging alternatives to conventional treatments. Therapies based on gene silencing using vector systems to deliver interference RNA to cells (i.e. against VEGF in diabetic retinopathy) are also a promising therapeutic option for the treatment of several diabetic complications. In conclusion, treatment of diabetes faces now a new era that is characterized by a variety of innovative therapeutic approaches that will improve quality of life in the near future.
Collapse
|
19
|
IL-7 receptor α blockade, an off-switch for autoreactive T cells. Proc Natl Acad Sci U S A 2012; 109:12270-1. [PMID: 22826232 DOI: 10.1073/pnas.1209749109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
20
|
Azzi J, Moore RF, Elyaman W, Mounayar M, El Haddad N, Yang S, Jurewicz M, Takakura A, Petrelli A, Fiorina P, Ruckle T, Abdi R. The novel therapeutic effect of phosphoinositide 3-kinase-γ inhibitor AS605240 in autoimmune diabetes. Diabetes 2012; 61:1509-18. [PMID: 22403300 PMCID: PMC3357271 DOI: 10.2337/db11-0134] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes (T1D) remains a major health problem worldwide, with a steadily rising incidence yet no cure. Phosphoinositide 3-kinase-γ (PI3Kγ), a member of a family of lipid kinases expressed primarily in leukocytes, has been the subject of substantial research for its role in inflammatory diseases. However, the role of PI3Kγ inhibition in suppressing autoimmune T1D remains to be explored. We tested the role of the PI3Kγ inhibitor AS605240 in preventing and reversing diabetes in NOD mice and assessed the mechanisms by which this inhibition abrogates T1D. Our data indicate that the PI3Kγ pathway is highly activated in T1D. In NOD mice, we found upregulated expression of phosphorylated Akt (PAkt) in splenocytes. Notably, T regulatory cells (Tregs) showed significantly lower expression of PAkt compared with effector T cells. Inhibition of the PI3Kγ pathway by AS605240 efficiently suppressed effector T cells and induced Treg expansion through the cAMP response element-binding pathway. AS605240 effectively prevented and reversed autoimmune diabetes in NOD mice and suppressed T-cell activation and the production of inflammatory cytokines by autoreactive T cells in vitro and in vivo. These studies demonstrate the key role of the PI3Kγ pathway in determining the balance of Tregs and autoreactive cells regulating autoimmune diabetes.
Collapse
Affiliation(s)
- Jamil Azzi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Robert F. Moore
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Wassim Elyaman
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Marwan Mounayar
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Najib El Haddad
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Sunmi Yang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Mollie Jurewicz
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Ayumi Takakura
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alessandra Petrelli
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Paolo Fiorina
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
| | - Thomas Ruckle
- Therapeutic Area Neurodegenerative Diseases, Merck Serono S.A., Geneva, Switzerland
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital and Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts
- Corresponding author: Reza Abdi,
| |
Collapse
|
21
|
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
|
22
|
Effect of influenza A virus non-structural protein 1(NS1) on a mouse model of diabetes mellitus induced by Streptozotocin. Biochem Biophys Res Commun 2012; 419:120-5. [DOI: 10.1016/j.bbrc.2012.01.146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 01/30/2012] [Indexed: 11/22/2022]
|
23
|
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]
|
24
|
Bresson D, Fousteri G, Manenkova Y, Croft M, von Herrath M. Antigen-specific prevention of type 1 diabetes in NOD mice is ameliorated by OX40 agonist treatment. J Autoimmun 2011; 37:342-51. [PMID: 22063316 DOI: 10.1016/j.jaut.2011.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/05/2011] [Accepted: 10/07/2011] [Indexed: 01/12/2023]
Abstract
Antigen-specific therapies are possibly the safest approach to prevent type 1 diabetes (T1D). However their clinical translation has yielded poor results and greater efforts need to be put into the development of novel strategies to ameliorate their clinical outcome. OX40 is a costimulatory molecule expressed by T cells after antigen recognition and has been implicated in the control effector but also regulatory T cells (Tregs) function in vivo. The activity of OX40 signal on Tregs function has been controversial. In this context we investigated whether an anti-OX40 agonist antibody treatment can ameliorate antigen-specific immune intervention for the prevention of T1D. We show that treatment of non-obese diabetic (NOD) mice with an OX40 agonistic antibody (OX86) reduced type 1 diabetes (T1D) incidence by inducing both CD4(+)CD25(+)Foxp3(+) Tregs and CD4(+)Foxp3(-) T cells expressing the latency-associated peptide (LAP). These OX86-induced CD4(+)Foxp3(-)LAP(+) T cells also demonstrated suppressive activity in vitro. A significant increase in protection was observed when OX86 was combined with insulin B9:23 (insB9:23) peptide immunizations. Synergy resulted from an expansion of IL-10-expressing insB9:23-reactive Tregs which augmented the proportion of CD4(+) T cells with in vivo suppressive activity. Consequently, CD4(+) T cells purified from OX86/insB9:23 combination treatment prevented T1D development when adoptively transferred into recipient mice. These findings suggest that the requirement for OX40 signaling by antigen-induced Tregs can be dominant over its well-documented need for effector memory cell function and may have potentially important implications for improving the clinical translation of antigen-specific prevention of T1D and possibly other autoimmune disorders.
Collapse
Affiliation(s)
- Damien Bresson
- Diabetes Center, Department of Developmental Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
25
|
Waldron-Lynch F, Herold KC. Immunomodulatory therapy to preserve pancreatic β-cell function in type 1 diabetes. Nat Rev Drug Discov 2011; 10:439-52. [DOI: 10.1038/nrd3402] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
26
|
Benitez-Aguirre P, Maahs DM. Report of the 36th ISPAD meeting, Buenos Aires, Argentina, 27-30 October 2010. Pediatr Diabetes 2010; 11:583-91. [PMID: 21118343 DOI: 10.1111/j.1399-5448.2010.00746.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Paul Benitez-Aguirre
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, Australia. paulb6ATchw.edu.au
| | | |
Collapse
|
27
|
Boettler T, von Herrath M. Immunotherapy of type 1 diabetes — How to rationally prioritize combination therapies in T1D. Int Immunopharmacol 2010; 10:1491-5. [DOI: 10.1016/j.intimp.2010.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 07/14/2010] [Accepted: 07/15/2010] [Indexed: 02/06/2023]
|
28
|
Abstract
Selected bacteria, viruses, parasites and nonliving, immunologically active microbial substances prevent autoimmune diabetes in animal models. Such agents might also have a protective effect in humans by providing immune stimuli critical during childhood development. The 'hygiene hypothesis' proposes that reduced exposure to environmental stimuli, including microbes, underlies the rising incidence of childhood autoimmune diseases, including type 1 diabetes mellitus (T1DM). This hypothesis is supported by data that highlight the importance of infant exposure to environmental microbes for appropriate development of the immune system, which might explain the observation that administration of microbes or their components inhibits autoimmune disease in animals. This finding raises the possibility of using live, nonpathogenic microbes (for example, probiotics) or microbial components to modulate or 're-educate' the immune system and thereby vaccinate against T1DM. Progress has been assisted by the identification of receptors and pathways through which gut microbes influence development of the immune system. Such mechanistic data have moved a field that was once regarded as being on the scientific fringe to the mainstream, and support increased funding to advance this promising area of research in the hope that it might deliver the long awaited answer of how to safely prevent T1DM.
Collapse
Affiliation(s)
- Nikolai Petrovsky
- Department of Endocrinology, Flinders Medical Centre, Flinders Drive, Bedford Park, Adelaide, SA 5042, Australia.
| |
Collapse
|
29
|
Sena CM, Bento CF, Pereira P, Seiça R. Diabetes mellitus: new challenges and innovative therapies. EPMA J 2010; 1:138-163. [PMID: 23199048 PMCID: PMC3405309 DOI: 10.1007/s13167-010-0010-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 02/04/2010] [Indexed: 12/25/2022]
Abstract
Diabetes mellitus is a widespread disease prevalence and incidence of which increases worldwide. The introduction of insulin therapy represented a major breakthrough in type 1 diabetes; however, frequent hyper- and hypoglycemia seriously affects the quality of life of these patients. New therapeutic approaches, such as whole pancreas transplant or pancreatic islet transplant, stem cell, gene therapy and islets encapsulation are discussed in this review. Regarding type 2 diabetes, therapy has been based on drugs that stimulate insulin secretion (sulphonylureas and rapid-acting secretagogues), reduce hepatic glucose production (biguanides), delay digestion and absorption of intestinal carbohydrate (alpha-glucosidase inhibitors) or improve insulin action (thiazolidinediones). This review is also focused on the newer therapeutically approaches such as incretin-based therapies, bariatric surgery, stem cells and other emerging therapies that promise to further extend the options available. Gene-based therapies are among the most promising emerging alternatives to conventional treatments. Some of these therapies rely on genetic modification of non-differentiated cells to express pancreatic endocrine developmental factors, promoting differentiation of non-endocrine cells into β-cells, enabling synthesis and secretion of insulin in a glucose-regulated manner. Alternative therapies based on gene silencing using vector systems to deliver interference RNA to cells (i.e. against VEGF in diabetic retinopathy) are also a promising therapeutic option for the treatment of several diabetic complications. In conclusion, treatment of diabetes faces now a new era that is characterized by a variety of innovative therapeutic approaches that will improve quality-life and allow personalized therapy-planning in the near future.
Collapse
Affiliation(s)
- Cristina M. Sena
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Subunit 1, polo 3, Azinhaga de Santa Comba, Celas, 3000-354 Coimbra, Portugal
- IBILI, University of Coimbra, Coimbra, Portugal
| | - Carla F. Bento
- IBILI, University of Coimbra, Coimbra, Portugal
- Centre of Ophthalmology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paulo Pereira
- IBILI, University of Coimbra, Coimbra, Portugal
- Centre of Ophthalmology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Raquel Seiça
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Subunit 1, polo 3, Azinhaga de Santa Comba, Celas, 3000-354 Coimbra, Portugal
- IBILI, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
30
|
Current literature in diabetes. Diabetes Metab Res Rev 2010; 26:i-xi. [PMID: 20474064 DOI: 10.1002/dmrr.1019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
31
|
Rewers M, Gottlieb P. Immunotherapy for the prevention and treatment of type 1 diabetes: human trials and a look into the future. Diabetes Care 2009; 32:1769-82. [PMID: 19794002 PMCID: PMC2752911 DOI: 10.2337/dc09-0374] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Marian Rewers
- From the Barbara Davis Center, University of Colorado, Health Sciences Center, Denver, Colorado
| | - Peter Gottlieb
- From the Barbara Davis Center, University of Colorado, Health Sciences Center, Denver, Colorado
| |
Collapse
|