1
|
Rendell M. Pharmacotherapy of type 1 diabetes - part 3: tomorrow. Expert Opin Pharmacother 2025; 26:535-550. [PMID: 40056035 DOI: 10.1080/14656566.2025.2468906] [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: 12/16/2024] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
INTRODUCTION The last 100 years have seen type 1 diabetes, a previously fatal disease, transformed by the administration of exogenous insulin. AREAS COVERED A standard literature search using the Google and Microsoft search engines and PubMed was performed. The development of synthetic insulins with varying onsets and duration of action improved glucose control, essential to mitigate the microvascular and macrovascular consequences of diabetes. Today insulin pumps guided by continuous glucose monitors are approaching the objective of normalized glucose levels. The area of greatest development is now in attempting to suppress the immune process which results in progressive destruction of the beta cell. It is possible to identify family members of patients with type 1 diabetes who may eventually develop the disease by measuring several beta cell antibodies. Very recently teplizumab, a CD3 inhibitor, has been approved to delay the onset of hyperglycemia in these individuals. EXPERT OPINION The future will see progress in immunosuppression, possibly using specific CAR-Treg cells directed at the beta cell antigens which trigger the immune process. In parallel, stem cell-derived beta cells may eventually make it possible to replace lost beta cells, resulting in a true cure for type 1 diabetes.
Collapse
Affiliation(s)
- Marc Rendell
- The Association of Diabetes Investigators, Omaha, NE, USA
- The Rose Salter Medical Research Foundation, Newport Coast, CA, USA
| |
Collapse
|
2
|
Herold KC, Delong T, Perdigoto AL, Biru N, Brusko TM, Walker LSK. The immunology of type 1 diabetes. Nat Rev Immunol 2024; 24:435-451. [PMID: 38308004 PMCID: PMC7616056 DOI: 10.1038/s41577-023-00985-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2023] [Indexed: 02/04/2024]
Abstract
Following the seminal discovery of insulin a century ago, treatment of individuals with type 1 diabetes (T1D) has been largely restricted to efforts to monitor and treat metabolic glucose dysregulation. The recent regulatory approval of the first immunotherapy that targets T cells as a means to delay the autoimmune destruction of pancreatic β-cells highlights the critical role of the immune system in disease pathogenesis and tends to pave the way for other immune-targeted interventions for T1D. Improving the efficacy of such interventions across the natural history of the disease will probably require a more detailed understanding of the immunobiology of T1D, as well as technologies to monitor residual β-cell mass and function. Here we provide an overview of the immune mechanisms that underpin the pathogenesis of T1D, with a particular emphasis on T cells.
Collapse
Affiliation(s)
- Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT, USA.
- Department of Internal Medicine, Yale University, New Haven, CT, USA.
| | - Thomas Delong
- Anschutz Medical Campus, University of Colorado, Denver, CO, USA
| | - Ana Luisa Perdigoto
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Internal Medicine, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Noah Biru
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, University College London, London, UK.
- Division of Infection & Immunity, University College London, London, UK.
| |
Collapse
|
3
|
Ashraf MT, Ahmed Rizvi SH, Kashif MAB, Shakeel Khan MK, Ahmed SH, Asghar MS. Efficacy of anti-CD3 monoclonal antibodies in delaying the progression of recent-onset type 1 diabetes mellitus: A systematic review, meta-analyses and meta-regression. Diabetes Obes Metab 2023; 25:3377-3389. [PMID: 37580969 DOI: 10.1111/dom.15237] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/11/2023] [Accepted: 07/22/2023] [Indexed: 08/16/2023]
Abstract
AIM Type 1 diabetes mellitus is widely recognized as a chronic autoimmune disease characterized by the pathogenic destruction of beta cells, resulting in the loss of endogenous insulin production. Insulin administration remains the primary therapy for symptomatic treatment. Recent studies showed that disease-modifying agents, such as anti-CD3 monoclonal antibodies, have shown promising outcomes in improving the management of the disease. In late 2022, teplizumab received approval from the US Food and Drug Administration (FDA) as the first disease-modifying agent for the treatment of type 1 diabetes. This review aims to evaluate the clinical evidence regarding the efficacy of anti-CD3 monoclonal antibodies in the prevention and treatment of type 1 diabetes. METHODS A comprehensive search of PubMed, Google Scholar, Scopus and Cochrane Central Register of Controlled Trials (CENTRAL) was conducted up to December 2022 to identify relevant randomized controlled trials. Meta-analysis was performed using a random-effects model, and odds ratios with 95% confidence intervals (CIs) were calculated to quantify the effects. The Cochrane risk of bias tool was employed for quality assessment. RESULTS In total, 11 randomized controlled trials involving 1397 participants (908 participants in the intervention arm, 489 participants in the control arm) were included in this review. The mean age of participants was 15 years, and the mean follow-up time was 2.04 years. Teplizumab was the most commonly studied intervention. Compared with placebo, anti-CD3 monoclonal antibody treatment significantly increased the C-peptide concentration in the area under the curve at shorter timeframes (mean difference = 0.114, 95% CI: 0.069 to 0.159, p = .000). Furthermore, anti-CD3 monoclonal antibodies significantly reduced the patients' insulin intake across all timeframes (mean difference = -0.123, 95% CI: -0.151 to -0.094, p < .001). However, no significant effect on glycated haemoglobin concentration was observed. CONCLUSION The findings of this review suggest that anti-CD3 monoclonal antibody treatment increases endogenous insulin production and improves the lifestyle of patients by reducing insulin dosage. Future studies should consider the limitations, including sample size, heterogeneity and duration of follow-up, to validate the generalizability of these findings further.
Collapse
Affiliation(s)
- Muhammad Talal Ashraf
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | | | | | | - Syed Hassan Ahmed
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | |
Collapse
|
4
|
Wang CJ, Petersone L, Edner NM, Heuts F, Ovcinnikovs V, Ntavli E, Kogimtzis A, Fabri A, Elfaki Y, Houghton LP, Hosse RJ, Schubert DA, Frei AP, Ross EM, Walker LSK. Costimulation blockade in combination with IL-2 permits regulatory T cell sparing immunomodulation that inhibits autoimmunity. Nat Commun 2022; 13:6757. [PMID: 36347877 PMCID: PMC9643453 DOI: 10.1038/s41467-022-34477-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
Blockade of CD28 costimulation with CTLA-4-Ig/Abatacept is used to dampen effector T cell responses in autoimmune and transplantation settings. However, a significant drawback of this approach is impaired regulatory T cell homeostasis that requires CD28 signaling. Therefore, strategies that restrict the effects of costimulation blockade to effector T cells would be advantageous. Here we probe the relative roles of CD28 and IL-2 in maintaining Treg. We find provision of IL-2 counteracts the regulatory T cell loss induced by costimulation blockade while minimally affecting the conventional T cell compartment. These data suggest that combining costimulation blockade with IL-2 treatment may selectively impair effector T cell responses while maintaining regulatory T cells. Using a mouse model of autoimmune diabetes, we show combined therapy supports regulatory T cell homeostasis and protects from disease. These findings are recapitulated in humanised mice using clinically relevant reagents and provide an exemplar for rational use of a second immunotherapy to offset known limitations of the first.
Collapse
Affiliation(s)
- Chun Jing Wang
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Lina Petersone
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Natalie M Edner
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Frank Heuts
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Vitalijs Ovcinnikovs
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Elisavet Ntavli
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Alexandros Kogimtzis
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Astrid Fabri
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Yassin Elfaki
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Luke P Houghton
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Ralf J Hosse
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development (pRED), Schlieren, Switzerland
| | - David A Schubert
- Roche Innovation Center Basel, Roche Pharma Research & Early Development (pRED), Basel, Switzerland
| | - Andreas P Frei
- Roche Innovation Center Basel, Roche Pharma Research & Early Development (pRED), Basel, Switzerland
| | - Ellen M Ross
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection & Immunity, London, UK.
| |
Collapse
|
5
|
Pham VT, Ciccaglione M, Ramirez DG, Benninger RKP. Ultrasound Imaging of Pancreatic Perfusion Dynamics Predicts Therapeutic Prevention of Diabetes in Preclinical Models of Type 1 Diabetes. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1336-1347. [PMID: 35473669 PMCID: PMC9149043 DOI: 10.1016/j.ultrasmedbio.2022.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/07/2022] [Accepted: 03/18/2022] [Indexed: 06/14/2023]
Abstract
In type 1 diabetes (T1D), immune-cell infiltration into islets of Langerhans (insulitis) and β-cell decline occur years before diabetes presents. There is a lack of validated clinical approaches for detecting insulitis and β-cell decline, to diagnose eventual diabetes and monitor the efficacy of therapeutic interventions. We previously determined that contrast-enhanced ultrasound measurements of pancreas perfusion dynamics predict disease progression in T1D pre-clinical models. Here, we test whether these measurements predict therapeutic prevention of T1D. We performed destruction-reperfusion measurements with size-isolated microbubbles in non-obese diabetic (NOD)-severe combined immunodeficiency (SCID) mice receiving an adoptive transfer of diabetogenic splenocytes. Mice received vehicle control or the following treatments: (i) anti-CD3 to block T-cell activation; (ii) anti-CD4 to deplete CD4+ T cells; (iii) verapamil to reduce β-cell apoptosis; or (iv) tauroursodeoxycholic acid (TUDCA) to reduce β-cell endoplasmic reticulum stress. We compared measurements of pancreas perfusion dynamics with subsequent progression to diabetes. Anti-CD3, anti-CD4, and verapamil delayed diabetes development. Blood flow dynamics was significantly altered in treated mice with delayed/absent diabetes development compared with untreated mice. Conversely, blood flow dynamics in treated mice with unchanged diabetes development was similar to that in untreated mice. Thus, measurement of pancreas perfusion dynamics predicts the successful prevention of diabetes. This strategy may provide a clinically deployable predictive marker for therapeutic prevention in asymptomatic T1D.
Collapse
Affiliation(s)
- Vinh T Pham
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark Ciccaglione
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David G Ramirez
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
6
|
Mignogna C, Maddaloni E, D'Onofrio L, Buzzetti R. Investigational therapies targeting CD3 for prevention and treatment of type 1 diabetes. Expert Opin Investig Drugs 2021; 30:1209-1219. [PMID: 34936848 DOI: 10.1080/13543784.2022.2022119] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Immunotherapies for type 1 diabetes mellitus (T1D) have been the focus of intense research over the past few decades; nevertheless, the results of clinical trials have not matched expectations. However, thanks to the recent and promising results on T1D prevention, among all the different immune-intervention tested strategies, clinical evidence on anti-CD3 monoclonal antibodies (mAb) deserve particular attention and in-depth evaluation. AREAS COVERED In this narrative review, we introduce the role of T-cells and their co-receptor CD3 in the pathogenesis of T1D and examine the potential of anti-CD3 mAbs as a treatment for preventing or curing T1D. We discuss pre-clinical studies, phase II/III clinical trials, testing the anti-CD3 mAb teplizumab in subjects at T1D high risk, and testing teplizumab and otelixizumab in T1D recent onset patients. In this work we discuss the current evidence gathered on anti-CD3 therapy to offer insights on the treatment strengths, limitations and unmet needs. EXPERT OPINION Recent phase II clinical trials with teplizumab in recent-onset T1D seem encouraging, but benefits associated with the use of anti-CD3 mAb in recent-onset T1D are still controversial. A better patient selection, based on immunological profiles and specific biomarkers, is crucial to improve clinical outcomes in T1D immunotherapies.
Collapse
Affiliation(s)
- Carmen Mignogna
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Ernesto Maddaloni
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Luca D'Onofrio
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | | |
Collapse
|
7
|
Stewart JM, Posgai AL, Leon JJ, Haller MJ, Keselowsky BG. Combination Treatment with Antigen-Specific Dual-Sized Microparticle System Plus Anti-CD3 Immunotherapy Fails to Synergize to Improve Late-Stage Type 1 Diabetes Prevention in Nonobese Diabetic Mice. ACS Biomater Sci Eng 2020; 6:5941-5958. [PMID: 33320581 PMCID: PMC8108782 DOI: 10.1021/acsbiomaterials.0c01075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) pathophysiology, while incompletely understood, has in part been attributed to aberrant presentation of self-antigen plus proinflammatory costimulation by professional antigen-presenting cells (APCs). Therapies targeting dendritic cells (DCs) offer an avenue to restore antigen-specific tolerance by promoting presentation of self-antigen in an anti-inflammatory or suppressive context. Here, we describe a subcutaneously administered, dual-sized biodegradable microparticle (MP) platform that includes phagocytosable (∼1 μm) and nonphagocytosable (∼30 μm) MPs to deliver pro-tolerogenic factors both intra- and extracellularly, as well as the T1D-associated autoantigen, insulin, to DCs for amelioration of autoimmunity. This MP platform resulted in increased recruitment of DCs, suppressive skewing of DC phenotype with diminished expression of CD86 and MHC-II, increased regulatory T cell (Treg) frequency, and upregulated expression of the checkpoint inhibitor programmed cell death protein 1 (PD-1) on T cells. When administered concomitantly with anti-CD3 antibody, which provides transient T cell depletion while preserving Treg populations, in 12-week-old nonobese diabetic (NOD) mice, regulatory immune populations persisted out to 20 weeks of age; however, combination anti-CD3 and dual-sized MP (dMP) therapy failed to synergistically inhibit diabetes onset.
Collapse
Affiliation(s)
- J. M. Stewart
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| | - A. L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32611
| | - J. J. Leon
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
| | - M. J. Haller
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611
| | - B. G. Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32611
| |
Collapse
|
8
|
Cabello-Kindelan C, Mackey S, Sands A, Rodriguez J, Vazquez C, Pugliese A, Bayer AL. Immunomodulation Followed by Antigen-Specific T reg Infusion Controls Islet Autoimmunity. Diabetes 2020; 69:215-227. [PMID: 31712320 PMCID: PMC6971488 DOI: 10.2337/db19-0061] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 11/06/2019] [Indexed: 12/21/2022]
Abstract
Optimal immune-based therapies for type 1 diabetes (T1D) should restore self-tolerance without inducing chronic immunosuppression. CD4+Foxp3+ regulatory T cells (Tregs) are a key cell population capable of facilitating durable immune tolerance. However, clinical trials with expanded Tregs in T1D and solid-organ transplant recipients are limited by poor Treg engraftment without host manipulation. We showed that Treg engraftment and therapeutic benefit in nonautoimmune models required ablative host conditioning. Here, we evaluated Treg engraftment and therapeutic efficacy in the nonobese diabetic (NOD) mouse model of autoimmune diabetes using nonablative, combinatorial regimens involving the anti-CD3 (αCD3), cyclophosphamide (CyP), and IAC (IL-2/JES6-1) antibody complex. We demonstrate that αCD3 alone induced substantial T-cell depletion, impacting both conventional T cells (Tconv) and Tregs, subsequently followed by more rapid rebound of Tregs Despite robust depletion of host Tconv and host Tregs, donor Tregs failed to engraft even with interleukin-2 (IL-2) support. A single dose of CyP after αCD3 depleted rebounding host Tregs and resulted in a 43-fold increase in donor Treg engraftment, yet polyclonal donor Tregs failed to reverse diabetes. However, infusion of autoantigen-specific Tregs after αCD3 alone resulted in robust Treg engraftment within the islets and induced remission in all mice. This novel combinatorial therapy promotes engraftment of autoantigen-specific donor Tregs and controls islet autoimmunity without long-term immunosuppression.
Collapse
Affiliation(s)
| | - Shane Mackey
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Alexander Sands
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Jennifer Rodriguez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Claudia Vazquez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Allison L Bayer
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
9
|
Dayan CM, Korah M, Tatovic D, Bundy BN, Herold KC. Changing the landscape for type 1 diabetes: the first step to prevention. Lancet 2019; 394:1286-1296. [PMID: 31533907 DOI: 10.1016/s0140-6736(19)32127-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/25/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022]
Abstract
Over several decades, studies have described the progression of autoimmune diabetes, from the first appearance of autoantibodies until, and after, the diagnosis of clinical disease with hyperglycaemia and insulin dependence. Despite the improved management of type 1 diabetes with exogenous insulin, most patients do not meet clinical glycaemic goals, and diabetes remains an important medical problem that affects children and adults. Clinical and preclinical studies have suggested strategies to prevent the diagnosis of type 1 diabetes in people at risk, but the outcomes of previous clinical trials have not met their primary endpoints of disease prevention or delay. The results from the TN-10 teplizumab prevention trial show that the diagnosis of type 1 diabetes can be delayed by treatment with a FcR non-binding monoclonal antibody to CD3 in people at high risk for disease. This Series paper discusses how this clinical achievement raises new questions about for whom, and when, immunological strategies might be developed to prevent type 1 diabetes, and how to achieve this goal.
Collapse
Affiliation(s)
- Colin M Dayan
- Diabetes Research Group, Cardiff University School of Medicine, Cardiff, UK
| | - Maria Korah
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, USA
| | - Danijela Tatovic
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, USA
| | - Brian N Bundy
- Health Informatics Institute, University of South Florida, Tampa, FL, USA
| | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
10
|
Gaglia J, Kissler S. Anti-CD3 Antibody for the Prevention of Type 1 Diabetes: A Story of Perseverance. Biochemistry 2019; 58:4107-4111. [PMID: 31523950 DOI: 10.1021/acs.biochem.9b00707] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by an insulin deficiency. Ever since the discovery of insulin almost 100 years ago, patients with T1D have relied on multiple daily insulin injections to survive an otherwise deadly disease. Despite decades of research and clinical trials, no treatment exists yet to prevent or cure T1D. A recent prevention trial using the anti-CD3 antibody teplizumab in individuals at a high risk of developing T1D has provided the first piece of evidence that a safe and transient intervention may be able to delay disease. In this Perspective, we review the 40-year long history of anti-CD3 and discuss how this antibody became a candidate for the treatment of autoimmune diabetes. The path that leads to its use in this latest clinical trial for T1D has been winding and strewn with setbacks. The molecular actions of the anti-CD3 antibody that target T lymphocytes are well-understood, but its systemic effect on immune function has proven more difficult to unravel. Moreover, preclinical data suggested that the utility of anti-CD3 for the prevention of T1D may be limited. However, the latest clinical data are encouraging and exemplify how a basic discovery can, decades later and with much perseverance, become a promising therapeutic candidate.
Collapse
Affiliation(s)
- Jason Gaglia
- Section for Immunobiology , Joslin Diabetes Center , Boston , Massachusetts 02215 , United States.,Department of Medicine , Harvard Medical School , Boston , Massachusetts 02215 , United States
| | - Stephan Kissler
- Section for Immunobiology , Joslin Diabetes Center , Boston , Massachusetts 02215 , United States.,Department of Medicine , Harvard Medical School , Boston , Massachusetts 02215 , United States
| |
Collapse
|
11
|
Syed I, Rubin de Celis MF, Mohan JF, Moraes-Vieira PM, Vijayakumar A, Nelson AT, Siegel D, Saghatelian A, Mathis D, Kahn BB. PAHSAs attenuate immune responses and promote β cell survival in autoimmune diabetic mice. J Clin Invest 2019; 129:3717-3731. [PMID: 31380811 PMCID: PMC6715391 DOI: 10.1172/jci122445] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/04/2019] [Indexed: 12/15/2022] Open
Abstract
Palmitic acid esters of hydroxy stearic acids (PAHSAs) are endogenous antidiabetic and antiinflammatory lipids. Here, we show that PAHSAs protect against type 1 diabetes (T1D) and promote β cell survival and function. Daily oral PAHSA administration to nonobese diabetic (NOD) mice delayed the onset of T1D and markedly reduced the incidence of T1D, whether PAHSAs were started before or after insulitis was established. PAHSAs reduced T and B cell infiltration and CD4+ and CD8+ T cell activation, while increasing Treg activation in pancreata of NOD mice. PAHSAs promoted β cell proliferation in both NOD mice and MIN6 cells and increased the number of β cells in NOD mice. PAHSAs attenuated cytokine-induced apoptotic and necrotic β cell death and increased β cell viability. The mechanism appears to involve a reduction of ER stress and MAPK signaling, since PAHSAs lowered ER stress in NOD mice, suppressed thapsigargin-induced PARP cleavage in human islets, and attenuated ERK1/2 and JNK1/2 activation in MIN6 cells. This appeared to be mediated in part by glucagon-like peptide 1 receptor (GLP-1R) and not the G protein-coupled receptor GPR40. PAHSAs also prevented impairment of glucose-stimulated insulin secretion and improved glucose tolerance in NOD mice. Thus, PAHSAs delayed the onset of T1D and reduced its incidence by attenuating immune responses and exerting direct protective effects on β cell survival and function.
Collapse
Affiliation(s)
- Ismail Syed
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Maria F. Rubin de Celis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - James F. Mohan
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Pedro M. Moraes-Vieira
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Archana Vijayakumar
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew T. Nelson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Dionicio Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Barbara B. Kahn
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Hosseini H, Yi L, Kanellakis P, Cao A, Tay C, Peter K, Bobik A, Toh BH, Kyaw T. Anti-TIM-1 Monoclonal Antibody (RMT1-10) Attenuates Atherosclerosis By Expanding IgM-producing B1a Cells. J Am Heart Assoc 2018; 7:JAHA.117.008447. [PMID: 29936416 PMCID: PMC6064881 DOI: 10.1161/jaha.117.008447] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Peritoneal B1a cells attenuate atherosclerosis by secreting natural polyclonal immunoglobulin M (IgM). Regulatory B cells expressing T‐cell immunoglobulin mucin domain‐1 (TIM‐1) expanded through TIM‐1 ligation by anti‐TIM‐1 monoclonal antibody (RMT1‐10) induces immune tolerance. Methods and Results We examined the capacity of RMT1‐10 to expand peritoneal B1a cells to prevent atherosclerosis development and retard progression of established atherosclerosis. RMT1‐10 treatment selectively doubled peritoneal B1a cells, tripled TIM‐1+ B1a cells and increased TIM‐1+IgM+interleukin (IL)‐10+ by 3‐fold and TIM‐1+IgM+IL‐10− B1a cells by 2.5‐fold. Similar expansion of B1a B cells was observed in spleens. These effects reduced atherosclerotic lesion size, increased plasma IgM and lesion IgM deposits, and decreased oxidatively modified low‐density lipoproteins in lesions. Lesion CD4+ and CD8+ T cells, macrophages and monocyte chemoattractant protein‐1, vascular cell adhesion molecule‐1, expression of proinflammatory cytokines monocyte chemoattractant protein‐1, vascular cell adhesion molecule‐1, IL1β, apoptotic cell numbers and necrotic cores were also reduced. RMT1‐10 treatment failed to expand peritoneal B1a cells and reduce atherosclerosis after splenectomy that reduces B1a cells, indicating that these effects are B1a cell‐dependent. Apolipoprotein E‐KO mice fed a high‐fat diet for 6 weeks before treatment with RMT1‐10 also increased TIM‐1+IgM+IL‐10+ and TIM‐1+IgM+IL‐10− B1a cells and IgM levels and attenuated progression of established atherosclerosis. Conclusions RMT1‐10 treatment attenuates atherosclerosis development and progression by selectively expanding IgM producing atheroprotective B1a cells. Antibody‐based in vivo expansion of B1a cells could be an attractive approach for treating atherosclerosis.
Collapse
Affiliation(s)
- Hamid Hosseini
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| | - Li Yi
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| | | | - Anh Cao
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Christopher Tay
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| | | | - Alex Bobik
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Immunology, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| | - Tin Kyaw
- Baker Heart and Diabetes Institute, Melbourne, Australia .,Centre for Inflammatory Diseases, Department of Medicine, Southern Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
13
|
Tahvili S, Törngren M, Holmberg D, Leanderson T, Ivars F. Paquinimod prevents development of diabetes in the non-obese diabetic (NOD) mouse. PLoS One 2018; 13:e0196598. [PMID: 29742113 PMCID: PMC5942776 DOI: 10.1371/journal.pone.0196598] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
Quinoline-3-carboxamides (Q compounds) are immunomodulatory compounds that have shown efficacy both in autoimmune disease and cancer. We have in here investigated the impact of one such compound, paquinimod, on the development of diabetes in the NOD mouse model for type I diabetes (T1D). In cohorts of NOD mice treated with paquinimod between weeks 10 to 20 of age and followed up until 40 weeks of age, we observed dose-dependent reduction in incidence of disease as well as delayed onset of disease. Further, in contrast to untreated controls, the majority of NOD mice treated from 15 weeks of age did not develop diabetes at 30 weeks of age. Importantly, these mice displayed significantly less insulitis, which correlated with selectively reduced number of splenic macrophages and splenic Ly6Chi inflammatory monocytes at end point as compared to untreated controls. Collectively, these results demonstrate that paquinimod treatment can significantly inhibit progression of insulitis to T1D in the NOD mouse. We propose that the effect of paquinimod on disease progression may be related to the reduced number of these myeloid cell populations. Our finding also indicates that this compound could be a candidate for clinical development towards diabetes therapy in humans.
Collapse
Affiliation(s)
- Sahar Tahvili
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Dan Holmberg
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Leanderson
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Active Biotech AB, Lund, Sweden
| | - Fredrik Ivars
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
14
|
Gill RG, Pagni PP, Kupfer T, Wasserfall CH, Deng S, Posgai A, Manenkova Y, Bel Hani A, Straub L, Bernstein P, Atkinson MA, Herold KC, von Herrath M, Staeva T, Ehlers MR, Nepom GT. A Preclinical Consortium Approach for Assessing the Efficacy of Combined Anti-CD3 Plus IL-1 Blockade in Reversing New-Onset Autoimmune Diabetes in NOD Mice. Diabetes 2016; 65:1310-6. [PMID: 26718498 PMCID: PMC5860426 DOI: 10.2337/db15-0492] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
There is an ongoing need to develop strategic combinations of therapeutic agents to prevent type 1 diabetes (T1D) or to preserve islet β-cell mass in new-onset disease. Although clinical trials using candidate therapeutics are commonly based on preclinical studies, concern is growing regarding the reproducibility as well as the potential clinical translation of reported results using animal models of human disorders. In response, the National Institutes of Health Immune Tolerance Network and JDRF established a multicenter consortium of academic institutions designed to assess the efficacy and intergroup reproducibility of clinically applicable immunotherapies for reversing new-onset disease in the NOD mouse model of T1D. Predicated on prior studies, this consortium conducted coordinated, prospective studies, using joint standard operating procedures, fixed criteria for study entry, and common reagents, to optimize combined anti-CD3 treatment plus interleukin-1 (IL-1) blockade to reverse new-onset disease in NOD mice. We did not find that IL-1 blockade with anti-IL-1β monoclonal antibody or IL-1trap provided additional benefit for reversing new-onset disease compared with anti-CD3 treatment alone. These results demonstrate the value of larger, multicenter preclinical studies for vetting and prioritizing therapeutics for future clinical use.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/therapeutic use
- Autoimmune Diseases/drug therapy
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Biomedical Research/methods
- CD3 Complex/chemistry
- CD3 Complex/metabolism
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Drug Administration Schedule
- Drug Therapy, Combination
- Female
- Immunoglobulin Fab Fragments/administration & dosage
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/therapeutic use
- Immunotherapy/methods
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors
- Interleukin-1 Receptor Accessory Protein/metabolism
- Interleukin-1beta/antagonists & inhibitors
- Interleukin-1beta/metabolism
- Mice, Inbred NOD
- Multicenter Studies as Topic
- Pilot Projects
- Receptors, Interleukin-1 Type I/antagonists & inhibitors
- Receptors, Interleukin-1 Type I/metabolism
- Recombinant Fusion Proteins/therapeutic use
- Reproducibility of Results
- Research Design
- Specific Pathogen-Free Organisms
- United States
Collapse
Affiliation(s)
- Ronald G Gill
- Departments of Surgery and Immunology, University of Colorado Denver, Aurora, CO
| | | | - Tinalyn Kupfer
- Departments of Surgery and Immunology, University of Colorado Denver, Aurora, CO
| | | | - Songyan Deng
- Yale University School of Medicine, New Haven, CT
| | - Amanda Posgai
- Department of Pathology, University of Florida, Gainesville, FL
| | | | - Amira Bel Hani
- La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Laura Straub
- Immune Tolerance Network, University of California, San Francisco, San Francisco, CA
| | | | - Mark A Atkinson
- Department of Pathology, University of Florida, Gainesville, FL
| | | | | | | | - Mario R Ehlers
- Immune Tolerance Network, University of California, San Francisco, San Francisco, CA
| | | |
Collapse
|
15
|
Kuznetsova A, Yu Y, Hollister-Lock J, Opare-Addo L, Rozzo A, Sadagurski M, Norquay L, Reed JE, El Khattabi I, Bonner-Weir S, Weir GC, Sharma A, White MF. Trimeprazine increases IRS2 in human islets and promotes pancreatic β cell growth and function in mice. JCI Insight 2016; 1. [PMID: 27152363 PMCID: PMC4854304 DOI: 10.1172/jci.insight.80749] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The capacity of pancreatic β cells to maintain glucose homeostasis during chronic physiologic and immunologic stress is important for cellular and metabolic homeostasis. Insulin receptor substrate 2 (IRS2) is a regulated adapter protein that links the insulin and IGF1 receptors to downstream signaling cascades. Since strategies to maintain or increase IRS2 expression can promote β cell growth, function, and survival, we conducted a screen to find small molecules that can increase IRS2 mRNA in isolated human pancreatic islets. We identified 77 compounds, including 15 that contained a tricyclic core. To establish the efficacy of our approach, one of the tricyclic compounds, trimeprazine tartrate, was investigated in isolated human islets and in mouse models. Trimeprazine is a first-generation antihistamine that acts as a partial agonist against the histamine H1 receptor (H1R) and other GPCRs, some of which are expressed on human islets. Trimeprazine promoted CREB phosphorylation and increased the concentration of IRS2 in islets. IRS2 was required for trimeprazine to increase nuclear Pdx1, islet mass, β cell replication and function, and glucose tolerance in mice. Moreover, trimeprazine synergized with anti-CD3 Abs to reduce the progression of diabetes in NOD mice. Finally, it increased the function of human islet transplants in streptozotocin-induced (STZ-induced) diabetic mice. Thus, trimeprazine, its analogs, or possibly other compounds that increase IRS2 in islets and β cells without adverse systemic effects might provide mechanism-based strategies to prevent the progression of diabetes.
Collapse
Affiliation(s)
- Alexandra Kuznetsova
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yue Yu
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Hollister-Lock
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Lynn Opare-Addo
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aldo Rozzo
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marianna Sadagurski
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lisa Norquay
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jessica E Reed
- Housey Pharmaceutical Research Laboratories, Southfield, Michigan, USA
| | - Ilham El Khattabi
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Bonner-Weir
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Gordon C Weir
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Arun Sharma
- Section of Islet Cell and Regenerative Biology, Department of Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Morris F White
- Division of Endocrinology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Affiliation(s)
- Xiangwei Xiao
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| |
Collapse
|
17
|
NLRP3 deficiency protects from type 1 diabetes through the regulation of chemotaxis into the pancreatic islets. Proc Natl Acad Sci U S A 2015; 112:11318-23. [PMID: 26305961 DOI: 10.1073/pnas.1513509112] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Studies in animal models and human subjects have shown that both innate and adaptive immunity contribute to the pathogenesis of type 1 diabetes (T1D). Whereas the role of TLR signaling pathways in T1D has been extensively studied, the contribution of the nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing protein (NLRP) 3 inflammasome pathway remains to be explored. In this study, we report that NLRP3 plays an important role in the development of T1D in the nonobese diabetic (NOD) mouse model. NLRP3 deficiency not only affected T-cell activation and Th1 differentiation, but also modulated pathogenic T-cell migration to the pancreatic islet. The presence of NLRP3 is critical for the expression of the chemokine receptors CCR5 and CXCR3 on T cells. More importantly, NLRP3 ablation reduced the expression of chemokine genes CCL5 and CXCL10 on pancreatic islet cells in an IRF-1-dependent manner. Our results suggest that molecules involved in chemotaxis, accompanied by the activation of the NLRP3 inflammasome, may be effective targets for the treatment of T1D.
Collapse
|
18
|
Hu C, Wong FS, Wen L. Type 1 diabetes and gut microbiota: Friend or foe? Pharmacol Res 2015; 98:9-15. [PMID: 25747961 PMCID: PMC4469505 DOI: 10.1016/j.phrs.2015.02.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 02/25/2015] [Accepted: 02/25/2015] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes is a T cell-mediated autoimmune disease. Environmental factors play an important role in the initiation of the disease in genetically predisposed individuals. With the improved control of infectious disease, the incidence of autoimmune diseases, particularly type 1 diabetes, has dramatically increased in developed countries. Increasing evidence suggests that gut microbiota are involved in the pathogenesis of type 1 diabetes. Here we focus on recent advances in this field and provide a rationale for novel therapeutic strategies targeting gut microbiota for the prevention of type 1 diabetes.
Collapse
Affiliation(s)
- Changyun Hu
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - F Susan Wong
- Institute of Molecular and Experimental Medicine, Cardiff University School of Medicine, Cardiff, UK
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
19
|
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
|
20
|
Papotto PH, Marengo EB, Sardinha LR, Goldberg AC, Rizzo LV. Immunotherapeutic strategies in autoimmune uveitis. Autoimmun Rev 2014; 13:909-16. [PMID: 24833504 PMCID: PMC4181827 DOI: 10.1016/j.autrev.2014.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 04/20/2014] [Indexed: 12/11/2022]
Abstract
Autoimmune uveitis is an organ-specific disorder characterized by irreversible lesions to the eye that predominantly affect people in their most productive years and is among the leading causes of visual deficit and blindness. Currently available therapies are effective in the treatment of a wide spectrum of uveitis, but are often associated with severe side effects. Here, we review ongoing research with promising immunomodulatory therapeutic strategies, describing their specific features, interactions and the responses triggered by the targeted immune molecules that aim to minimize clinical complications and the likelihood of disease relapse. We first review the main features of the disease, diagnostic tools, and traditional forms of therapy, as well as the animal models predominantly used to understand the pathogenesis and test the novel intervention approaches aiming to control the acute immune and inflammatory responses and to dampen chronic responses. Both exploratory research and clinical trials have targeted either the blockade of effector pathways or of their companion co-stimulatory molecules. Examples of targets are T cell receptors (CD3), their co-stimulatory receptors (CD28, CTLA-4) and corresponding ligands (B7-1 and B7-2, also known as CD80 and CD86), and cytokines like IL-2 and their receptors. Here, we summarize the available evidence on effectiveness of these treatments in human and experimental uveitis and highlight a novel CD28 antagonist monovalent Fab′ antibody, FR104, which has shown preclinical efficacy suppressing effector T cells while enhancing regulatory T cell function and immune tolerance in a humanized graft-versus-host disease (GVHD) mice model and is currently being tested in a mouse autoimmune uveitis model with encouraging results.
Collapse
Affiliation(s)
- Pedro Henrique Papotto
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627-701, 2-SS Bloco A, 05651-901 São Paulo, Brazil
| | - Eliana Blini Marengo
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627-701, 2-SS Bloco A, 05651-901 São Paulo, Brazil
| | - Luiz Roberto Sardinha
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627-701, 2-SS Bloco A, 05651-901 São Paulo, Brazil
| | - Anna Carla Goldberg
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627-701, 2-SS Bloco A, 05651-901 São Paulo, Brazil; Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (iii-INCT), Brazil
| | - Luiz Vicente Rizzo
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627-701, 2-SS Bloco A, 05651-901 São Paulo, Brazil; Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (iii-INCT), Brazil.
| |
Collapse
|
21
|
Diaz-de-Durana Y, Lau J, Knee D, Filippi C, Londei M, McNamara P, Nasoff M, DiDonato M, Glynne R, Herman AE. IL-2 immunotherapy reveals potential for innate beta cell regeneration in the non-obese diabetic mouse model of autoimmune diabetes. PLoS One 2013; 8:e78483. [PMID: 24205242 PMCID: PMC3813455 DOI: 10.1371/journal.pone.0078483] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/12/2013] [Indexed: 01/09/2023] Open
Abstract
Type-1 diabetes (T1D) is an autoimmune disease targeting insulin-producing beta cells, resulting in dependence on exogenous insulin. To date, significant efforts have been invested to develop immune-modulatory therapies for T1D treatment. Previously, IL-2 immunotherapy was demonstrated to prevent and reverse T1D at onset in the non-obese diabetic (NOD) mouse model, revealing potential as a therapy in early disease stage in humans. In the NOD model, IL-2 deficiency contributes to a loss of regulatory T cell function. This deficiency can be augmented with IL-2 or antibody bound to IL-2 (Ab/IL-2) therapy, resulting in regulatory T cell expansion and potentiation. However, an understanding of the mechanism by which reconstituted regulatory T cell function allows for reversal of diabetes after onset is not clearly understood. Here, we describe that Ab/IL-2 immunotherapy treatment, given at the time of diabetes onset in NOD mice, not only correlated with reversal of diabetes and expansion of Treg cells, but also demonstrated the ability to significantly increase beta cell proliferation. Proliferation appeared specific to Ab/IL-2 immunotherapy, as anti-CD3 therapy did not have a similar effect. Furthermore, to assess the effect of Ab/IL-2 immunotherapy well after the development of diabetes, we tested the effect of delaying treatment for 4 weeks after diabetes onset, when beta cells were virtually absent. At this late stage after diabetes onset, Ab/IL-2 treatment was not sufficient to reverse hyperglycemia. However, it did promote survival in the absence of exogenous insulin. Proliferation of beta cells could not account for this improvement as few beta cells remained. Rather, abnormal insulin and glucagon dual-expressing cells were the only insulin-expressing cells observed in islets from mice with established disease. Thus, these data suggest that in diabetic NOD mice, beta cells have an innate capacity for regeneration both early and late in disease, which is revealed through IL-2 immunotherapy.
Collapse
Affiliation(s)
- Yaiza Diaz-de-Durana
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Janet Lau
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Deborah Knee
- Biotherapeutics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Christophe Filippi
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Marco Londei
- Translational Medicine, Novartis Institutes of Biomedical Research, San Diego, California, United States of America
| | - Peter McNamara
- Pharmacology Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Marc Nasoff
- Biotherapeutics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Michael DiDonato
- Structural Biology Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| | - Richard Glynne
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
- * E-mail:
| | - Ann E. Herman
- Genetics Department, Genomics Institute of the Novartis Research Foundation, San Diego, California, United States of America
| |
Collapse
|
22
|
Li L, Nishio J, van Maurik A, Mathis D, Benoist C. Differential response of regulatory and conventional CD4⁺ lymphocytes to CD3 engagement: clues to a possible mechanism of anti-CD3 action? JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:3694-704. [PMID: 23986534 PMCID: PMC3932531 DOI: 10.4049/jimmunol.1300408] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Several clinical trials have shown anti-CD3 treatment to be a promising therapy for autoimmune diabetes, but its mechanism of action remains unclear. Foxp3(+) regulatory T cells (Tregs) are likely to be involved, but through unknown mechanistic pathways. We profiled the transcriptional consequences in CD4(+) Tregs and conventional T cells (Tconvs) in the first hours and days after anti-CD3 treatment of NOD mice. Anti-CD3 treatment led to a transient transcriptional response, terminating faster than most Ag-induced responses. Most transcripts were similarly induced in Tregs and Tconvs, but several were differential, in particular, those encoding the IL-7R and transcription factors Id2/3 and Gfi1, upregulated in Tregs but repressed in Tconvs. Because IL-7R was a plausible candidate for driving the homeostatic response of Tregs to anti-CD3, we tested its relevance by supplementation of anti-CD3 treatment with IL-7/anti-IL-7 complexes. Although ineffective alone, IL-7 significantly improved the rate of remission induced by anti-CD3. Four anti-human CD3 mAbs exhibited the same differential effect on IL-7R expression in human as in mouse cells, suggesting that the mechanism also underlies therapeutic effect in human cells, and perhaps a rationale for testing a combination of anti-CD3 and IL-7 for the treatment of recent-onset human type 1 diabetes. Thus, systems-level analysis of the response to anti-CD3 in the early phase of the treatment demonstrates different responses in Tregs and Tconvs, and provides new leads to a mechanistic understanding of its mechanism of action in reverting recent-onset diabetes.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- CD3 Complex/immunology
- CD3 Complex/metabolism
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Drug Synergism
- Gene Expression Regulation/drug effects
- Humans
- Interleukin-7/pharmacology
- Mice
- Mice, Transgenic
- Protein Binding
- Receptors, Interleukin-7/genetics
- Receptors, Interleukin-7/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Li Li
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Junko Nishio
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - André van Maurik
- Immuno Inflammation, GlaxoSmithKline, Stevenage, SG1 2NY, United Kingdom
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
23
|
Hu C, Ding H, Zhang X, Wong FS, Wen L. Combination treatment with anti-CD20 and oral anti-CD3 prevents and reverses autoimmune diabetes. Diabetes 2013; 62:2849-58. [PMID: 23447122 PMCID: PMC3717853 DOI: 10.2337/db12-1175] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease, although B cells also play an important role in T1D development. Both T cell- and B cell-directed immunotherapies have shown efficacy in the prevention and reversal of T1D. However, whether the combined strategy of targeting both T and B cells could further improve therapeutic efficacy remains to be explored. We show that combined treatment with intravenous antihuman CD20 (hCD20) and oral anti-CD3 significantly delays diabetes development in prediabetic hCD20 transgenic NOD mice. More importantly, the combined treatment reverses diabetes in >60% of mice newly diagnosed with diabetes. Further mechanistic studies demonstrated that the addition of oral anti-CD3 to the B-cell depletion therapy synergistically enhances the suppressive function of regulatory T cells. Of note, the oral anti-CD3 treatment induced a fraction of interleukin (IL)-10-producing CD4 T cells in the small intestine through IL-10- and IL-27-producing dendritic cells. Thus, the findings demonstrate that combining anti-CD20 and oral anti-CD3 is superior to anti-CD20 monotherapy for restoring normoglycemia in diabetic NOD mice, providing important preclinical evidence for the optimization of B cell-directed therapy for T1D.
Collapse
Affiliation(s)
- Changyun Hu
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Heyuan Ding
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Endocrinology Section, The 5th People’s Hospital, Fudan University, Shanghai, China
| | - Xiaojun Zhang
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - F. Susan Wong
- Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff, U.K
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Corresponding author: Li Wen,
| |
Collapse
|
24
|
Brooks-Worrell B, Palmer JP. Prevention versus intervention of type 1 diabetes. Clin Immunol 2013; 149:332-8. [PMID: 23803322 DOI: 10.1016/j.clim.2013.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 05/29/2013] [Accepted: 05/30/2013] [Indexed: 01/10/2023]
Abstract
Type 1 diabetes (T1D) is a cell-mediated autoimmune disease. New cases of T1D are on the increase and exogenous insulin therapy is the only intervention regularly initiated for T1D patients. Though tremendous strides have been made in prediction of T1D, prevention and intervention strategies have not experienced the same success. In this review, we will discuss some possible reasons why new intervention therapies for T1D have not been implemented into the mainstream treatment regimen for T1D patients. We will also discuss potential caveats for why prevention and intervention trials in T1D may not have experienced the same success as prediction trials.
Collapse
Affiliation(s)
- Barbara Brooks-Worrell
- Department of Medicine, University of Washington, Seattle, WA 98108, USA; Department of Medicine, VA Puget Sound Health Care System, Seattle, WA 98108, USA.
| | | |
Collapse
|
25
|
Daifotis AG, Koenig S, Chatenoud L, Herold KC. Anti-CD3 clinical trials in type 1 diabetes mellitus. Clin Immunol 2013; 149:268-78. [PMID: 23726024 DOI: 10.1016/j.clim.2013.05.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/29/2013] [Accepted: 05/01/2013] [Indexed: 12/21/2022]
Abstract
Two humanized, anti-CD3 mAbs with reduced FcR binding, teplizumab and otelixizumab, have been evaluated in over 1500 subjects, ages 7-45, with new and recently diagnosed T1D with a range of intravenous doses (3-48mg) and regimens (6-14 days, single or repeat courses). In general, studies that used adequate dosing demonstrated improvement in stimulated C-peptide responses and reduced need for exogenous insulin for two years and even longer after diagnosis. Drug treatment causes a transient reduction in circulating T cells, but the available data suggest that the mechanism of action may involve induction of regulatory mechanisms. The adverse effects of anti-CD3 treatment are infusion-related and transient. The studies have identified significant differences in efficacy among patient groups suggesting that a key aspect for development of this immune therapy is identification of the demographic, metabolic, and immunologic features that distinguish subjects who are most likely to show beneficial clinical responses.
Collapse
|
26
|
Gan MJ, Albanese-O'Neill A, Haller MJ. Type 1 diabetes: current concepts in epidemiology, pathophysiology, clinical care, and research. Curr Probl Pediatr Adolesc Health Care 2012; 42:269-91. [PMID: 23046732 DOI: 10.1016/j.cppeds.2012.07.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease mediated by a combination of genetic and environmental triggers resulting in lymphocytic infiltration of pancreatic islets, destruction of beta cells, and lifelong dependency on exogenous insulin. Although T1D is prevalent (1 in 300) and its incidence is steadily increasing worldwide (3% per year), the exact gene-environment interactions precipitating the disease remain unknown. Living with T1D is challenging for patients, families, and caregivers. Because of the relative paucity of pediatric endocrinologists, general pediatricians and other subspecialists may occasionally be faced with the task of managing diabetes-related complaints. Herein, we provide a comprehensive review of the natural history, pathophysiology, and contemporary management of T1D. In addition, recent advances in T1D research are discussed.
Collapse
Affiliation(s)
- Mary Joyce Gan
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | | | | |
Collapse
|
27
|
A humanised mouse model of cytokine release: comparison of CD3-specific antibody fragments. J Immunol Methods 2012; 384:33-42. [PMID: 22796190 DOI: 10.1016/j.jim.2012.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/03/2012] [Accepted: 07/03/2012] [Indexed: 01/11/2023]
Abstract
CD3-specific antibodies have shown clinical efficacy in both transplantation and autoimmunity. However, targeting CD3 in this way can lead to T-cell activation and a serious cytokine release syndrome mediated by Fcγ receptor binding. An in vivo mouse model has been developed using severe combined immunodeficient (SCID) mice to detect human T-cell depletion and cytokine release into the circulation after administration of OKT3. This system has been used to evaluate OKT3 antibody fragments lacking the entire Fc region alongside whole antibody constructs. These data clearly show that cytokine release is detected with all OKT3 antibody constructs and fragments tested and these can be ranked from highest to lowest as follows: mIgG2a>hIgG1 (Ala-Ala)>hIgG1 diFab' maleimide (DFM)>hIgG1 F(ab')₂>mIgG2a F(ab')₂>hIgG1 Fab'. Furthermore, the monovalent hIgG1 Fab' fragment gives the least cytokine release but it does not deplete human T-cells in this assay format. This suggests that T-cell activation may be playing a role in the mechanism of action of anti-CD3 antibodies and consequently the unwanted cytokine release is potentially unavoidable for this class of molecules. This model system provides a useful tool to aid in understanding and reducing the potential risks of cytokine release following antibody therapy.
Collapse
|
28
|
Ablamunits V, Henegariu O, Hansen JB, Opare-Addo L, Preston-Hurlburt P, Santamaria P, Mandrup-Poulsen T, Herold KC. Synergistic reversal of type 1 diabetes in NOD mice with anti-CD3 and interleukin-1 blockade: evidence of improved immune regulation. Diabetes 2012; 61:145-54. [PMID: 22043003 PMCID: PMC3237664 DOI: 10.2337/db11-1033] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammatory cytokines are involved in autoimmune diabetes: among the most prominent is interleukin (IL)-1β. We postulated that blockade of IL-1β would modulate the effects of anti-CD3 monoclonal antibody (mAb) in treating diabetes in NOD mice. To test this, we treated hyperglycemic NOD mice with F(ab')(2) fragments of anti-CD3 mAb with or without IL-1 receptor antagonist (IL-1RA), or anti-IL-1β mAb. We studied the reversal of diabetes and effects of treatment on the immune system. Mice that received a combination of anti-CD3 mAb with IL-1RA showed a more rapid rate of remission of diabetes than mice treated with anti-CD3 mAb or IL-1RA alone. Combination-treated mice had increased IL-5, IL-4, and interferon (IFN)-γ levels in circulation. There were reduced pathogenic NOD-relevant V7 peptide-V7(+) T cells in the pancreatic lymph nodes. Their splenocytes secreted more IL-10, had increased arginase expression in macrophages and dendritic cells, and had delayed adoptive transfer of diabetes. After 1 month, there were increased concentrations of IgG1 isotype antibodies and reduced intrapancreatic expression of IFN-γ, IL-6, and IL-17 despite normal splenocyte cytokine secretion. These studies indicate that the combination of anti-CD3 mAb with IL-1RA is synergistic in reversal of diabetes through a combination of mechanisms. The combination causes persistent remission from islet inflammation.
Collapse
Affiliation(s)
- Vitaly Ablamunits
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Octavian Henegariu
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jakob Bondo Hansen
- Hagedorn Research Institute, Gentofte, Denmark, and Institute of Biomedicine, University of Copenhagen, Copenhagen, Denmark
| | - Lynn Opare-Addo
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Paula Preston-Hurlburt
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre and Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Thomas Mandrup-Poulsen
- Hagedorn Research Institute, Gentofte, Denmark, and Institute of Biomedicine, University of Copenhagen, Copenhagen, Denmark
| | - Kevan C. Herold
- Department of Immunobiology and Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Corresponding author: Kevan C. Herold,
| |
Collapse
|
29
|
Induction of immunological tolerance by oral anti-CD3. Clin Dev Immunol 2011; 2012:425021. [PMID: 22162715 PMCID: PMC3227236 DOI: 10.1155/2012/425021] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Accepted: 09/04/2011] [Indexed: 12/11/2022]
Abstract
In recent years, our knowledge about immunoregulation and autoimmunity has significantly advanced, but nontoxic and more effective treatments for different inflammatory and autoimmune diseases are still lacking. Oral tolerance is of unique immunologic importance because it is a continuous natural immunologic event driven by exogenous antigen and is an attractive approach for treatment of these conditions. Parenteral administration of anti-CD3 monoclonal antibody is an approved therapy for transplantation in humans and is effective in autoimmune diabetes. Orally administered anti-CD3 monoclonal antibody is biologically active in the gut and suppresses experimental models of autoimmune diseases. Orally delivered antibody does not have side effects including cytokine release syndromes, thus oral anti-CD3 antibody is clinically applicable for chronic therapy. Here we review findings that identify a novel and powerful immunologic approach that is widely applicable for the treatment of human autoimmune conditions.
Collapse
|
30
|
Whitfield-Larry F, Young EF, Talmage G, Fudge E, Azam A, Patel S, Largay J, Byrd W, Buse J, Calikoglu AS, Shultz LD, Frelinger JA. HLA-A2-matched peripheral blood mononuclear cells from type 1 diabetic patients, but not nondiabetic donors, transfer insulitis to NOD-scid/γc(null)/HLA-A2 transgenic mice concurrent with the expansion of islet-specific CD8+ T cells. Diabetes 2011; 60:1726-33. [PMID: 21521873 PMCID: PMC3114397 DOI: 10.2337/db10-1287] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Type 1 diabetes is an autoimmune disease characterized by the destruction of insulin-producing β-cells. NOD mice provide a useful tool for understanding disease pathogenesis and progression. Although much has been learned from studies with NOD mice, increased understanding of human type 1 diabetes can be gained by evaluating the pathogenic potential of human diabetogenic effector cells in vivo. Therefore, our objective in this study was to develop a small-animal model using human effector cells to study type 1 diabetes. RESEARCH DESIGN AND METHODS We adoptively transferred HLA-A2-matched peripheral blood mononuclear cells (PBMCs) from type 1 diabetic patients and nondiabetic control subjects into transgenic NOD-scid/γc(null)/HLA-A*0201 (NOD-scid/γc(null)/A2) mice. At various times after adoptive transfer, we determined the ability of these mice to support the survival and proliferation of the human lymphoid cells. Human lymphocytes were isolated and assessed from the blood, spleen, pancreatic lymph node and islets of NOD-scid/γc(null)/A2 mice after transfer. RESULTS Human T and B cells proliferate and survive for at least 6 weeks and were recovered from the blood, spleen, draining pancreatic lymph node, and most importantly, islets of NOD-scid/γc(null)/A2 mice. Lymphocytes from type 1 diabetic patients preferentially infiltrate the islets of NOD-scid/γc(null)/A2 mice. In contrast, PBMCs from nondiabetic HLA-A2-matched donors showed significantly less islet infiltration. Moreover, in mice that received PBMCs from type 1 diabetic patients, we identified epitope-specific CD8(+) T cells among the islet infiltrates. CONCLUSIONS We show that insulitis is transferred to NOD-scid/γc(null)/A2 mice that received HLA-A2-matched PBMCs from type 1 diabetic patients. In addition, many of the infiltrating CD8(+) T cells are epitope-specific and produce interferon-γ after in vitro peptide stimulation. This indicates that NOD-scid/γc(null)/A2 mice transferred with HLA-A2-matched PBMCs from type 1 diabetic patients may serve as a useful tool for studying epitope-specific T-cell-mediated responses in patients with type 1 diabetes.
Collapse
Affiliation(s)
- Fatima Whitfield-Larry
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill North Carolina
- Corresponding author: Fatima Whitfield-Larry,
| | - Ellen F. Young
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill North Carolina
| | - Garrick Talmage
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill North Carolina
| | - Elizabeth Fudge
- Division of Endocrinology, Departments of Medicine and Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Anita Azam
- Division of Endocrinology, Departments of Medicine and Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Shipra Patel
- Division of Endocrinology, Departments of Medicine and Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph Largay
- Division of Endocrinology, Departments of Medicine and Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Warren Byrd
- Division of Endocrinology, Departments of Medicine and Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - John Buse
- Division of Endocrinology, Departments of Medicine and Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ali S. Calikoglu
- Division of Endocrinology, Departments of Medicine and Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Jeffrey A. Frelinger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill North Carolina
| |
Collapse
|
31
|
Mfarrej B, Keir M, Dada S, Trikudanathan S, Sayegh MH, Sharpe AH, Guleria I. Anti-CD3 mAb treatment cures PDL1-/-.NOD mice of diabetes but precipitates fatal myocarditis. Clin Immunol 2011; 140:47-53. [PMID: 21498129 DOI: 10.1016/j.clim.2011.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 03/13/2011] [Accepted: 03/15/2011] [Indexed: 01/11/2023]
Abstract
Anti-CD3 mAb is an effective therapy that can reverse diabetes in NOD mice and has therapeutic potential in patients with type 1 diabetes (T1D). We administered anti-CD3 to PDL1-/-.NOD mice in order to determine whether this treatment would reverse the development of diabetes in these mice. Mice injected with anti-CD3 mAb neonatally were protected from T1D. However, all of these anti-CD3 mAb treated PDL1-/-.NOD mice developed a wasting disease between 12 and 20 weeks of age with sudden deterioration and weight loss, leading to death within 3-5 days of development of illness. Histology revealed severe inflammation in the heart and skeletal muscles. These results suggest that deficiency of PDL1 in NOD background has the potential to lead to immune-mediated tissue damage in organs other than the pancreas, but this cannot be appreciated in PDL1-/-.NOD mice as the mice develop T1D at an early age and die from diabetes prior to manifesting other autoimmune diseases.
Collapse
Affiliation(s)
- Bechara Mfarrej
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Ke Y, Jiang G, Sun D, Kaplan HJ, Shao H. Anti-CD3 antibody ameliorates experimental autoimmune uveitis by inducing both IL-10 and TGF-β dependent regulatory T cells. Clin Immunol 2011; 138:311-20. [PMID: 21256812 PMCID: PMC3046397 DOI: 10.1016/j.clim.2010.12.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 11/30/2010] [Accepted: 12/21/2010] [Indexed: 01/12/2023]
Abstract
Chronic/recurrent autoimmune (idiopathic) uveitis is difficult to treat and they account for approximately 10% of legal blindness in the Western world. As it has been reported that anti-CD3 antibody can enhance T cell regulatory function, we investigated its effects in vivo on experimental autoimmune uveitis (EAU), a model for autoimmune uveitis in humans. B10RIII mice immunized with an uveitogenic peptide were treated with the F(ab')(2) fragment of anti-CD3 mAb either before or at clinical disease onset. Evaluation of EAU and cellular responses showed that disease was inhibited and the activation and expansion of pathogenic T cells selectively reduced, whereas functions of Treg in vivo were enhanced. Moreover, mice treated with anti-CD3 mAb were resistant to a second challenge with antigen and thus protected from recurrence of disease. Our results demonstrate that anti-CD3 mAb is a potent inhibitor of autoimmune uveitis.
Collapse
Affiliation(s)
- Yan Ke
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY, USA
| | - Guomin Jiang
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY, USA
| | - Deming Sun
- Doheny Eye Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA90033, USA
| | - Henry J. Kaplan
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY, USA
| | - Hui Shao
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
33
|
Van Belle TL, Coppieters KT, Von Herrath MG. Type 1 Diabetes: Etiology, Immunology, and Therapeutic Strategies. Physiol Rev 2011; 91:79-118. [DOI: 10.1152/physrev.00003.2010] [Citation(s) in RCA: 679] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease in which destruction or damaging of the beta-cells in the islets of Langerhans results in insulin deficiency and hyperglycemia. We only know for sure that autoimmunity is the predominant effector mechanism of T1D, but may not be its primary cause. T1D precipitates in genetically susceptible individuals, very likely as a result of an environmental trigger. Current genetic data point towards the following genes as susceptibility genes: HLA, insulin, PTPN22, IL2Ra, and CTLA4. Epidemiological and other studies suggest a triggering role for enteroviruses, while other microorganisms might provide protection. Efficacious prevention of T1D will require detection of the earliest events in the process. So far, autoantibodies are most widely used as serum biomarker, but T-cell readouts and metabolome studies might strengthen and bring forward diagnosis. Current preventive clinical trials mostly focus on environmental triggers. Therapeutic trials test the efficacy of antigen-specific and antigen-nonspecific immune interventions, but also include restoration of the affected beta-cell mass by islet transplantation, neogenesis and regeneration, and combinations thereof. In this comprehensive review, we explain the genetic, environmental, and immunological data underlying the prevention and intervention strategies to constrain T1D.
Collapse
Affiliation(s)
- Tom L. Van Belle
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Ken T. Coppieters
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Matthias G. Von Herrath
- Center for Type 1 Diabetes Research, La Jolla Institute for Allergy and Immunology, La Jolla, California
| |
Collapse
|
34
|
Lin M, Yin N, Murphy B, Medof ME, Segerer S, Heeger PS, Schröppel B. Immune cell-derived c3 is required for autoimmune diabetes induced by multiple low doses of streptozotocin. Diabetes 2010; 59:2247-52. [PMID: 20584999 PMCID: PMC2927947 DOI: 10.2337/db10-0044] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The complement system contributes to autoimmune injury, but its involvement in promoting the development of autoimmune diabetes is unknown. In this study, our goal was to ascertain the role of complement C3 in autoimmune diabetes. RESEARCH DESIGN AND METHODS Susceptibility to diabetes development after multiple low-dose streptozotocin treatment in wild-type (WT) and C3-deficient mice was analyzed. Bone marrow chimeras, luminex, and quantitative reverse transcription PCR assays were performed to evaluate the phenotypic and immunologic impact of C3 in the development of this diabetes model. RESULTS Coincident with the induced elevations in blood glucose levels, we documented alternative pathway complement component gene expression within the islets of the diabetic WT mice. When we repeated the experiments with C3-deficient mice, we observed complete resistance to disease, as assessed by the absence of histologic insulitis and the absence of T-cell reactivity to islet antigens. Studies of WT chimeras bearing C3-deficient bone marrow cells showed that bone marrow cell-derived C3, and not serum C3, is involved in the induction of diabetes in this model. CONCLUSIONS The data reveal a key role for immune cell-derived C3 in the pathogenesis of murine multiple low-dose streptozotocin-induced diabetes and support the concept that immune cell mediated diabetes is in part complement-dependent.
Collapse
Affiliation(s)
- Marvin Lin
- Division of Nephrology, Mount Sinai School of Medicine, New York, New York
| | - Na Yin
- Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, New York
| | - Barbara Murphy
- Division of Nephrology, Mount Sinai School of Medicine, New York, New York
- Transplantation Institute, Mount Sinai School of Medicine, New York, New York
| | - M. Edward Medof
- Institute of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Stephan Segerer
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Peter S. Heeger
- Division of Nephrology, Mount Sinai School of Medicine, New York, New York
- Transplantation Institute, Mount Sinai School of Medicine, New York, New York
| | - Bernd Schröppel
- Division of Nephrology, Mount Sinai School of Medicine, New York, New York
- Transplantation Institute, Mount Sinai School of Medicine, New York, New York
- Corresponding author: Bernd Schröppel,
| |
Collapse
|
35
|
Abstract
Despite improvements in understanding of the natural history of type 1 diabetes (T1D), an intervention capable of consistently and safely preventing or reversing the disease has not been developed. The inability to cure this disorder is largely because of the complex pathophysiology of T1D, continued struggles to identify its precise etiologic triggers, and voids in understanding of the immunologic mechanisms that specifically target pancreatic beta cells. Rapidly improving technologies for managing T1D require critical discussions about equipoise, especially when considering interventions deemed high risk in terms of their safety. This article reviews the conceptual basis for prevention versus intervention trials in settings of T1D, past experiences of clinical trials studying these purposes, and controversial issues regarding disease interdiction, and seeks to provide a roadmap for future efforts to cure this disorder.
Collapse
Affiliation(s)
- Michael J Haller
- Division of Endocrinology, Department of Pediatrics, University of Florida, PO Box 100296, Gainesville, FL 32610, USA.
| | | | | |
Collapse
|
36
|
Nishio J, Feuerer M, Wong J, Mathis D, Benoist C. Anti-CD3 therapy permits regulatory T cells to surmount T cell receptor-specified peripheral niche constraints. ACTA ACUST UNITED AC 2010; 207:1879-89. [PMID: 20679403 PMCID: PMC2931163 DOI: 10.1084/jem.20100205] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Treatment with anti-CD3 is a promising therapeutic approach for autoimmune diabetes, but its mechanism of action remains unclear. Foxp3+ regulatory T (T reg) cells may be involved, but the evidence has been conflicting. We investigated this issue in mice derived from the NOD model, which were engineered so that T reg populations were perturbed, or could be manipulated by acute ablation or transfer. The data highlighted the involvement of Foxp3+ cells in anti-CD3 action. Rather than a generic influence on all T reg cells, the therapeutic effect seemed to involve an ∼50–60-fold expansion of previously constrained T reg cell populations; this expansion occurred not through conversion from Foxp3− conventional T (T conv) cells, but from a proliferative expansion. We found that T reg cells are normally constrained by TCR-specific niches in secondary lymphoid organs, and that intraclonal competition restrains their possibility for conversion and expansion in the spleen and lymph nodes, much as niche competition limits their selection in the thymus. The strong perturbations induced by anti-CD3 overcame these niche limitations, in a process dependent on receptors for interleukin-2 (IL-2) and IL-7.
Collapse
Affiliation(s)
- Junko Nishio
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
37
|
Abstract
IMPORTANCE OF THE FIELD Type 1 diabetes mellitus (T1D) is a T-cell mediated autoimmune disease with selective destruction of beta cells. Immunological interventions are directed at arresting the loss of beta-cell function with the promise that this will make it easier for patients to control their glucose levels. AREAS COVERED IN THIS REVIEW This review provides a summary of the preclinical and clinical research published between 1992 and 2009 using teplizumab and other anti-CD3 antibodies to arrest the loss of beta-cell function in new onset T1D. Data from animal and human studies on the probable mechanism of action of teplizumab are also reviewed. WHAT THE READER WILL GAIN A broad perspective on the use of teplizumab in inducing disease specific tolerance. TAKE HOME MESSAGE In Phase I/II randomized control trials, in patients with new onset T1D, teplizumab slowed the rate of loss of beta-cell function over 2 years of follow-up. Treated patients had better glycemic control and lower insulin requirements. Adverse events so far are mild and of limited duration. Phase III clinical trials are underway to confirm these results and to determine if two courses of drug have greater efficacy in arresting loss of beta-cell function.
Collapse
Affiliation(s)
- Umesh B Masharani
- Division of Endocrinology and Metabolism, Department of Medicine, University of California-San Francisco, 400 Parnassus Avenue, San Francisco, CA 94143, USA.
| | | |
Collapse
|
38
|
Luo X, Herold KC, Miller SD. Immunotherapy of type 1 diabetes: where are we and where should we be going? Immunity 2010; 32:488-99. [PMID: 20412759 PMCID: PMC2860878 DOI: 10.1016/j.immuni.2010.04.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 03/22/2010] [Accepted: 03/31/2010] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disorder characterized by destruction of insulin-producing pancreatic beta cells. Many broad-based immunosuppressive and antigen-specific immunoregulatory therapies have been and are currently being evaluated for their utility in the prevention and treatment of T1D. Looking forward, this review discusses the potential therapeutic use of antigen-specific tolerance strategies, including tolerance induced by "tolerogenic" antigen-presenting cells pulsed with diabetogenic antigens and transfer of induced or expanded regulatory T cells, which have demonstrated efficacy in nonobese diabetic (NOD) mice. Depending on the time of therapeutic intervention in the T1D disease process, antigen-specific immunoregulatory strategies may be employed as monotherapies, or in combination with short-term tolerance-promoting immunoregulatory drugs and/or drugs promoting differentiation of insulin-producing beta cells from endogenous progenitors.
Collapse
Affiliation(s)
- Xunrong Luo
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Kevan C. Herold
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Stephen D. Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
39
|
Raskin P, Mohan A. Emerging treatments for the prevention of type 1 diabetes. Expert Opin Emerg Drugs 2010; 15:225-36. [DOI: 10.1517/14728211003694631] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
40
|
Cernea S, Herold KC. Monitoring of antigen-specific CD8 T cells in patients with type 1 diabetes treated with antiCD3 monoclonal antibodies. Clin Immunol 2010; 134:121-9. [PMID: 19837003 DOI: 10.1016/j.clim.2009.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2009] [Revised: 08/22/2009] [Accepted: 09/12/2009] [Indexed: 01/11/2023]
Abstract
The way in which anti-CD3 monoclonal antibodies (mAbs) modify human immune responses in type 1 diabetes (T1DM) is not known. We prepared a panel of Class I HLA-A2.1 tetramers with peptides from diabetes-associated antigens and studied the frequency and phenotype of the cells in patients with T1DM and blood donors and in patients treated with anti-CD3 mAb (Teplizumab). More patients with T1DM showed positive staining for at least 1 tetramer using frozen and fresh samples (p<0.05). Three months following treatment with anti-CD3 mAb, the proportion of GAD65- and InsB-peptide reactive CD8+ T cells increased (p<0.05). The phenotype of these cells was modulated from naïve to effector memoryRA+. We concludethat Class I MHC tetramers can identify antigen specific CD8+ T cells in patients with T1DM. The frequency of certain specificities increases after treatment with anti-CD3 mAb. Their modulated phenotype may have functional consequences for their pathogenicity.
Collapse
Affiliation(s)
- Simona Cernea
- Department of Immunobiology, Yale University, 10 Amistad Street, 131D, New Haven, CT 06520, USA
| | | |
Collapse
|
41
|
Mehta DS, Christmas RA, Waldmann H, Rosenzweig M. Partial and transient modulation of the CD3-T-cell receptor complex, elicited by low-dose regimens of monoclonal anti-CD3, is sufficient to induce disease remission in non-obese diabetic mice. Immunology 2010; 130:103-13. [PMID: 20059577 DOI: 10.1111/j.1365-2567.2009.03217.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
It has been established that a total of 250 microg of monoclonal anti-mouse CD3 F(ab')(2) fragments, administered daily (50 microg per dose), induces remission of diabetes in the non-obese diabetic (NOD) mouse model of autoimmune diabetes by preventing beta cells from undergoing further autoimmune attack. We evaluated lower-dose regimens of monoclonal anti-CD3 F(ab')(2) in diabetic NOD mice for their efficacy and associated pharmacodynamic (PD) effects, including CD3-T-cell receptor (TCR) complex modulation, complete blood counts and proportions of circulating CD4(+), CD8(+) and CD4(+) FoxP3(+) T cells. Four doses of 2 microg (total dose 8 microg) induced 53% remission of diabetes, similarly to the 250 microg dose regimen, whereas four doses of 1 microg induced only 16% remission. While the 250 microg dose regimen produced nearly complete and sustained modulation of the CD3 -TCR complex, lower doses, spaced 3 days apart, which induced similar remission rates, elicited patterns of transient and partial modulation. In treated mice, the proportions of circulating CD4(+) and CD8(+) T cells decreased, whereas the proportions of CD4(+) FoxP3(+) T cells increased; these effects were transient. Mice with greater residual beta-cell function, estimated using blood glucose and C-peptide levels at the initiation of treatment, were more likely to enter remission than mice with more advanced disease. Thus, lower doses of monoclonal anti-CD3 that produced only partial and transient modulation of the CD3-TCR complex induced remission rates comparable to higher doses of monoclonal anti-CD3. Accordingly, in a clinical setting, lower-dose regimens may be efficacious and may also improve the safety profile of therapy with monoclonal anti-CD3, potentially including reductions in cytokine release-related syndromes and maintenance of pathogen-specific immunosurveillance during treatment.
Collapse
|
42
|
Abstract
The use of anti-CD3 monoclonal antibodies (mAbs) has moved from the bench to the bedside. The experience with the anti-human CD3 mAb OKT3 for treatment of transplant rejection identified limitations that were largely overcome with the creation of humanized non-FcR binding antibodies: Teplizumab, Otelixizumab and Visilizumab. Preclinical studies showed the ability of the drugs to reverse hyperglycaemia in diabetic non-obese diabetic (NOD) mice providing rationale for clinical trials with the agents. The former two drugs have been tested in subjects with new onset type 1 diabetes. They have both shown, in randomized clinical trials, an ability to reduce the loss of insulin production over the first 2 years of the disease. In addition, the need for exogenous insulin to maintain glucose control has been reduced. However, these agents alone do not restore normal glucose control, and future approaches will likely require combinations of agents with complementary immune or metabolic activity.
Collapse
Affiliation(s)
- Adam Kaufman
- Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | | |
Collapse
|
43
|
Podojil JR, Miller SD. Molecular mechanisms of T-cell receptor and costimulatory molecule ligation/blockade in autoimmune disease therapy. Immunol Rev 2009; 229:337-55. [PMID: 19426232 PMCID: PMC2845642 DOI: 10.1111/j.1600-065x.2009.00773.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
SUMMARY Pro-inflammatory CD4(+) T-cell-mediated autoimmune diseases, such as multiple sclerosis and type 1 diabetes, are hypothesized to be initiated and maintained by activated antigen-presenting cells presenting self antigen to self-reactive interferon-gamma and interleukin-17-producing CD4(+) T-helper (Th) type 1/Th17 cells. To date, the majority of Food and Drug Administration-approved therapies for autoimmune disease primarily focus on the global inhibition of immune inflammatory activity. The goal of ongoing research in this field is to develop both therapies that inhibit/eliminate activated autoreactive cells as well as antigen-specific treatments, which allow for the directed blockade of the deleterious effects of self-reactive immune cell function. According to the two-signal hypothesis, activation of a naive antigen-specific CD4(+) T cell requires both stimulation of the T-cell receptor (TCR) (signal 1) and stimulation of costimulatory molecules (signal 2). There also exists a balance between pro-inflammatory and anti-inflammatory immune cell activity, which is regulated by the type and strength of the activating signal as well as the local cytokine milieu in which the naive CD4(+) T cell is activated. To this end, the majority of ongoing research is focused on the delivery of suboptimal TCR stimulation in the absence of costimulatory molecule stimulation, or potential blockade of stimulatory accessory molecules. Therefore, the signaling pathways involved in the induction of CD4(+) T-cell anergy, as apposed to activation, are topics of intense interest.
Collapse
Affiliation(s)
- Joseph R Podojil
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
44
|
The protective effect of simvastatin against low dose streptozotocin induced type 1 diabetes in mice is independent of inhibition of HMG-CoA reductase. Biochem Biophys Res Commun 2009; 379:1076-9. [PMID: 19150339 DOI: 10.1016/j.bbrc.2009.01.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Accepted: 01/04/2009] [Indexed: 11/20/2022]
Abstract
Besides a cholesterol-lowering effect, simvastatin possesses anti-inflammatory properties attributed to inhibition of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase and/or direct binding to, and inhibition of, the integrin lymphocyte function associated antigen-1 (LFA-1). We have shown that simvastatin protects against multiple low dose streptozotocin (MLDS) induced type 1 diabetes in mice. Presently, we examined if this effect could be abolished by co-administration of mevalonic acid, thus determining if the protective effect is dependent or independent of inhibition of HMG-CoA reductase. Mevalonic acid did not affect the protective effect of simvastatin against MLDS diabetes. Moreover, spleens from these mice did not show any signs of toxic side-effects, thus excluding the possibility that the protective effect is secondary to a general inflammatory response. We suggest that simvastatin's protective effect mainly is independent of HMG-CoA reductase inhibition. This implies that inhibition of LFA-1 activation is important for the protective effect exerted by simvastatin.
Collapse
|
45
|
Ochi H, Abraham M, Ishikawa H, Frenkel D, Yang K, Basso A, Wu H, Chen ML, Gandhi R, Miller A, Maron R, Weiner HL. New immunosuppressive approaches: oral administration of CD3-specific antibody to treat autoimmunity. J Neurol Sci 2008; 274:9-12. [PMID: 18804221 PMCID: PMC3167084 DOI: 10.1016/j.jns.2008.07.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 07/10/2008] [Accepted: 07/24/2008] [Indexed: 01/12/2023]
Abstract
One of the major goals for the immunotherapy of autoimmune diseases is the induction of regulatory T cells that mediate immunologic tolerance. Parenteral administration of anti-CD3 monoclonal antibody is an approved therapy for transplantation in humans and is effective in autoimmune diabetes. We have found that oral administration of anti-CD3 monoclonal antibody is biologically active in the gut and suppresses experimental autoimmune encephalomyelitis both prior to disease induction and at the height of disease. Oral anti-CD3 antibody acts by inducing a unique type of regulatory T cell characterized by latency-associated peptide (LAP) on its cell surface that functions in vivo and in vitro via TGF-beta dependent mechanism. Orally delivered antibody would not have side effects including cytokine release syndromes, thus oral anti-CD3 antibody is clinically applicable for chronic therapy. These findings identify a novel and powerful immunologic approach that is widely applicable for the treatment of human autoimmune conditions.
Collapse
Affiliation(s)
- Hirofumi Ochi
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Michal Abraham
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Hiroki Ishikawa
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Dan Frenkel
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kaiyong Yang
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Alexandre Basso
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Henry Wu
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Mei-Ling Chen
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Roopali Gandhi
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Ariel Miller
- Carmel Medical Center, Neuroimmunology Unit, Department of Neurology, 7 Michal Street, Haifa 34362, Israel
| | - Ruth Maron
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Howard L. Weiner
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
46
|
Abstract
BACKGROUND The critical role for regulatory T cells (Treg) in suppressing autoimmune pathology is now well recognised. However, the extent to which defects in regulation can be blamed for the onset of diseases like type 1 diabetes is not clear. OBJECTIVE To collate the available data from mouse models and from studies of type 1 diabetes patients, with a view to understanding the status of the natural Treg compartment in this disease setting. RESULTS/CONCLUSION Available evidence suggests that natural Treg are not under-represented in type 1 diabetes, and that Treg function is only likely to be suboptimal in a subset of patients. Emerging therapeutic strategies that attempt to exploit our knowledge of Treg biology to restore effective immune regulation in type I diabetes are discussed.
Collapse
Affiliation(s)
- Lucy S K Walker
- University of Birmingham Medical School, Medical Research Council Centre for Immune Regulation, Birmingham, B15 2TT, UK.
| |
Collapse
|
47
|
Jonson CO, Pihl M, Nyholm C, Cilio CM, Ludvigsson J, Faresjö M. Regulatory T cell-associated activity in photopheresis-induced immune tolerance in recent onset type 1 diabetes children. Clin Exp Immunol 2008; 153:174-81. [PMID: 18549445 DOI: 10.1111/j.1365-2249.2008.03625.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Extracorporeal photochemotherapy (ECP) has demonstrated immunological effects. The proposed cytotoxic lymphocyte antigen 4 (CTLA-4) involvement, together with forkhead box P3 (FoxP3) and transforming growth factor (TGF)-beta are associated with regulatory T cell activity. The aim of the study was to evaluate the regulatory T cell-associated effect of ECP in recent onset type 1 diabetic (T1D) children. Children (n = 20) with T1D received photopheresis 8-methoxypsoralen + ECP or placebo + shampheresis. Peripheral blood mononuclear cells (PBMC) collected pretreatment (day 1) and post-treatment (day 90) were stimulated with phytohaemagglutinin (PHA) and T1D-associated glutamic acid decarboxylase 65 (GAD(65)) peptide a.a. 247-279. CTLA-4, sCTLA-4, FoxP3 and TGF-beta mRNA transcription was quantified. Photopheresis-treated individuals' relative mRNA expression was generally maintained during the course of the study. Placebo individuals increased in spontaneous CTLA-4 mRNA (P < 0.05) but decreased in expression after stimulation with GAD(65)-peptide (P < 0.05) and PHA (P < 0.05). Spontaneous TGF-beta (P < 0.05) increased whereas PHA- (P < 0.01) and GAD(65)-peptide (P < 0.01)-induced TGF-beta expression decreased in the placebo group, whereas it was maintained in the treated group. Without intervention, expression of CTLA-4 and TGF-beta, stimulated with PHA and GAD(65) peptide, decreased with time, with a parallel reduction of GAD(65)-peptide and PHA-stimulated TGF-beta expression. These parameters were counteracted by ECP. In conclusion, our results indicate that ECP maintains regulatory T cell-associated activity in recent-onset T1D.
Collapse
Affiliation(s)
- C-O Jonson
- Division of Pediatrics and Diabetes Research Centre, Department of Molecular and Clinical Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | | | | | | | | | | |
Collapse
|
48
|
Rydgren T, Vaarala O, Sandler S. Simvastatin protects against multiple low-dose streptozotocin-induced type 1 diabetes in CD-1 mice and recurrence of disease in nonobese diabetic mice. J Pharmacol Exp Ther 2007; 323:180-5. [PMID: 17636011 DOI: 10.1124/jpet.107.122655] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Statins are drugs well known for their cholesterol-lowering properties. Lately, statins have been shown to possess anti-inflammatory properties that might be attributed to inhibition of leukocyte adhesion and migration to sites of inflammation. Therefore, we have explored the effects of administration of simvastatin (30 mg/kg body weight given i.p. once a day, from days 4-14) on the development of diabetes induced by multiple low-dose streptozotocin (MLDS) in CD-1 mice, a type 1 diabetes model. We found that treatment with simvastatin could delay and in certain mice fully protect against MLDS-induced diabetes. The protective effect could last up to 3 weeks after simvastatin treatment was ended. Morphological examinations of the pancreas suggest that simvastatin might reduce the islet inflammation. Based on experiments in vitro, using isolated pancreatic islets, we conclude that the protective effect of simvastatin is not mediated by a direct effect on streptozotocin action but rather the result of an immunomodulatory effect. This was reinforced by the finding that simvastatin treatment also prolonged islet function in the recurrence of disease model in diabetic nonobese diabetic mice.
Collapse
Affiliation(s)
- Tobias Rydgren
- Department of Medical Cell Biology, Biomedicum, P.O. Box 571, SE-75123 Uppsala, Sweden.
| | | | | |
Collapse
|
49
|
Sia C, Weinem M. "Persistence of Diabetes" - Why Has Research into Type 1 Diabetes not Made Significant Advances? Rev Diabet Stud 2007; 3:156-60. [PMID: 17487339 PMCID: PMC1828289 DOI: 10.1900/rds.2006.3.156] [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: 11/03/2022] Open
Affiliation(s)
- Charles Sia
- Vaccine Center, National Health Research Institutes, 35 Keyan Road, Zhunan Township, Miaoli County, Taiwan
| | - Michael Weinem
- Society for Biomedical Diabetes Research, 47057 Duisburg, Germany
| |
Collapse
|
50
|
Abstract
Endowed with the ability to actively suppress an immune response, regulatory T cells (Tregs) hold the promise of halting ongoing pathogenic autoimmunity and restoring self-tolerance in patients suffering from autoimmune diseases. Through many in vitro and in vivo studies, we have learned that Tregs can function in the lymph nodes as well as in the peripheral tissues. In vivo, Tregs act through dendritic cells to limit autoreactive T-cell activation, thus preventing their differentiation and acquisition of effector functions. By limiting the supply of activated pathogenic cells, Tregs prevent or slow down the progression of autoimmune diseases. However, this protective mechanism appears insufficient in autoimmune individuals, likely because of a shortage of Tregs cells and/or the development and accumulation of Treg-resistant pathogenic T cells over the long disease course. Thus, restoration of self-tolerance in these patients will likely require purging of pathogenic T cells along with infusion of Tregs with increased ability to control ongoing tissue injury. In this review, we highlight advances in dissecting Treg function in vivo in autoimmune settings and summarize multiple studies that have overcome the limitations of the low abundance of Tregs and their hypoproliferative phenotype to develop Treg-based therapies.
Collapse
Affiliation(s)
- Qizhi Tang
- UCSF Diabetes Center, Department of Medicine and Department of Pathology, University of California, San Francisco, CA 94143-0540, USA
| | | |
Collapse
|