Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jun 4.
Published in final edited form as: Nat Rev Immunol. 2024 Feb 2;24(6):435–451. doi: 10.1038/s41577-023-00985-4

The immunology of type 1 diabetes

Kevan C Herold 1,2, Thomas Delong 3, Ana Luisa Perdigoto 2,4, Noah Biru 1, Todd M Brusko 5, Lucy S K Walker 6,7
PMCID: PMC7616056  NIHMSID: NIHMS1977272  EMSID: EMS194751  PMID: 38308004

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.

Introduction

Type 1 diabetes (T1D) is an autoimmune disease that results in the killing of pancreatic islet β-cells, leading to metabolic failure requiring lifelong insulin treatment. T1D occurs in individuals with a genetic predisposition, in whom disease onset and progression is triggered by environmental or immunological events. Although the need for multiple factors – genetic, environmental and immunological – to induce the disease would suggest that T1D is a rare event, it is not uncommon. In 2021, there were an estimated 8.4 million people worldwide with T1D, with around 510,000 new cases diagnosed in that year1. Of these, 18% were younger than 20 years, and 64% were aged 20–59 years. The remainder were aged 60 years or older. Approximately 35,000 non-diagnosed individuals died within 12 months of symptom onset. The rates between countries vary greatly: 20% of individuals with T1D were in low-income and lower-middle-income countries. With modelling, using random-forest regression of published T1D mortality data, the remaining life expectancy for a 10-year-old individual diagnosed in a high-income country was reduced by approximately 11 years and in a low-income country by 49 years (ref. 1).

T1D develops in a clinically silent manner over a period of months to years. Its progression has been defined by key parameters that appear after the initiation of autoimmunity. Stage 1 is defined by the presence of two or more autoantibodies, stage 2 is defined by two or more autoantibodies and β-cell dysfunction as β-cell mass is progressively eliminated, and stage 3 equates to clinical T1D. Individuals diagnosed as stage 1 have a 35–50% risk of progressing to clinical T1D within 5–6 years. This risk is raised to 75% for those at stage 2, with a median time to diagnosis of 2 years2,3 (Table 1).

Table 1 ∣.

Measures of autoimmunity and disease activity in individuals with and at risk for T1D

Antigen, measured
parameter or technique
Comment Challenges
Autoantibody assays
Unknown Islet cell antibody (ICA) was the first autoantibody described; involves identification of serum binding to group O pancreas; not specific for β-cells The levels of autoantibodies may fluctuate and they do not reflect active β-cell killing
Glutamic acid decarboxylase 65 (GAD65) GAD65 is an enzyme that synthesizes the neurotransmitter GABA at the nerve terminals and synapses. It is found in islets of Langerhans. The affinity of autoantibodies to this antigen is higher in individuals who carry HLA-DR3. The presence of high-affinity glutamic acid decarboxylase antibody (GADA) may indicate high risk218
ICA512/IA-2 (IA2A) An autoantigen with a tyrosine phosphatase-like domain. Children who seroconverted early in life (median age <2 years) and develop insulin autoantibodies (IAA) and insulinoma-associated antigen 2 autoantibodies (IA2A) have the highest risk of progression to T1D, and this risk was unaffected by the presence or absence of GADA status219
ZnT8 (ZnT8A) Zinc transporter found on β-cells
(pro)Insulin (IAA) Antibodies to insulin and proinsulin, which are predominantly found in children carrying HLA-DR4. The presence of IAA to (pro) insulin allows to identify high risk of progression to T1D220-222
Modified (citrullinated) antigens Antibodies to citrullinated glucokinase and carbonylated P4Hb146,223. These can be found with T cells reactive to modified peptides
β-Cell assays
Measurement of INS DNA with unmethylated CpG sites This assay detects epigenetic marks of β-cell-derived INS DNA in the serum The half-life of circulating DNA is short and the number of dying β-cells at any one time is small
Relative proinsulin levels Reduced proinsulin processing and increased relative levels of proinsulin (compared to C-peptide or insulin), as measured by enzyme linked immunosorbent assays (ELISA) in serum, are associated with β-cell stress responses Choice of assay may affect the sensitivity of the assay. Age is a confounder
Functional assays Impaired first-phase insulin secretory responses and glucose intolerance are indicators of β-cell dysfunction In prevention studies, there is a poor correlation between C-peptide responses and glucose responses to an oral glucose tolerance test (OGTT)3. Assessment of insulin secretory dynamics are not widely used and their sensitivity to successful immune therapies is not clear.
B cell assays
Anergic B cells At-risk patients appear to have lower levels of anergic B cells82 B cells are not the direct cause of β-cell killing but may facilitate autoreactive T cell responses by acting as antigen-presenting cells. The frequency of anergic B cells has not been evaluated as a biomarker
T cell assays
ELIspots/flow cytometry cell activation CD4+ T cells that are specific for multiple autoantigens can be detected224,225 The sensitivity of the assay is limiting because the frequency of autoantigen-specific cells is on the order of 1:105 or less
Tetramers and Qdot probes with ‘fluorescent’ labels Class I and class II tetramers have been used to identify autoantigen-specific T cells from enriched cell populations. The specificity for identifying antigen-specific T cells may be improved by using multiple fluorochromes for tetramers or Qdots226. Preproinsulin-reactive cells have been linked to β-cell killing110 The frequency of autoantigen-specific T cells in the peripheral blood may be similar in individuals with T1D and healthy controls. Therefore, the frequency of the autoantigen-specific T cells may not be a specific biomarker of the disease
T cell libraries CD8+ T cell libraries detect autoantigen-specific T cells from expanded enriched CD8+ T cell subpopulations227 Only the relative frequency of the autoantigen-specific cells can be determined
Proliferation assays Proliferative or activation responses (for example, expression of CD40L) to pools or single peptides can be measured228 The absolute frequency of cells cannot be ascertained. Cultures are frequently supplemented with cytokines which may increase background responses
Immunoblot Islet antigens transferred to nitrocellulose and added to cell proliferation assays225 The precise antigen(s) are not defined.

This table lists assays that have been used clinically to identify individuals with and at-risk for type 1 diabetes (T1D) and to monitor disease activity.

Here we review the immunological basis of T1D and discuss how preclinical and clinical studies, as well as investigations of human samples, have provided new insights and opportunities for the development of therapeutics. We also provide guidance for interventions that prevent disease development or induce remission of the disease.

Genetic factors in T1D

Relatives of individuals with T1D have a significantly increased risk of developing the disease, but almost 90% of newly diagnosed individuals do not have a relative with the disease4. In first-degree relatives, the risk ranges from 3% in children of female probands to 5% in children of male probands, 8% in siblings and can be greater than 70% in identical twins5-7. The human leucocyte antigen (HLA) genes account for approximately 40–50% of familial predisposition8. The most frequent HLA haplotypes associated with T1D are DR3-DQ2 and DR4-DQ8, which are present in up to 90% of individuals with T1D (refs. 5,8). The highest-risk HLA genotype (DR3/DR4-DQB1*03:02) has an odds ratio of greater than 16 for the diagnosis of T1D by the age 15 years (ref. 9). A molecular basis for this genetic association was the finding that protective HLA-DQ alleles encoded an aspartic acid at position 57 of the HLA β-chain, allowing the binding of negatively charged peptide residues10-12. The high-risk HLA–peptide complex is speculated to promote the activation of autoreactive diabetogenic T cells.

Genetic studies have identified 143 regions of the genome that are associated with susceptibility for T1D, comprising nearly 60 independent candidate genes13 (Fig. 1). Substantial genetic insights into the disease have been gained from studies in non-obese diabetic (NOD) mice, which spontaneously develop T1D, for example via comparisons with congenic animals that carry discrete regions of DNA from another strain. Many of the risk loci in both mice and humans are associated with genes that are involved in the regulation of the immune response, including PTPN22, CTLA4 and IL2RA (ref. 5). Reduced expression of IL-2RA has been postulated to account for the reduced fitness and function of regulatory T (Treg) cells that is observed in T1D (ref. 14). T1D-associated gene variants are enriched in the open chromatin of T and B cells, and particularly in stimulated CD4+ effector T cells, suggesting that they may exert functional effects in these cell types13. The second strongest genetic association (after the HLA–MHC haplotype) involves a variable number of tandem repeats (VNTRs) located upstream of the gene encoding proinsulin (INS)15. Here, a high number of VNTRs is associated with a high expression of proinsulin in the thymus, which confers a dominant protective effect, purportedly by enhancing T cell tolerance15-17.

Fig. 1 ∣. Risk genes for type 1 diabetes encode proteins that impact T cell development and function.

Fig. 1 ∣

Certain haplotypes of the human HLA class II locus and a high number of repeats of the insulin variable number of tandem repeat (VNTR) region have been linked with autoimmune responses in pancreatic islets in individuals with type 1 diabetes. Approximately 50 other candidate risk genes have been found to encode a variety of proteins that are involved in T cell function, activation and differentiation, including kinases and phosphatases, transcription factors, receptors and ligands, cytokines, cytokine receptors and T cell effector molecules, structural and adaptor proteins, and costimulatory or co-inhibitory proteins. Additional risk genes are thought to encode proteins that are involved in T cell activation or differentiation indirectly by modifying antigen-presenting cells or by targeting β-cells (not shown). APC, antigen-presenting cell.

Environmental factors implicated in T1D

A number of large observational studies (ENDIA, GPPAD, TEDDY, DAISY) are currently underway to explore environmental factors such as viral exposure, dietary intake and the microbiome, which have been implicated in T1D development18-21. Viral infections have long been suspected to have the potential to break immunological tolerance to self-antigens22. The idea that a viral agent can promote a loss in immune tolerance and initiate T1D extends back to the earliest reports of cellular infiltrates in the pancreas of organ donors with T1D (ref. 23). These efforts have been reinvigorated by using modern molecular sequencing for viral identification and histopathological analyses of the pancreas from organ donors with T1D (refs. 24,25). Multiple approaches, including the analysis of stool samples, established a link between enterovirus infection and T1D, and persistent infection is associated with the appearance of islet autoantibodies and progression to overt clinical disease26. Establishing causal relationships between T1D and specific viruses remains challenging, as it would require intensive sampling of large populations over long periods of time to capture the timing of infection relative to the initiation of islet autoimmunity and T1D. Furthermore, enterovirus infection might be just one of many possible viral triggers for T1D, adding to the challenge of unravelling causality27. An intriguing recent observation concerns the efficacy of the antivirals pleconaril and ribavirin in new-onset T1D in a phase II randomized trial28. It has also been suggested that the incidence of T1D increased during the COVID-19 pandemic in certain populations29,30. However, a direct association between SARS-CoV-2 infection and islet autoimmunity has not been established31,32.

Individuals with T1D often have features of a dysregulated microbiome, possibly linked to changes in gut permeability33 or viral infection34,35. Interestingly, high-risk HLA haplotypes (such as HLA class II DR3/DR4-DQ2/8) were associated with a potential loss of tolerance and antibody development to host commensal microorganisms36. The microbiome also affects mucosal invariant T cells, innate-like T cells that function to preserve gut-barrier integrity, which have also been implicated in the pathogenesis of T1D (refs. 37,38). The ability of the microbiome to alter multiple aspects of immunity suggests that it tends to affect not only the initiation and progression of T1D but potentially also responses to immune modulating therapies39 (Fig. 2).

Fig. 2 ∣. The influence of the microbiome in T1D.

Fig. 2 ∣

Several mechanisms have been proposed to explain associations of the microbiome with type 1 diabetes (T1D). An analysis of stool samples established a link between enterovirus infection and T1D, and persistent infection is associated with islet autoimmunity and progression to overt clinical disease26. Invariant immune cells such as mucosal-associated invariant T cells, innate lymphoid cells and others have a role in the maintenance of the intestinal barrier. Their dysfunction and loss of microbiome diversity can lead to loss of tolerance to commensal bacteria and allow the outgrowth of inflammation-inducing species that may affect local tolerance mechanisms, enhance the function of autoreactive cells or even activate immune cells that have cross-reactivity between commensal bacteria and autoantigens36. In addition, metabolic products of the microbiome, such as short-chain fatty acids (SCFA), may affect systemic immune regulation, including via direct effects on mucosal-associated invariant T (MAIT) cells34,37,38,229,230. Antibodies to commensal microbiota have been identified in individuals at risk for T1D, and in non-obese diabetic mice, cross-reactive antigens that are recognized by CD8+ T cells were found (not shown)24,25,112 Moreover, microbial exposure may also affect the efficacy of immunotherapies for T1D (ref. 39).

The role of innate immunity

Innate immune cells, such as dendritic cells, macrophages, neutrophils and natural killer (NK) cells, are thought to play a role at the earliest stages of T1D. Dendritic cells in the pancreatic lymph nodes serve as antigen-presenting cells that can secrete inflammatory mediators such as IL-12 and IL-15, which can promote the expression of costimulatory molecules and activate autoreactive T cells40-42. Macrophages that reside in pancreatic islets have also been implicated in both the initiation and the ongoing destruction of pancreatic β-cells, in part through their ability to secrete cytokines (such as TNF and IL-1β, which can stimulate the secretion of IL-6) and reactive oxygen species40,43-48. In NOD mice that have received pathological (BDC2.5) CD4+ T cells or have been treated with monoclonal antibodies (mAbs) targeted at PD1 to accelerate disease onset, the depletion of macrophages can prevent the disease49,50.

Neutrophils are present in the exocrine pancreas and may promote the initiation of T1D by secreting cytokines and chemotactic factors that impact other immune cells, including macrophages and dendritic cells40,51-57. The role of NK cells in T1D is not fully understood, perhaps in part owing to the diversity of NK cell types, but they have also been linked to T1D pathogenesis58,59.

Inflammatory mediators such as IL-1 and type 1 interferons are involved in the innate immune response in T1D. Patients with recently diagnosed T1D have higher serum levels of macrophage-derived IL-1β compared with controls, and monocytes from individuals with T1D secrete elevated levels of IL-1β and IL-6 (refs. 60,61). Moreover, NOD mice and humans with T1D have higher expression of interferon (IFN) response markers in islet cells, and a transient IFN-induced gene signature precedes the emergence of islet autoantibodies in humans with T1D (ref. 62). Blocking the IFNα receptor in young NOD mice delayed onset of diabetes, pointing to a role for this cytokine early in the disease process63-66. Mechanistically, IFNα has been shown to promote the presentation of self-antigens by islet cells and, consequently, the detection of these cells by cytotoxic T cells. Moreover, it induces the secretion of various chemokines that are involved in the recruitment of immune cells such as T cells and NK cells62-67. For example, pancreatic islets from individuals with T1D were shown to express IFN-responsive genes that are involved in antigen presentation, endoplasmic reticulum stress and apoptosis68-71. IFNα can also act in synergy with IL-1β to induce β-cell killing68-71. A polymorphism in the TYK2 gene, which encodes a Janus kinase (JAK) that is responsive to IFNα, is associated with T1D, and knocking out TYK2 in human stem cell-derived islets reduces their sensitivity to T cell-mediated killing72.

Autoantibodies and B cells

The likelihood of progression to T1D can be predicted by measuring islet autoantibody levels and specificities73. In individuals with a genetic predisposition to T1D, the first autoantibodies generally appear at ages 1–2 years and are specific for insulin, or at ages 4–5 years and are specific for either glutamic acid decarboxylase (GAD) or insulin74. Genetic factors may dictate whether GAD-binding (typically in individuals carrying HLA-DR3) or insulin-binding (typically in individuals carrying HLA-DR4) autoantibodies appear first. In an analysis of 24,662 participants from five prospective studies, the progression of autoantibody reactivity was shown to differ by age, sex and HLA-DR haplotype75. Currently, a model that combines a genetic risk score with the number of islet autoantibodies provides the best prediction of progression to T1D (ref. 76). The trigger that initiates the appearance of the first autoantibodies remains elusive but could conceivably encompass β-cell damage or stress, possibly secondary to viral infection(s). Intriguingly, a small increase in postprandial blood glucose can be detected around 2 months before seroconversion, supporting the idea that β-cell perturbations may be associated with the initiation of autoantibody production77.

Autoantibodies in T1D are not believed to be pathogenic. However, B cells have an important role in disease pathogenesis, most probably as antigen-presenting cells. Loss of B cell antigen presentation, but not loss of antibody secretion, is sufficient to prevent the disease in the NOD model78,79. NOD mice with B cells specific for the model antigen HEL, which were unable to take up islet antigen through their B cell receptor, were protected from T1D development and lacked T cell responses to the islet autoantigen GAD80. Conversely, selective B cell recognition of insulin can enhance diabetes in NOD mice81. Loss of anergy of high-affinity insulin-binding B cells has been identified in patients prior to and at the time of T1D diagnosis82. Interestingly, people diagnosed with T1D at <7 years of age have a higher number of B cells in the islet infiltrate and fewer remaining β-cells than those diagnosed at an older age83.

Features of autoreactive T cells

An abundance of data from NOD mice and other preclinical models indicates an important role for T cells in T1D development and progression. Evidence for a pathogenic role of T cells in humans was provided by clinical studies four decades ago, when it was shown that treatment with cyclosporin A, an immunosuppressive drug that suppressed T cell receptor signalling, promoted the preservation of β-cell function (determined by measuring stimulated C-peptide levels, a by-product of insulin production) and reduced requirements for exogenous insulin. Cyclosporin A or chronic broad-spectrum immune-suppressive drugs (such as a combination of azathioprine with prednisone) were abandoned largely because of toxicity concerns. Information about the architecture, participating cells and mediators in autoimmune insulitis has been greatly expanded with the availability of pancreases from deceased donors obtained by the Network for Pancreatic Organ donors with Diabetes programme (nPOD; www.jdrfnpod.org), as well as from pancreatic biopsies from living individuals with T1D (DiViD; www.oslodiabetes.no)84. Tissue imaging mass cytometric analysis of human T1D pancreatic tissue revealed that CD4+ and CD8+ T cells are recruited to the islets simultaneously, with a trend for insulitis to be associated with islets that retain insulin-containing β-cells85. In human insulitis, CD8+ T cells outnumber CD4+ T cells86,87. Of note, in both NOD mice and human insulitis, not all islets are affected equally: within the same pancreas and even within the same region, there may be islets that are completely free of immune cells and others that show more substantial cellular infiltrates (Fig. 3a). The reason(s) for this heterogeneity remain unclear. The extent of insulitis is inversely correlated with the duration of diabetes, with one study identifying insulitis in all donors examined within 1 year of diagnosis and in 19% of those with disease durations of longer than 1 year87.

Fig. 3 ∣. Insulitis and CD8+ T cells in type 1 diabetes.

Fig. 3 ∣

a, Confocal microscopy of islets from a 12-week-old non-obese diabetic (NOD) mouse. Islets were stained for insulin (green), CD45 (red) and nuclei (blue). The two examples shown are of an islet without insulitis (left) and with immune cell infiltration (right) from the same mouse, illustrating the heterogeneity of β-cell destruction during type 1 diabetes pathogenesis. (Image taken in the Yale Center for Cellular & Molecular Imaging Confocal Facility). b, Stem-like autoreactive CD8+ memory T cells targeting β-cell antigens in type 1 diabetes have properties of both stem memory T (TSCM) cells and effector memory T (TEM) cells. Middle row: the stem-like population has a genome methylation profile that reflects this intermediate phenotype, which is reflected by the methylation patterns of key transcription factors related to differentiation (EOMES, TBX21, TCF7), effector functions (PRF1, GZMK, IFNγ) and exhaustion (TOX, BATF). Bottom row: in NOD mice, this stem-like population of T cells resides in the pancreatic lymph nodes, wherein the cells can differentiate into short-lived TEM cells that migrate to the pancreas115 and destroy β-cells.

CD4+ T cells

CD4+ T cells specific to proinsulin, GAD and insulinoma-associated protein-2 (IA2) have been isolated from the pancreatic islets of organ donors who had T1D (refs. 88-90). In NOD mice, many of the islet-infiltrating CD4+ T cells bind a fragment of the insulin B chain (amino acids B:9–23), and a single amino acid mutation in this region abolishes immunogenicity and the disease91. Autoantigen-reactive CD4+ T cells can also be identified in the peripheral blood using tetramer staining or peptide restimulation coupled with activation marker expression. The frequencies of activated (CD38+) islet antigen-specific memory CD4+ T cells (reactive against GAD65, IGRP, ZnT8, ChgA, PPI78-90K88S or PPI35-47R46E) in the peripheral blood are elevated in people with T1D (ref. 92) (Table 1). Pathogenic CD4+ T cells in T1D were initially thought to have a T helper 1 (TH1) phenotype, with TH2 cytokines generally regarded as protective; however, in recent years, this view has become more nuanced93. Some evidence suggests a pathogenic role for a TH17 response in T1D (refs. 94,95).

Intriguingly, an increased frequency of circulating T cells with a follicular helper (TFH) phenotype has been noted in individuals with T1D, and the hallmark TFH cytokine IL-21 appears to be elevated, consistent with a role for TFH cells in pathogenesis96-98 (Fig. 4). TFH cells support the formation of ectopic B cell germinal centres. A recent work suggests that ectopic germinal centres form in the thymus of NOD mice, supported by local TFH cells99. T peripheral helper (TPH) cells, which are phenotypically similar to TFH cells but thought to act in peripheral tissues, are also increased at T1D onset and in at-risk individuals who subsequently progress to T1D (ref. 100). TFH and TPH cells are highly sensitive to costimulation through the interaction of CD28 with CD80/CD86 (ref. 101), which means that they may be targeted by drugs that block these receptors. One such drug is abatacept, a fusion protein composed of the extracellular domain of CTLA4 (a CD28 homologue that binds CD80/CD86 with higher affinity) linked to a modified Fc portion of human IgG1.

Fig. 4 ∣. T cell phenotypes associated with B cell help are linked to T1D.

Fig. 4 ∣

Elevated levels of T cells with a follicular helper (TFH) phenotype and increased IL-21 production by T cells have been noted in mouse models and individuals with T1D (ref. 101). In mouse models of diabetes, insulin-specific T cells can exhibit a TFH phenotype and provide help to B cells that mount insulin-specific antibody responses231. Insulin-specific T cells with a TFH phenotype have also been documented in individuals who have recently developed islet-targeted autoantibodies232. T cells with a T peripheral helper (TPH) phenotype are also increased in individuals at onset of T1D and in at-risk individuals who go on to develop diabetes100. Like TFH cells, TPH cells expresses inducible T cell costimulator (ICOS) and PD1 but lack expression of CXCR5 and instead can express chemokine receptors associated with migration to inflammatory sites (for example, CCR2 and CCR5). Whether TPH cells recognizes T1D-associated antigens such as insulin remains unclear. TCR, T cell receptor.

Both TFH and TPH cells interact with B cells. Tertiary lymphoid organ (TLO)-like structures, which are aggregates of T cells and B cells that can be organized to some extent by tissue stroma102, have been identified in the pancreas of NOD mice103, but the presence of these structures in human pancreases has been uncertain. Recently, a study of organ donors has shown that out of 21 individuals with T1D who had insulitis, 12 had evidence of pancreatic TLOs. Curiously, the mean age of diagnosis was significantly lower in donors with TLOs than in those without, and well-organized TLOs were only found in individuals with very short disease duration. TLOs were also identified in three out of six autoantibody-positive non-diabetic donors examined104.

CD8+ T cells

Autoantigen-reactive CD8+ T cells can be identified in the pancreas and peripheral blood of individuals with T1D (Fig. 3b), as well as in the peripheral blood of control participants105-107 (Table 1). There is an enrichment of islet autoantigen (such as ZnT8186–194)-reactive CD8+ T cells in the pancreas of T1D donors in spite of the similar frequency in blood from individuals with T1D versus non-diabetic controls108. The proportion of patients with islet-infiltrating preproinsulin (PPI)-reactive CD8+ T cells is estimated to be as high as 60–70%, and these cells can kill human β-cells in vitro109,110. Interestingly, CD8+ T cells with different autoantigen specificities can be found within the same islet, particularly in patients with long-standing disease111.

The differentiation state of islet-reactive CD8+ T cells in the peripheral blood has been investigated in several studies. Notably, a 2-year longitudinal study in children under 12 years of age with T1D has found that the frequency of CD57+ effector memory islet-specific CD8+ T cells declined as C-peptide levels fell, consistent with a role for antigen load in promoting the antigen-specific T cell response112. An exhaustion-like profile, with expression of EOMES, 2B4, PD1, TIGIT and CD160, was associated with slow disease progression113. Compared with responses to conventional antigens such as viral antigens, the interactions of CD8+ T cells with autoantigens displays lower avidity but also longer persistence. An analysis of the epigenome of T1D-specific autoantigen-reactive CD8+ T cells (identified with class I MHC tetramers) revealed stem-like epigenetic features, which are postulated to account for the longevity of these cells despite constant exposure to autoantigen114. Consistent with this, autoantigen-reactive CD8+ T cells in the pancreatic draining lymph node in NOD mice exhibited a high self-renewal capacity with the ability to differentiate into short-lived effector cells that can destroy β-cells115. Because islet-reactive CD8+ T cells are found in healthy individuals108,113,116, how these cells are triggered to initiate immune pathology is an open question. Interestingly, commensal bacteria within the gut microbiome can represent a source of antigenic peptides that cross-react with IGRP-specific CD8+ T cells, suggesting a mechanism by which the microbiome might modulate the course of the disease117-119.

Crosstalk between β-cells and immune cells

There is evidence that crosstalk between immune cells and β-cells contributes to T1D pathogenesis because β-cells release insulin peptide fragments into the circulation that can stimulate CD4+ T cells, even at distant sites120. Pro-inflammatory cytokines (such as IFNγ and TNF) can induce the expression of class I and II HLA molecules on β-cells121. The hyperexpression of HLA molecules in pancreatic islets of individuals with T1D, which is thought to enhance islet antigen recognition, was first described over 40 years ago122,123. At early stages of the disease, this may involve IFNα acting via a TYK2-STAT2-IRF9 axis, and at later stages, IFNγ-driven STAT1 activation may contribute to high levels of HLA expression69,72,124.

Dying or senescent β-cells provide a source of antigen for autoreactive T cells125. Inflammatory conditions in the pancreas induce the dioxygenase TET2 in β-cells, and Tet2-deficient mouse β-cells are resistant to killing by T cells126. Separately, thioredoxin-interacting protein (TXNIP) can mediate oxidative stress, inhibit cell proliferation and induce apoptosis by inhibiting the thioredoxin system. Elevated levels of TXNIP are found in human diabetic islets and induce β-cell apoptosis, whereas β-cell-specific TXNIP deficiency protected against diabetes in NOD and streptozotocin-induced mouse models127. The calcium channel blocker verapamil can inhibit TXNIP and was shown to have beneficial effects on β-cell function in individuals with newly diagnosed T1D (ref. 128). Treatment with verapamil reduced the levels of chromogranin, a β-cell autoantigen, and was suggested to decrease the levels of circulating pro-inflammatory TFH cells129.

Islet proteins may undergo various types of modifications, leading to the emergence of neoantigens that can be recognized by T cells130,131. For example, CD4+ T cells from a patient with T1D were found to be reactive to an oxidized epitope of the insulin A-chain containing a novel disulfide bridge132. Another type of post-translational modification that occurs in β-cells is represented by hybrid insulin peptides (HIPs), which are proinsulin fragments fused to other β-cell peptides through peptide bonds (Fig. 5). The presence of HIPs in human and mouse pancreatic islets has been validated through mass spectrometric analyses120,133-135. HIPs differ from conventionally modified epitopes in that they encompass entire amino acid sequences not found in the genome. This results in the introduction of fresh contact residues for interactions with both MHC and T cell receptors (TCRs). Significantly elevated levels of pro-inflammatory (IFNγ-secreting) T cells targeting HIPs have been detected in the peripheral blood of individuals with recent-onset T1D, setting them apart from non-diabetic control subjects136,137. HIP-reactive T cells have also been detected in residual pancreatic islets of organ donors with T1D (refs. 88,135,136). Furthermore, various diabetes-triggering CD4+ T cell clones that target HIPs have been identified in NOD mice138-140. In experiments using NOD mice engineered to possess only a monoclonal population of T cells, it was revealed that only a small subset of 17 autoantigen-specific TCRs tested had the capability to initiate islet infiltration and β-cell destruction independently141. Notably, pathogenic TCRs were derived from the CD4 T cell clones BDC-2.5, NY-4.1 and BDC-6.9, all of which respond to HIPs in which a common C-peptide fragment is linked to different β-cell peptides135,138,140. Another pathogenic TCR was derived from BDC-10.1, which targets the same HIP as BDC-2.5 (Fig. 5). Lastly, a TCR responsive to the unmodified insulin B:9–23 epitope was also identified as pathogenic; however, its potency was lower, resulting in diabetes in only 33% of the mice, compared with the 100% incidence for both BDC-2.5 and BDC-10.1, as well as the 71% and 56% observed for NY4.1 and BDC-6.9, respectively. The remaining TCRs which were focused on epitopes derived from GAD65, IA2 and phogrin (IA2-beta) were not independently pathogenic. These findings highlight that autoreactivity does not equate to pathogenicity141 and that a distinct group of HIPs (2.5HIP, 6.9HIP and HIP11; see Fig. 5) are targeted by the most pathogenic CD4+ T cell clones in NOD mice. Recent data established that these pathogenic HIPs are generated owing to the reversed proteolytic action of aspartic protease cathepsin D (CatD)142, suggesting that CatD may serve as a therapeutic target in T1D. Other modifications of insulin that have been identified as T cell epitopes in T1D include deamidations, introduced by tissue transglutaminase 2 and different forms of protein oxidation, induced by reactive oxygen species143,144. Further forms of post-translational modifications that are linked to disease pathogenesis in individuals with T1D are citrullination and carbonylation of epitopes derived from proteins other than insulin88,145,146. Lastly, defective ribosomal initiation products of proinsulin (DRIPs) have also been suggested as T cell epitopes in T1D (ref. 147).

Fig. 5 ∣. Disease-relevant hybrid insulin peptides (HIPs).

Fig. 5 ∣

The figure shows HIPs that were confidently identified from pancreatic islets by mass spectrometry and for which disease-relevant HIP-specific T cells were also identified in peripheral blood mononuclear cells. For example, the CD4+ T cell clone E2 was isolated from peripheral blood mononuclear cells of individuals with recent-onset T1D (ref. 136) or from residual islets of organ donors with T1D (such as T cells expressing the TCR GSE.8E3). Moreover, experiments in NOD mice identified various diabetes-triggering CD4+ T cell clones that target HIPs bearing a distinct C-peptide (InsC) fragment (ending in the amino acid sequence LAL) linked to the N-termini of peptides derived from chromogranin A (ChgA), proinsulin (C-peptide) or islet amyloid polypeptide (IAPP). These HIPs are formed by the aspartic protease cathepsin D through a reversed proteolytic transpeptidation reaction. Peptide sequences highlighted in blue originate from C-peptide, whereas sequences highlighted in red originate from ChgA, IAPP or InsC.

Failures of regulation: Treg cells in T1D

Several candidate genes associated with T1D, such as IL2RA, CTLA4, PTPN2 and PTPN22, have important functions in Treg cells. There is not a quantitative deficiency of Treg cells in T1D; however, a number of reports suggest that Treg cells in individuals with T1D may be dysfunctional and memory effector T cells from patients may be resistant to suppression148. Impaired IL-2R signalling has been suggested as a mechanism by which the metabolic and functional fitness of Treg cells is compromised in individuals with T1D (ref. 149). Defects in the IL-2R pathway and reduced phosphorylation of STAT5 may lead to insufficient FOXP3 expression in Treg cells14. Likewise, the production of IFNγ by Treg cells has been found and interpreted as a measure of Treg cell instability in T1D (ref. 150). Cytokines such as IL-12, IL-23 and IL-21 have also been reported to drive Treg cell lineage instability150,151, and there may be a preferential expansion of TFH cells over follicular Treg cells in secondary lymphoid organs152.

Immune modulation as a therapy for T1D

The insights into pathological immune responses in T1D have led to efforts to develop biomarkers of disease activity. Challenges remain because certain parameters that can easily be measured in the serum (such as autoantibodies and cytokines) are not the direct cause of β-cell killing, and the contributions of immune cells measured in the blood may not precisely reflect events in the pancreas. Clinical trials have, therefore, used clinical parameters for defining their entry criteria, but there is heterogeneity in the duration and progression of the disease between participants. This heterogeneity is captured in the idea of disease ‘endotypes’, which are based on immunological and demographic differences between patients and their responses to therapies – age being an important feature153. Many studies have enrolled patients with recent-onset stage 3 T1D, most frequently in early teenage years, with a primary end point based on stimulated C-peptide responses compared with placebo control groups (Fig. 6 and Box 1). In the sub-sections that follow, we provide selected highlights from this area and refer the reader to other reviews for more extensive coverage of the field154-156.

Fig. 6 ∣. Drugs and mechanisms that have shown efficacy in T1D.

Fig. 6 ∣

The targets of these therapies have included Innate and inflammatory mediators and pathways, B cells, costimulatory molecules, T cells and β-cells128,158,163,164,173,179,185,188,202,203. The principle targets of drug action are highlighted in red. The T cells depicted are generic and may include several different subsets (for example, CD4+,CD8+, TFH cells and others). mAb, monoclonal antibody.

Box 1. Road to development of therapies for the treatment and prevention of T1D.

  • Discovery stage: Concept development based on cellular mechanisms, genetics and tissue pathology. Targets for therapy, timing of therapy and adverse events are investigated. Preclinical models allow to study genetics and cellular mechanisms, as well as testing therapeutics for efficacy, timing and adverse events. NOD mice, transgenic NOD mice (such as BDC2.5), RIP LCMV transgenic mice and DO11 × RIP-mOVA mice are typically used. Moreover, human and mouse islets can be studied in vitro and ex vivo. For example, nPOD has provided pancreatic specimens from patients at-risk and those who have died with T1D (ref. 240).

  • Early-phase clinical studies: These studies of limited size aim to determine the safety, biological and clinical efficacy, proof of mechanism and early evidence for ‘prospect of benefit’. They are most often carried out in adults but may include children (healthy individuals or those with recent-onset stage 3 T1D). They can involve testing new drugs or repurposing of approved drugs such as abatacept and rituximab173,179.

  • Later-phase clinical studies: These studies test the preservation of β-cell function and clinical parameters and the diagnosis of stage 2 or 3 T1D. Individuals are typically diagnosed with new-onset T1D (around 100 days from diagnosis) and can include children. They also include smaller-size prevention studies in individuals with stage 1 or 2 T1D.

Therapies targeting innate immune pathways

Given the early role of innate immune cells in triggering autoimmune diabetes, cytokines and chemokines implicated in innate immune responses have been proposed as potential pharmacological targets. In a clinical study in children with new-onset T1D, etanercept, a fusion protein that binds TNF and blocks its activity, led to a reduction in haemoglobin A1c (HbA1c) and improved endogenous insulin responses157. Moreover, a highly successful trial of the TNF-targeting antibody golimumab showed improvement in C-peptide responses and clinical parameters when it was administered to participants aged 6–21 years with stage 3 T1D for ≤52 weeks158.

However, trials with agents that block IL-1β (such as the mAbs anakinra or canakinumab) have failed to show benefit159. Similarly, tocilizumab, a mAb that blocks IL-6R, did not slow β-cell loss160, nor did blocking CXCR1/2, which is important for neutrophil recruitment54,161,162. Imatinib, a small-molecule tyrosine kinase inhibitor that affects inflammatory pathways in immune cells and pancreatic β-cells, showed modest effects on C-peptide responses at 1 year163.

Because of their pleotropic effects on inflammatory pathways, there is a strong rationale for using JAK inhibitors to treat patients with T1D. Indeed, a pilot trial of baricitinib in patients with new-onset T1D has shown improvement in β-cell function after 1 year164.

Antigens as targets

Antigen-specific strategies such as vaccination have been tested in individuals with T1D and, given their general safety, have also been trialled in patients at risk for stage 3 T1D. Subcutaneous immunization with GAD-alum (Diamyd®), which is thought to modulate T cell responses to GAD65, initially showed improvement in stimulated C-peptide responses, but subsequent trials failed to reproduce this finding165-167. It has been suggested that intralymphatic administration of this antigen may be more effective168. Parenteral and oral administration of insulin have been trialled to test whether mucosal delivery of antigen affects responses, but these trials have failed to meet their end points169-171. In prevention studies, post hoc analyses have suggested that some at-risk individuals who have impaired pancreatic β-cell responses may experience some delay in diagnosis of clinical T1D when treated with oral insulin171. A therapeutic dose for orally administered antigens is challenging to identify: doses that are higher than those used in prevention studies have shown greater systemic antigen responses172.

B cell-directed immunotherapy

Given that preclinical data suggest a pathogenic role for B cells in T1D, therapies targeting this cell population have been investigated. A clinical trial testing rituximab, a CD20-targeted mAb that depletes B cells (four doses over 1 month), has been shown to preserve β-cell function and delay disease progression in individuals with new-onset T1D173. However, the benefit was short-lived174. The initial beneficial effects may be attributable to the ability of the drug to interrupt T cell-B cell collaboration, given that antibody responses to the T cell-dependent antigen phiX174 were blocked. However, the fact that the effects were temporary may be owing to the inability of transient B cell depletion to reset defective early B cell tolerance checkpoints, resulting in repletion with autoreactive B cells175. In addition, non-responders had a higher expression of T cell-associated gene expression modules after rituximab treatment, consistent with the idea that T cell responses also need to be controlled during B cell-directed immunotherapy176.

Costimulation blockade

A key control point for adaptive immune responses is the provision of costimulation through the T cell surface receptor CD28. Blocking CD28 costimulatory signals with CTLA-4Ig showed efficacy in NOD mice177. A clinical trial of the CTLA-4-Ig molecule abatacept, given at days 1, 14 and 28 and then monthly for 2 years in individuals with recent-onset stage 3 T1D, has shown a reduced decline in C-peptide levels compared with placebo178,179. Building on this success, a trial was conducted in patients at an earlier stage of the disease (stage 1), with the idea that earlier blockade of T cell activation would prove more effective. In this trial (TrialNet, TN18), abatacept was given monthly for 1 year, and although it showed a delay in the rate of progression, the result did not meet statistical significance (P = 0.11)180. In both trials, abatacept reduced the levels of activated TFH cells, but it also reduced the frequency of Treg cells that may have interfered with the efficacy and persistence of the effect101,181. The critical role of CD28 for the activation of effectors and regulators suggests that careful choice of timing, and possibly combination therapies (for example, co-administering low-dose IL-2 to preserve Treg cells), may be needed182.

Anti-thymocyte globulin

Broad targeting of T cells was tested with anti-thymocyte globulin (ATG), which is routinely used for the treatment of transplant rejection and consists of rabbit antibodies against human thymocytes. In individuals with T1D it was tested at reduced dosages in order to mitigate the known adverse effects of cytokine release syndrome. A 2-day course of low-dose ATG was shown to preserve C-peptide responses in individuals with established T1D in a pilot trial and in new-onset T1D in a subsequent placebo-controlled trial (TN19)183-186. Drug administration led to the depletion of CD4+ and CD8+ T cells. The way in which ATG causes long-term responses are yet unknown, but may have similarities to the effects of teplizumab on immune cells (discussed in the sub-sections that follow). Specifically, low-dose ATG appears to primarily impact CD4+ T cells and promote an exhaustion phenotype, while simultaneously preserving Treg cells187.

Alefacept

Two 12-week courses of the LFA3Ig fusion protein alefacept, which blocks T cell proliferation and activation, reduced loss of stimulated C-peptide responses in new-onset T1D. Importantly, there was also a significant reduction in the frequency of severe hypoglycaemia188. It depleted CD4+ and CD8+ central and effector memory T cells, but preserved Treg cells, thereby increasing the ratios of Treg cells to central and effector memory T cells. These findings supported the strategy of improving the balance between immune effectors and regulators for treatment.

Teplizumab

Teplizumab, a Fc-receptor non-binding humanized CD3-targeted mAb, was the first immunotherapy for T1D to receive FDA approval (November 2022). The earliest work examining the therapeutic potential of CD3-targeted mAbs began in the transplant field, in which the mAb OKT3 was shown to prevent allograft rejection. However, treatment-related cytokine release syndrome owing to the production of TNF by activated T cells and the development of anti-mouse antibodies limited enthusiasm for its translation in humans189-191. Early preclinical experiments in T1D using a modified CD3-targeted mAb consisting of F(ab′)2 fragments of the 145-2C11 mAb have shown that it could prevent the induction of autoimmune diabetes in a mouse model of T1D induced by low-dose streptozotocin, which kills pancreatic β-cells192. Another study has found that brief treatment of hyperglycaemic NOD mice with CD3-targeted mAbs (with either the whole antibody or F(ab′)2 fragments) can reverse the disease and, importantly, that the disease did not recur after drug treatment was discontinued193,194. This observation highlighted the reversibility of the disease even at a time when hyperglycaemia had set in, which was subsequently shown to entail the recovery of β-cell granulation in cells that were non-functional, but not destroyed, at diagnosis195. Two humanized FcR non-binding CD3-targeted mAbs, teplizumab and otelixizumab, were produced and each contains two alanines instead of leucines in the Fc portion of the IgG1 molecule196. In a randomized controlled phase II trial of 42 patients, a single 12-day or 14-day course of teplizumab was shown to improve C-peptide responses to a mixed meal even 2 years after treatment, and without the severe cytokine release syndrome that was routinely observed with OKT3 (refs. 197,198). Otelixizumab was tested in a randomized placebo-controlled trial of 80 patients and also showed clinical benefit as reflected by reduced insulin requirements 18 months after the single course of treatment shortly after diagnosis199. Importantly, like the preclinical studies and unlike continuous immune therapies in the past, the sustained efficacy did not require continuous administration.

A randomized phase II trial (AbATE), sponsored by the Immune Tolerance Network, tested the effects of two courses of teplizumab, 1 year apart, on β-cell function after 2 years200. At the same time, a phase III industry-sponsored trial (Protégé) was initiated, in which teplizumab or placebo was given at varying dosages, at diagnosis and at 6 months, with a second (Protégé Encore) trial initiated afterwards. The Protégé trial used a new composite primary end point of insulin use and HbA1c levels. Despite not meeting the composite end point, β-cell function (as measured by stimulated C-peptide response) was improved with teplizumab compared with placebo, confirming the results from earlier trials. A subsequent study has tested treatment of patients more than 4 months after diagnosis, but in whom clinically significant levels of β-cell function could be detected (defined as a stimulated C-peptide level of at least 0.2 pmol ml−1). This trial also confirmed improved effects of the drug on β-cell function, although therapeutic outcomes were not as favourable as those observed in new-onset patients201. A combined analysis of data from five studies of teplizumab in patients with stage 3 T1D has shown consistent improvement in stimulated C-peptide responses and reduced insulin usage202. Furthermore, a recently completed phase III clinical trial (PROTECT) of children and adolescents with new-onset T1D has confirmed these earlier findings on C-peptide preservation at 18 months after enrolment and suggested improvements in insulin use, continuous glucose metrics, hypoglycaemia and patient reported outcomes203.

The TN10 study, conducted by the National Institute of Diabetes Digestive and Kidney Diseases T1D TrialNet, tested whether a single course of teplizumab would delay or prevent progression to clinical disease in individuals with pre-diabetes, defined as stage 2 T1D with a high-risk progression to stage 3 T1D. The median time from enrolment to diagnosis with stage 3 T1D was 27 months in placebo-treated patients, which was delayed by approximately 2 years with teplizumab treatment3,204. However, some participants were not diagnosed with T1D for much longer periods of time (that is, >10 years after treatment). In this study, stimulated C-peptide responses, which were lower in this at-risk cohort than in healthy age-matched individuals, improved rapidly after treatment3,205.

The mechanism of action of teplizumab is attributable to the nature of the partial agonist signal that is delivered to T cells (Fig. 7). These findings, based on studies of peripheral blood cells from treated patients, extend the observational studies that slow rates of spontaneous disease in at-risk individuals correlate with T cell exhaustion signatures identified in treated patients by expression of an EOMES signature and the expression of TIGIT and KLRG1 on CD8+ T cells206.

Fig. 7 ∣. Mechanism of action of the CD3-targeting monoclonal antibody teplizumab.

Fig. 7 ∣

Early studies in animal models of the Fc receptor (FcR) non-binding monoclonal antibody (mAb) teplizumab have shown that by binding CD3, it induces a partial T cell receptor signal that induces clonal anergy and, thereby, inhibits β-cell killing233. In vitro and in vivo experiments have demonstrated that it induces a relative expansion of CD8+ T cells and the secretion of IL-10; similar results were also found with a FcR non-binding version of the CD3-targeted mAb. Moreover, teplizumab induced a transient decline in circulating lymphocytes, which reflected the migration of T cells to the gut wall, wherein activation signals led to the induction of CD4+TGFβ+ or CD8+IL-10+ T cells234,235. Moreover, FcR non-binding CD3-targeted mAbs induced a selective preservation of Treg cells, and another study has reported that teplizumab induced TGF β+ Treg cells that have a role in the restoration of self-tolerance in the pancreatic draining lymph nodes236,237. a, Observations from the initial clinical trials showed activation of CD8+ T cells in vivo, and a transcriptome analysis of bulk RNA from peripheral blood mononuclear cells (PBMCs) from clinical responders in the AbATE trial, showed changes in CD8+ T cells that were later found to involve the induction of the transcription factor EOMES and increased expression of the receptors KLRG1 and TIGIT, suggesting that the cells were ‘partially exhausted’197,198,238. b, A long-term follow-up study of responders from the AbATE trial has shown an increased expression of PD1 on CD8+ T cells and an increase in PD1+ central memory CD8+ T cells and anergic CD8+ T cells, which may explain the long-term effects of teplizumab239. Autoantigen-reactive T cell receptors from CD8+ T cells have weak avidity for the antigen–MHC complex. The partial agonist signal delivered by the FcR non-binding CD3-targeted mAb may preferentially affect TCR signalling in response to such weak interactions, given that responses to conventional antigens such as viral antigens recover quickly after drug administration. APC, antigen-presenting cell.

Augmenting immune regulatory function

To enhance the number and function of Treg cells, a clinical trial was initiated to test a combination of sub-cutaneous administration of IL-2 (4.5 × 106 IU three times per week for 1 month) and rapamycin. However, it showed a deterioration of C-peptide responses along with expansion of NK cells, CD8+ effector cells and eosinophils207. Other groups have carefully titrated the dose of recombinant human IL-2 (rhIL-2) in order to selectively enhance the expansion of Treg cells without expanding NK cells or CD8+ effector T cells208,209. In a phase I–II trial, a low dose of rhIL-2 was associated with C-peptide preservation for a small subgroup of study participants210. A more direct approach to harness Treg cells involves cellular therapy, which was enabled by technological advances in expanding Treg cells ex vivo211. In a pilot trial, Treg cell adoptive cellular therapy was given alone or with rhIL-2 to enhance Treg cell survival. The infusions were considered safe but the combination trial with rhIL-2 was stopped because of transient worsening of C-peptide responses. Mechanistic studies have shown expansion of CD8+ effector T cells in patients receiving rhIL-2 (refs. 212,213). However, a subsequent phase III trial of expanded autologous Treg cells failed to meet its clinical end point of improvement in stimulated C-peptide responses at 1 year212,213. Redirecting T cell specificity through TCR gene transfer or by using chimeric antigen receptor expression may be required to improve the efficacy and safety of this approach. Moreover, strategies to improve the survival and stability of transferred cells are also being developed214.

Conclusions and future directions

Decades of mechanistic research are beginning to bear fruit in the quest to modulate the autoimmune response to pancreatic β-cells in T1D, as highlighted by the approval of teplizumab. Progress has been made in understanding the differentiation state of pathological CD4+ and CD8+ T cells and the nature of the autoantigens that drive the disease. More remains to be learned about the mechanisms that underpin stage 1 and stage 2 T1D and the molecular pathways and environmental influences that promote disease progression. We anticipate that the application of high-parameter imaging techniques to pancreatic tissue samples from patients or mice with T1D will allow new insights into the identity and phenotype of islet-infiltrating immune cell populations. The results of clinical studies have highlighted several areas that could be further developed.

Importantly, trials to date have targeted single molecules or cell types. The failure of current therapies to provide long-term protection suggests that combinations or repeated administration of therapeutic agents are needed. For example, targeting B cells, TFH cells and IL-21 production simultaneously may prevent the activation of pathogenic effector CD8+ T more efficiently than targeting single components, but such an approach has not yet been trialled215. Mechanistic studies of samples from individuals with T1D who fail to respond to immune-targeted drugs, or in whom the efficacy of successful drugs wanes, have suggested rational combinations of drugs to prevent disease recurrence – for example, the T cell signature associated with poor response to rituximab (see in the previous section) has led to a clinical trial in which abatacept is given after rituximab176,216. The recent success of the JAK inhibitor baricitinib in patients with recent-onset T1D may involve the inhibition of inflammatory pathways involved in immune cell activation, but it may also prevent the damaging effects of inflammatory cytokines directly on β-cells217. Agents to improve β-cell function, expand their mass or stabilize glycaemia, when combined with an immune therapy, may achieve the desired metabolic control that occurs only with functioning endogenous β-cells. A combination of an agent to arrest the destructive immune response that occurs at or prior to diagnosis, followed by a more specific treatment that can be administered repeatedly, such as an antigen that is delivered in a tolerogenic manner, may represent a safe approach to sustain endogenous β-cell function. Therapies with smaller target cell populations, including antigen-specific therapies, may allow repeated administration with acceptable safety profiles.

Not all therapies have or would be expected to work in all patients: genetic, demographic and immune markers may be developed to optimize the choice of drugs for individuals. In addition, the timing of interventions may be important. Different therapies may work maximally at different disease stages, and delayed intervention may present the added challenge of memory responses. It is important to consider that even in patients at stage 1 T1D, an auto-immune response has already been initiated. Primary prevention is clearly an important goal; however, testing such approaches would require very large enrolment, a very safe agent and long follow-up. Nonetheless, with the approval of the first immune-cell targeted therapy to change the course of T1D, a new frontier has been opened.

Acknowledgements

K.C.H. acknowledges support from the US National Institutes of Health (NIH) through grant numbers DK057846, DK129523, DK045735, AI66387, DK106993 (Type 1 Diabetes TrialNet) and CA2277473. KCH and NB received funding from T32AI155317. A.L.P. also acknowledges support from NIH through grant numbers DK106993 (Type 1 Diabetes TrialNet) and CA215110. T.M.B. acknowledges support from the National Institutes of Allergy and Infectious Diseases (NIAID) through grant number P01AI042288 and National Institute of Diabetes Digestive and Kidney Diseases through grant numbers DK106191 and DK122638. L.S.K.W. acknowledges support from the Medical Research Council (MR/N001435/1), Wellcome Trust (220772/Z/20/Z), Diabetes UK (20/0006172) and Connect Immune Research (22931). We are grateful to Natalie Edner for the assistance with figure preparation.

Footnotes

Competing interests

K.C.H. is a co-inventor on a patent for the use of teplizumab to delay type 1 diabetes and has consulted for Sanofi, Provention Bio, GSK, Abata, Gentibio and Idorsia. He is on the scientific advisory board for Sonoma and NexImmune.

References

  • 1.Gregory GA et al. Global incidence, prevalence, and mortality of type 1 diabetes in 2021 with projection to 2040: a modelling study. Lancet Diabetes Endocrinol. 10, 741–760 (2022). [DOI] [PubMed] [Google Scholar]
  • 2.Evans-Molina C et al. β Cell dysfunction exists more than 5 years before type 1 diabetes diagnosis. JCI Insight 3, e120877 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Sims EK et al. Teplizumab improves and stabilizes beta cell function in antibody-positive high-risk individuals. Sci. Transl. Med 13, eabc8980 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Turtinen M. et al. Characteristics of familial type 1 diabetes: effects of the relationship to the affected family member on phenotype and genotype at diagnosis. Diabetologia 62, 2025–2039 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Redondo MJ, Steck AK & Pugliese A. Genetics of type 1 diabetes. Pediatr. Diabetes 19, 346–353 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mrena S. et al. Models for predicting type 1 diabetes in siblings of affected children. Diabetes Care 29, 662–667 (2006). [DOI] [PubMed] [Google Scholar]
  • 7.Dorman JS et al. Type 1 diabetes in offspring of parents with type 1 diabetes: the tip of an autoimmune iceberg? Pediatr. Diabetes 1, 17–22 (2000). [DOI] [PubMed] [Google Scholar]
  • 8.Noble JA & Valdes AM Genetics of the HLA region in the prediction of type 1 diabetes. Curr. Diabetes Rep 11, 533–542 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Aly TA et al. Genetic prediction of autoimmunity: initial oligogenic prediction of anti-islet autoimmunity amongst DR3/DR4-DQ8 relatives of patients with type 1A diabetes. J. Autoimmun 25, 40–45 (2005). [DOI] [PubMed] [Google Scholar]
  • 10.Astill TP, Ellis RJ, Arif S, Tree TI & Peakman M Promiscuous binding of proinsulin peptides to type 1 diabetes-permissive and -protective HLA class II molecules. Diabetologia 46, 496–503 (2003). [DOI] [PubMed] [Google Scholar]
  • 11. Todd JA, Bell JI & McDevitt HO HLA-DQ beta gene contributes to susceptibility and resistance to insulin-dependent diabetes mellitus. Nature 329, 599–604 (1987). This study established that the amino acid sequence of the HLA-DQβ chain is linked to predisposition to T1D in a manner dependent on the identity of amino acid residue 57.
  • 12.Gioia L et al. Position beta57 of I-A(g7) controls early anti-insulin responses in NOD mice, linking an MHC susceptibility allele to type 1 diabetes onset. Sci. Immunol 4, eaaw6329 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Robertson CC et al. Fine-mapping, trans-ancestral and genomic analyses identify causal variants, cells, genes and drug targets for type 1 diabetes. Nat. Genet 53, 962–971 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Garg G. et al. Type 1 diabetes-associated IL2RA variation lowers IL-2 signaling and contributes to diminished CD4+CD25+ regulatory T cell function. J. Immunol 188, 4644–4653 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pugliese A. et al. The insulin gene is transcribed in the human thymus and transcription levels correlated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nat. Genet 15, 293–297 (1997). [DOI] [PubMed] [Google Scholar]
  • 16.Sabater L. et al. Insulin alleles and autoimmune regulator (AIRE) gene expression both influence insulin expression in the thymus. J. Autoimmun 25, 312–318 (2005). [DOI] [PubMed] [Google Scholar]
  • 17.Vafiadis P. et al. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nat. Genet 15, 289–292 (1997). [DOI] [PubMed] [Google Scholar]
  • 18.Oakey H. et al. Protocol for a nested case-control study design for omics investigations in the Environmental Determinants of Islet Autoimmunity cohort. Ann. Med 55, 2198255 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ziegler AG et al. Primary prevention of beta-cell autoimmunity and type 1 diabetes — the Global Platform for the Prevention of Autoimmune Diabetes (GPPAD) perspectives. Mol. Metab 5, 255–262 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Krischer JP et al. Genetic and environmental interactions modify the risk of diabetes-related autoimmunity by 6 years of age: the TEDDY study. Diabetes Care 40, 1194–1202 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rewers M. et al. Newborn screening for HLA markers associated with IDDM: Diabetes Autoimmunity Study in the Young (DAISY). Diabetologia 39, 807–812 (1996). [DOI] [PubMed] [Google Scholar]
  • 22.Bach JF Autoimmune diseases as the loss of active “self-control”. Ann. N. Y. Acad. Sci 998, 161–177 (2003). [DOI] [PubMed] [Google Scholar]
  • 23.Gepts W. Pathologic anatomy of the pancreas in juvenile diabetes mellitus. Diabetes 14, 619–633 (1965). [DOI] [PubMed] [Google Scholar]
  • 24.Krogvold L. et al. Detection of a low-grade enteroviral infection in the islets of Langerhans of living patients newly diagnosed with type 1 diabetes. Diabetes 64, 1682–1687 (2015). [DOI] [PubMed] [Google Scholar]
  • 25.Oikarinen S. et al. Characterisation of enterovirus RNA detected in the pancreas and other specimens of live patients with newly diagnosed type 1 diabetes in the DiViD study. Diabetologia 64, 2491–2501 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Nekoua MP, Alidjinou EK & Hober D Persistent coxsackievirus B infection and pathogenesis of type 1 diabetes mellitus. Nat. Rev. Endocrinol 18, 503–516 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Rodriguez-Calvo T Enterovirus infection and type 1 diabetes: unraveling the crime scene. Clin. Exp. Immunol 195, 15–24 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Krogvold L. et al. Pleconaril and ribavirin in new-onset type 1 diabetes: a phase 2 randomized trial. Nat. Med 29, 2902–2908 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kamrath C. et al. Incidence of type 1 diabetes in children and adolescents during the COVID-19 pandemic in Germany: results from the DPV Registry. Diabetes Care 45, 1762–1771 (2022). [DOI] [PubMed] [Google Scholar]
  • 30.D’Souza D. et al. Incidence of diabetes in children and adolescents during the COVID-19 pandemic: a systematic review and meta-analysis. JAMA Netw. Open 6, e2321281 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Hippich M. et al. A public health antibody screening indicates a 6-fold higher SARS-CoV-2 exposure rate than reported cases in children. Med 2, 149–163 e144 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Rewers M. et al. SARS-CoV-2 infections and presymptomatic type 1 diabetes autoimmunity in children and adolescents from Colorado, USA, and Bavaria, Germany. JAMA 328, 1252–1255 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Del Chierico F et al. Pathophysiology of type 1 diabetes and gut microbiota role. Int. J. Mol. Sci 23, 14650 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Morse ZJ, Simister RL, Crowe SA, Horwitz MS & Osborne LC Virus induced dysbiosis promotes type 1 diabetes onset. Front. Immunol 14, 1096323 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gavin PG, Kim KW, Craig ME, Hill MM & Hamilton-Williams EE Multi-omic interactions in the gut of children at the onset of islet autoimmunity. Microbiome 10, 230 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Paun A. et al. Association of HLA-dependent islet autoimmunity with systemic antibody responses to intestinal commensal bacteria in children. Sci. Immunol 4, eaau8125 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rouxel O. et al. Cytotoxic and regulatory roles of mucosal-associated invariant T cells in type 1 diabetes. Nat. Immunol 18, 1321–1331 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Nel I. et al. MAIT cell alterations in adults with recent-onset and long-term type 1 diabetes. Diabetologia 64, 2306–2321 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gulden E. et al. Microbiota control immune regulation in humanized mice. JCI Insight 2, e91709 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sun L. et al. Two to tango: dialogue between adaptive and innate immunity in type 1 diabetes. J. Diabetes Res 2020, 4106518 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ferris ST et al. A minor subset of Batf3-dependent antigen-presenting cells in islets of Langerhans is essential for the development of autoimmune diabetes. Immunity 41, 657–669 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zirpel H & Roep BO Islet-resident dendritic cells and macrophages in type 1 diabetes: in search of bigfoot’s print. Front. Endocrinol 12, 666795 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Citro A, Campo F, Dugnani E & Piemonti L Innate immunity mediated inflammation and beta cell function: neighbors or enemies? Front. Endocrinol 11, 606332 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Calderon B. et al. The pancreas anatomy conditions the origin and properties of resident macrophages. J. Exp. Med 212, 1497–1512 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zakharov PN, Hu H, Wan X & Unanue ER Single-cell RNA sequencing of murine islets shows high cellular complexity at all stages of autoimmune diabetes. J. Exp. Med 217, e20192362 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Carrero JA et al. Resident macrophages of pancreatic islets have a seminal role in the initiation of autoimmune diabetes of NOD mice. Proc. Natl Acad. Sci. USA 114, E10418–E10427 (2017). This paper demonstrates the essential role of islet-resident macrophages in the development of autoimmune diabetes in NOD mice.
  • 47.Dror E. et al. Postprandial macrophage-derived IL-1beta stimulates insulin, and both synergistically promote glucose disposal and inflammation. Nat. Immunol 18, 283–292 (2017). [DOI] [PubMed] [Google Scholar]
  • 48.Alleva DG, Pavlovich RP, Grant C, Kaser SB & Beller DI Aberrant macrophage cytokine production is a conserved feature among autoimmune-prone mouse strains: elevated interleukin (IL)-12 and an imbalance in tumor necrosis factor-alpha and IL-10 define a unique cytokine profile in macrophages from young nonobese diabetic mice. Diabetes 49, 1106–1115 (2000). [DOI] [PubMed] [Google Scholar]
  • 49.Calderon B, Carrero JA, Miller MJ & Unanue ER Entry of diabetogenic T cells into islets induces changes that lead to amplification of the cellular response. Proc. Natl Acad. Sci. USA 108, 1567–1572 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hu H, Zakharov PN, Peterson OJ & Unanue ER Cytocidal macrophages in symbiosis with CD4 and CD8 T cells cause acute diabetes following checkpoint blockade of PD-1 in NOD mice. Proc. Natl Acad. Sci. USA 117, 31319–31330 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Vecchio F. et al. Abnormal neutrophil signature in the blood and pancreas of presymptomatic and symptomatic type 1 diabetes. JCI Insight 3, e122146 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Huang J. et al. Distinct neutrophil counts and functions in newly diagnosed type 1 diabetes, latent autoimmune diabetes in adults, and type 2 diabetes. Diabetes Metab. Res. Rev 35, e3064 (2019). [DOI] [PubMed] [Google Scholar]
  • 53. Diana J. et al. Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat. Med 19, 65–73 (2013). This study highlights the essential role of innate immune cells and their crosstalk in the initiation of autoimmune diabetes.
  • 54.Diana J & Lehuen A Macrophages and beta-cells are responsible for CXCR2-mediated neutrophil infiltration of the pancreas during autoimmune diabetes. EMBO Mol. Med 6, 1090–1104 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Battaglia M, Petrelli A & Vecchio F Neutrophils and type 1 diabetes: current knowledge and suggested future directions. Curr. Opin. Endocrinol. Diabetes Obes 26, 201–206 (2019). [DOI] [PubMed] [Google Scholar]
  • 56.Valle A. et al. Reduction of circulating neutrophils precedes and accompanies type 1 diabetes. Diabetes 62, 2072–2077 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.de Boer P. et al. Large-scale electron microscopy database for human type 1 diabetes. Nat. Commun 11, 2475 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gardner G & Fraker CA Natural killer cells as key mediators in type I diabetes immunopathology. Front. Immunol 12, 722979 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Poirot L, Benoist C & Mathis D Natural killer cells distinguish innocuous and destructive forms of pancreatic islet autoimmunity. Proc. Natl Acad. Sci. USA 101, 8102–8107 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hussain MJ et al. Elevated serum levels of macrophage-derived cytokines precede and accompany the onset of IDDM. Diabetologia 39, 60–69 (1996). [DOI] [PubMed] [Google Scholar]
  • 61.Bradshaw EM et al. Monocytes from patients with type 1 diabetes spontaneously secrete proinflammatory cytokines inducing Th17 cells. J. Immunol 183, 4432–4439 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Apaolaza PS et al. Islet expression of type I interferon response sensors is associated with immune infiltration and viral infection in type 1 diabetes. Sci. Adv 7, eabd6527 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Li Q. et al. Interferon-alpha initiates type 1 diabetes in nonobese diabetic mice. Proc. Natl Acad. Sci. USA 105, 12439–12444 (2008). This study is one of the seminal papers demonstrating the role of IFNα as an initiator of T1D.
  • 64.Rodrigues KB et al. Innate immune stimulation of whole blood reveals IFN-1 hyper-responsiveness in type 1 diabetes. Diabetologia 63, 1576–1587 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kallionpaa H. et al. Innate immune activity is detected prior to seroconversion in children with HLA-conferred type 1 diabetes susceptibility. Diabetes 63, 2402–2414 (2014). [DOI] [PubMed] [Google Scholar]
  • 66.Ferreira RC et al. A type I interferon transcriptional signature precedes autoimmunity in children genetically at risk for type 1 diabetes. Diabetes 63, 2538–2550 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lombardi A, Tsomos E, Hammerstad SS & Tomer Y Interferon alpha: the key trigger of type 1 diabetes. J. Autoimmun 94, 7–15 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Lundberg M, Krogvold L, Kuric E, Dahl-Jorgensen K & Skog O Expression of interferon-stimulated genes in insulitic pancreatic islets of patients recently diagnosed with type 1 diabetes. Diabetes 65, 3104–3110 (2016). [DOI] [PubMed] [Google Scholar]
  • 69.Marroqui L. et al. Interferon-alpha mediates human beta cell HLA class I overexpression, endoplasmic reticulum stress and apoptosis, three hallmarks of early human type 1 diabetes. Diabetologia 60, 656–667 (2017). [DOI] [PubMed] [Google Scholar]
  • 70.Lombardi A & Tomer Y Interferon alpha impairs insulin production in human beta cells via endoplasmic reticulum stress. J. Autoimmun 80, 48–55 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Coomans de Brachene A et al. IFN-alpha induces a preferential long-lasting expression of MHC class I in human pancreatic beta cells. Diabetologia 61, 636–640 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chandra V. et al. The type 1 diabetes gene TYK2 regulates β-cell development and its responses to interferon-α. Nat. Commun 13, 6363 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Ziegler AG et al. Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. JAMA 309, 2473–2479 (2013). This is a landmark study establishing the link between multiple islet autoantibody positivity and risk of progressing to stage 3 diabetes.
  • 74.Krischer JP et al. Predicting islet cell autoimmunity and type 1 diabetes: an 8-year TEDDY study progress report. Diabetes Care 41, 1051–1060 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kwon BC et al. Progression of type 1 diabetes from latency to symptomatic disease is predicted by distinct autoimmune trajectories. Nat. Commun 13, 1514 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Redondo MJ et al. A type 1 diabetes genetic risk score predicts progression of islet autoimmunity and development of type 1 diabetes in individuals at risk. Diabetes Care 41, 1887–1894 (2018). This representative study capitalizes on the ability to create polygenic risk scores for population screening efforts to identify at-risk subjects and those with distinct HLA genotypes or risk variants for pathway targeted therapies.
  • 77.Warncke K. et al. Elevations in blood glucose before and after the appearance of islet autoantibodies in children. J. Clin. Invest 132, e162123 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wong FS et al. Investigation of the role of B-cells in type 1 diabetes in the NOD mouse. Diabetes 53, 2581–2587 (2004). [DOI] [PubMed] [Google Scholar]
  • 79.Hu CY et al. Treatment with CD20-specific antibody prevents and reverses autoimmune diabetes in mice. J. Clin. Invest 117, 3857–3867 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Silveira PA et al. The preferential ability of B lymphocytes to act as diabetogenic APC in NOD mice depends on expression of self-antigen-specific immunoglobulin receptors. Eur. J. Immunol 32, 3657–3666 (2002). [DOI] [PubMed] [Google Scholar]
  • 81.Hulbert C, Riseili B, Rojas M & Thomas JW B cell specificity contributes to the outcome of diabetes in nonobese diabetic mice. J. Immunol 167, 5535–5538 (2001). [DOI] [PubMed] [Google Scholar]
  • 82.Smith MJ et al. Loss of anergic B cells in prediabetic and new-onset type 1 diabetic patients. Diabetes 64, 1703–1712 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Leete P. et al. Studies of insulin and proinsulin in pancreas and serum support the existence of aetiopathological endotypes of type 1 diabetes associated with age at diagnosis. Diabetologia 63, 1258–1267 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Campbell-Thompson ML et al. The diagnosis of insulitis in human type 1 diabetes. Diabetologia 56, 2541–2543 (2013). [DOI] [PubMed] [Google Scholar]
  • 85.Damond N. et al. A map of human type 1 diabetes progression by imaging mass cytometry. Cell Metab. 29, 755–768 e755 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Willcox A, Richardson SJ, Bone AJ, Foulis AK & Morgan NG Analysis of islet inflammation in human type 1 diabetes. Clin. Exp. Immunol 155, 173–181 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Campbell-Thompson M. et al. Insulitis and beta-cell mass in the natural history of type 1. Diabetes 65, 719–731 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Babon JA et al. Analysis of self-antigen specificity of islet-infiltrating T cells from human donors with type 1 diabetes. Nat. Med 22, 1482–1487 (2016). This study identified several autoreactive T cells directly isolated from organ donors with T1D.
  • 89.Landry LG et al. Proinsulin-reactive CD4 T cells in the islets of type 1 diabetes organ donors. Front. Endocrinol 12, 622647 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Pathiraja V. et al. Proinsulin-specific, HLA-DQ8, and HLA-DQ8-transdimer-restricted CD4+ T cells infiltrate islets in type 1 diabetes. Diabetes 64, 172–182 (2015). [DOI] [PubMed] [Google Scholar]
  • 91.Nakayama M. et al. Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice. Nature 435, 220–223 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Yang J. et al. Antigen-specific T cell analysis reveals that active immune responses to beta cell antigens are focused on a unique set of epitopes. J. Immunol 199, 91–96 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Walker LS & von Herrath M CD4 T cell differentiation in type 1 diabetes. Clin. Exp. Immunol 183, 16–29 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Ferraro A et al. Expansion of Th17 cells and functional defects in T regulatory cells are key features of the pancreatic lymph nodes in patients with type 1 diabetes. Diabetes 60, 2903–2913 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Arif S. et al. Peripheral and islet interleukin-17 pathway activation characterizes human autoimmune diabetes and promotes cytokine-mediated beta-cell death. Diabetes 60, 2112–2119 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Kenefeck R. et al. Follicular helper T cell signature in type 1 diabetes. J. Clin. Invest 125, 292–303 (2015). This study demonstrated that TFH cells and IL-21 are overproduced in T1D, challenging the established TH1 paradigm.
  • 97.Ferreira RC et al. IL-21 production by CD4+ effector T cells and frequency of circulating follicular helper T cells are increased in type 1 diabetes patients. Diabetologia 58, 781–790 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Viisanen T. et al. Circulating CXCR5+PD-1+ICOS+ follicular T helper cells are increased close to the diagnosis of type 1 diabetes in children with multiple autoantibodies. Diabetes 66, 437–447 (2017). [DOI] [PubMed] [Google Scholar]
  • 99.Pinto AI, Smith J, Kissack MR, Hogg KG & Green EA Thymic B cell-mediated attack of thymic stroma precedes type 1 diabetes development. Front. Immunol 9, 1281 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ekman I. et al. Circulating CXCR5PD-1hi peripheral T helper cells are associated with progression to type 1 diabetes. Diabetologia 62, 1681–1688 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Edner NM et al. Follicular helper T cell profiles predict response to costimulation blockade in type 1 diabetes. Nat. Immunol 21, 1244–1255 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kendall PL, Yu G, Woodward EJ & Thomas JW Tertiary lymphoid structures in the pancreas promote selection of B lymphocytes in autoimmune diabetes. J. Immunol 178, 5643–5651 (2007). [DOI] [PubMed] [Google Scholar]
  • 103.Penaranda C, Tang Q, Ruddle NH & Bluestone JA Prevention of diabetes by FTY720-mediated stabilization of peri-islet tertiary lymphoid organs. Diabetes 59, 1461–1468 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Korpos E et al. Identification and characterisation of tertiary lymphoid organs in human type 1 diabetes. Diabetologia 64, 1626–1641 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Mallone R. et al. CD8+ T-cell responses identify beta-cell autoimmunity in human type 1 diabetes. Diabetes 56, 613–621 (2007). [DOI] [PubMed] [Google Scholar]
  • 106.Cerosaletti K. et al. Single-cell RNA sequencing reveals expanded clones of islet antigen-reactive CD4+ T cells in peripheral blood of subjects with type 1 diabetes. J. Immunol 199, 323–335 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Anderson AM et al. Human islet T cells are highly reactive to preproinsulin in type 1 diabetes. Proc. Natl Acad. Sci. USA 118, e2107208118 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Culina S. et al. Islet-reactive CD8+ T cell frequencies in the pancreas, but not in blood, distinguish type 1 diabetic patients from healthy donors. Sci. Immunol 3, eaao4013 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bender C, Rodriguez-Calvo T, Amirian N, Coppieters KT & von Herrath MG The healthy exocrine pancreas contains preproinsulin-specific CD8 T cells that attack islets in type 1 diabetes. Sci. Adv 6, eabc5586 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Skowera A. et al. CTLs are targeted to kill beta cells in patients with type 1 diabetes through recognition of a glucose-regulated preproinsulin epitope. J. Clin. Invest 118, 3390–3402 (2008). In this study, preproinsulin-specific CTLs that were capable of killing β-cells were isolated and characterized from peripheral blood of patients. The presence of these cells with a direct mechanism for β-cell killing suggests a pathological mechanism.
  • 111. Coppieters KT et al. Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from recent onset and long-term type 1 diabetes patients. J. Exp. Med 209, 51–60 (2012). This is a seminal paper showing direct HLA multimer staining of in situ autoreactive T cells.
  • 112.Yeo L. et al. Autoreactive T effector memory differentiation mirrors beta cell function in type 1 diabetes. J. Clin. Invest 128, 3460–3474 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wiedeman AE et al. Autoreactive CD8+ T cell exhaustion distinguishes subjects with slow type 1 diabetes progression. J. Clin. Invest 130, 480–490 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Abdelsamed HA et al. Beta cell-specific CD8+ T cells maintain stem cell memory-associated epigenetic programs during type 1 diabetes. Nat. Immunol 21, 578–587 (2020). In this study, tetramer-sorted antigen specific CD8+ T cells from individualswith T1D were found to have an epigenetic signature suggesting stem cell memory features. Together with Bender et al. (2020), this observation suggests novel features of autoreactive CD8+ T cells.
  • 115.Gearty SV et al. An autoimmune stem-like CD8 T cell population drives type 1 diabetes. Nature 602, 156–161 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Skowera A. et al. β-Cell-specific CD8 T cell phenotype in type 1 diabetes reflects chronic autoantigen exposure. Diabetes 64, 916–925 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Tai N. et al. Microbial antigen mimics activate diabetogenic CD8 T cells in NOD mice. J. Exp. Med 213, 2129–2146 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Hebbandi Nanjundappa R. et al. A gut microbial mimic that hijacks diabetogenic autoreactivity to suppress colitis. Cell 171, 655–667.17 (2017). [DOI] [PubMed] [Google Scholar]
  • 119.Okada M. et al. Islet-specific CD8+ T cells gain effector function in the gut lymphoid tissues via bystander activation not molecular mimicry. Immunol. Cell Biol 101, 36–48 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wan X. et al. Pancreatic islets communicate with lymphoid tissues via exocytosis of insulin peptides. Nature 560, 107–111 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Quesada-Masachs E. et al. Upregulation of HLA class II in pancreatic beta cells from organ donors with type 1 diabetes. Diabetologia 65, 387–401 (2022). [DOI] [PubMed] [Google Scholar]
  • 122.Bottazzo GF et al. In situ characterization of autoimmune phenomena and expression of HLA molecules in the pancreas in diabetic insulitis. N. Engl. J. Med 313, 353–360 (1985). [DOI] [PubMed] [Google Scholar]
  • 123. Foulis AK, Farquharson MA & Hardman R Aberrant expression of class II major histocompatibility complex molecules by B cells and hyperexpression of class I major histocompatibility complex molecules by insulin containing islets in type 1 (insulin-dependent) diabetes mellitus. Diabetologia 30, 333–343 (1987). Together with Bottazo et al. (1985), this study revealed that HLA molecules can be aberrantly expressed in the pancreatic islets of donors with T1D, raising the possibility that this contributes to immune-mediated destruction.
  • 124.Marroqui L. et al. TYK2, a candidate gene for type 1 diabetes, modulates apoptosis and the innate immune response in human pancreatic beta-cells. Diabetes 64, 3808–3817 (2015). [DOI] [PubMed] [Google Scholar]
  • 125.Thompson PJ et al. Targeted elimination of senescent beta cells prevents type 1 diabetes. Cell Metab. 29, 1045–1060.e10 (2019). [DOI] [PubMed] [Google Scholar]
  • 126.Rui J. et al. Tet2 controls the responses of beta cells to inflammation in autoimmune diabetes. Nat. Commun 12, 5074 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Shalev A. Minireview: Thioredoxin-interacting protein: regulation and function in the pancreatic beta-cell. Mol. Endocrinol 28, 1211–1220 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Forlenza GP et al. Effect of verapamil on pancreatic beta cell function in newly diagnosed pediatric type 1 diabetes: a randomized clinical trial. JAMA 329, 990–999 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Xu G. et al. Exploratory study reveals far reaching systemic and cellular effects of verapamil treatment in subjects with type 1 diabetes. Nat. Commun 13, 1159 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Purcell AW, Sechi S & DiLorenzo TP The evolving landscape of autoantigen discovery and characterization in type 1 diabetes. Diabetes 68, 879–886 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Rodriguez-Calvo T, Johnson JD, Overbergh L & Dunne JL Neoepitopes in type 1 diabetes: etiological insights, biomarkers and therapeutic targets. Front. Immunol 12, 667989 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Mannering SI et al. The insulin A-chain epitope recognized by human T cells is posttranslationally modified. J. Exp. Med 202, 1191–1197 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wiles TA et al. Identification of hybrid insulin peptides (HIPs) in mouse and human islets by mass spectrometry. J. Proteome Res 18, 814–825 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Wiles TA, Saba LM & Delong T Peptide-spectrum match validation with internal standards (p-vis): internally-controlled validation of mass spectrometry-based peptide identifications. J. Proteome Res 20, 236–249 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wiles TA et al. Characterization of human CD4 T cells specific for a C-Peptide/C-peptide hybrid insulin peptide. Front. Immunol 12, 668680 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Baker RL et al. Hybrid insulin peptides are autoantigens in type 1 diabetes. Diabetes 68, 1830–1840 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Mitchell AM et al. T-cell responses to hybrid insulin peptides prior to type 1 diabetes development. Proc. Natl Acad. Sci. USA 118, e2019129118 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Delong T. et al. Pathogenic CD4 T cells in type 1 diabetes recognize epitopes formed by peptide fusion. Science 351, 711–714 (2016). This study in diabetic mice and human pancreatic samples suggests a role for hybrid peptides in breaking immune tolerance.
  • 139.Wiles TA et al. An insulin-IAPP hybrid peptide is an endogenous antigen for CD4 T cells in the non-obese diabetic mouse. J. Autoimmun 78, 11–18 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Parras D, Sole P, Delong T, Santamaria P & Serra P Recognition of multiple hybrid insulin peptides by a single highly diabetogenic T-cell receptor. Front. Immunol 12, 737428 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Burton AR et al. On the pathogenicity of autoantigen-specific T-cell receptors. Diabetes 57, 1321–1330 (2008). [DOI] [PubMed] [Google Scholar]
  • 142. Crawford SA et al. Cathepsin D drives the formation of hybrid insulin peptides relevant to the pathogenesis of type 1 diabetes. Diabetes 71, 2793–2803 (2022). This study shows that HIPs, formed by peptide bond cross-linking between proinsulin fragments and other peptides in pancreatic β-cells, are identified in the CD4 T cell targets of indiviuals with T1D; cathepsin D is revealed as the key protease driving HIP formation.
  • 143.Daniel D, Gill RG, Schloot N & Wegmann D Epitope specificity, cytokine production profile and diabetogenic activity of insulin-specific T cell clones isolated from NOD mice. Eur. J. Immunol 25, 1056–1062 (1995). [DOI] [PubMed] [Google Scholar]
  • 144.Strollo R. et al. Autoantibody and T cell responses to oxidative post-translationally modified insulin neoantigenic peptides in type 1 diabetes. Diabetologia 66, 132–146 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Mallone R, Brezar V & Boitard C T cell recognition of autoantigens in human type 1 diabetes: clinical perspectives. Clin. Dev. Immunol 2011, 513210 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yang ML et al. Carbonyl posttranslational modification associated with early-onset type 1 diabetes autoimmunity. Diabetes 71, 1979–1993 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Kracht MJ et al. Autoimmunity against a defective ribosomal insulin gene product in type 1 diabetes. Nat. Med 23, 501–507 (2017). This study explores a new source of immunogenic polypeptides originating from a nonconventional open reading frame within human insulin mRNA, which can be targeted by cytotoxic CD8+ T cells present in individuals with T1D, potentially contributing to β-cell destruction in the disease.
  • 148.Schneider A. et al. The effector T cells of diabetic subjects are resistant to regulation via CD4+ FOXP3+ regulatory T cells. J. Immunol 181, 7350–7355 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hulme MA, Wasserfall CH, Atkinson MA & Brusko TM Central role for interleukin-2 in type 1 diabetes. Diabetes 61, 14–22 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.McClymont SA et al. Plasticity of human regulatory T cells in healthy subjects and patients with type 1 diabetes. J. Immunol 186, 3918–3926 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Dean JW et al. Innate inflammation drives NK cell activation to impair Treg activity. J. Autoimmun 108, 102417 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Vecchione A. et al. Reduced follicular regulatory T cells in spleen and pancreatic lymph nodes of patients with type 1 diabetes. Diabetes 70, 2892–2902 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Battaglia M. et al. Introducing the endotype concept to address the challenge of disease heterogeneity in type 1 diabetes. Diabetes Care 43, 5–12 (2020). This study shows that demographic and immunological factors may be used to identify individuals with T1D in whom the mechanisms of disease and responses to therapies differ. Endotypes may be defined by these parameters.
  • 154.Bluestone JA, Buckner JH & Herold KC Immunotherapy: building a bridge to a cure for type 1 diabetes. Science 373, 510–516 (2021). [DOI] [PubMed] [Google Scholar]
  • 155.Dayan CM et al. Preventing type 1 diabetes in childhood. Science 373, 506–510 (2021). [DOI] [PubMed] [Google Scholar]
  • 156.Pearson JA, McKinney EF & Walker LSK 100 years post-insulin: immunotherapy as the next frontier in type 1 diabetes. Immunother. Adv 1, ltab024 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Mastrandrea L. et al. Etanercept treatment in children with new-onset type 1 diabetes: pilot randomized, placebo-controlled, double-blind study. Diabetes Care 32, 1244–1249 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Quattrin T. et al. Golimumab and beta-cell function in youth with new-onset type 1 diabetes. N. Engl. J. Med 383, 2007–2017 (2020). [DOI] [PubMed] [Google Scholar]
  • 159.Moran A. et al. Interleukin-1 antagonism in type 1 diabetes of recent onset: two multicentre, randomised, double-blind, placebo-controlled trials. Lancet 381, 1905–1915 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Greenbaum CJ et al. IL-6 receptor blockade does not slow beta cell loss in new-onset type 1 diabetes. JCI Insight 6, e150074 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Piemonti L et al. Ladarixin, an inhibitor of the interleukin-8 receptors CXCR1 and CXCR2, in new-onset type 1 diabetes: a multicentre, randomized, double-blind, placebo-controlled trial. Diabetes Obes. Metab 24, 1840–1849 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Citro A. et al. CXCR1/2 inhibition blocks and reverses type 1 diabetes in mice. Diabetes 64, 1329–1340 (2015). [DOI] [PubMed] [Google Scholar]
  • 163.Gitelman SE et al. Imatinib therapy for patients with recent-onset type 1 diabetes: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Diabetes Endocrinol. 9, 502–514 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Waibel M. et al. Baricitinib and beta-cell function in patients with new-onset type 1 diabetes. N. Engl. J. Med 389, 2140–2150 (2023). [DOI] [PubMed] [Google Scholar]
  • 165.Ludvigsson J. et al. GAD treatment and insulin secretion in recent-onset type 1 diabetes. N. Engl. J. Med 359, 1909–1920 (2008). [DOI] [PubMed] [Google Scholar]
  • 166.Ludvigsson J. et al. GAD65 antigen therapy in recently diagnosed type 1 diabetes mellitus. N. Engl. J. Med 366, 433–442 (2012). [DOI] [PubMed] [Google Scholar]
  • 167.Wherrett DK et al. Antigen-based therapy with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset type 1 diabetes: a randomised double-blind trial. Lancet 378, 319–327 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Nowak C. et al. Intralymphatic GAD-alum (Diamyd®) improves glycemic control in type 1 diabetes with HLA DR3-DQ2. J. Clin. Endocrinol. Metab 107, 2644–2651 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Diabetes Prevention Trial — Type 1 Diabetes Study Group. Effects of insulin in relatives of patients with type 1 diabetes mellitus. N. Engl. J. Med 346, 1685–1691 (2002). [DOI] [PubMed] [Google Scholar]
  • 170.Skyler JS et al. Effects of oral insulin in relatives of patients with type 1 diabetes: The Diabetes Prevention Trial — Type 1. Diabetes Care 28, 1068–1076 (2005). [DOI] [PubMed] [Google Scholar]
  • 171.Writing Committee for the Type 1 Diabetes TrialNet Oral Insulin Study Group. et al. Effect of oral insulin on prevention of diabetes in relatives of patients with type 1 diabetes: a randomized clinical trial. JAMA 318, 1891–1902 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Bonifacio E. et al. Effects of high-dose oral insulin on immune responses in children at high risk for type 1 diabetes: the Pre-POINT randomized clinical trial. JAMA 313, 1541–1549 (2015). [DOI] [PubMed] [Google Scholar]
  • 173. Pescovitz MD et al. Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N. Engl. J. Med 361, 2143–2152 (2009). This study highlights a role for B cells in the pathogenesis in T1D and demonstrates that treatment with rituximab can preserve β-cell function in individuals with T1D.
  • 174.Pescovitz MD et al. B-lymphocyte depletion with rituximab and beta-cell function: two-year results. Diabetes Care 37, 453–459 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Chamberlain N. et al. Rituximab does not reset defective early B cell tolerance checkpoints. J. Clin. Invest 126, 282–287 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Linsley PS et al. Elevated T cell levels in peripheral blood predict poor clinical response following rituximab treatment in new-onset type 1 diabetes. Genes Immun. 20, 293–307 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Lenschow DJ et al. Differential effects of anti-B7-1 and anti-B7-2 monoclonal antibody treatment on the development of diabetes in the nonobese diabetic mouse. J. Exp. Med 181, 1145–1155 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Orban T. et al. Costimulation modulation with abatacept in patients with recent-onset type 1 diabetes: follow-up 1 year after cessation of treatment. Diabetes Care 37, 1069–1075 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Orban T. et al. Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial. Lancet 378, 412–419 (2011). This randomized placebo-controlled clinical trial shows that treatment with CTLA4Ig over 2 years attenuated loss of β-cell function.
  • 180.Russell WE et al. Abatacept for delay of type 1 diabetes progression in stage 1 relatives at risk: a randomized, double-masked, controlled trial. Diabetes Care 46, 1005–1013 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Orban T. et al. Reduction in CD4 central memory T-cell subset in costimulation modulator abatacept-treated patients with recent-onset type 1 diabetes is associated with slower C-peptide decline. Diabetes 63, 3449–3457 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Wang CJ et al. Costimulation blockade in combination with IL-2 permits regulatory T cell sparing immunomodulation that inhibits autoimmunity. Nat. Commun 13, 6757 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Haller MJ et al. Anti-thymocyte globulin/G-CSF treatment preserves beta cell function in patients with established type 1 diabetes. J. Clin. Invest 125, 448–455 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Haller MJ et al. Antithymocyte globulin plus G-CSF combination therapy leads to sustained immunomodulatory and metabolic effects in a subset of responders with established type 1 diabetes. Diabetes 65, 3765–3775 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Haller MJ et al. Low-dose anti-thymocyte globulin preserves C-peptide, reduces HbA1c, and increases regulatory to conventional T-cell ratios in new-onset type 1 diabetes: two-year clinical trial data. Diabetes 68, 1267–1276 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Haller MJ et al. Low-dose anti-thymocyte globulin (ATG) preserves beta-cell function and improves HbA1c in new-onset type 1 diabetes. Diabetes Care 41, 1917–1925 (2018). This randomized placebo-controlled clinical trial shows that ATG treatment reduced the decline in β-cell function in patients with new-onset T1D.
  • 187.Jacobsen LM et al. Responders to low-dose ATG induce CD4 T cell exhaustion in type 1 diabetes. JCI Insight 8, e161812 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Rigby MR et al. Alefacept provides sustained clinical and immunological effects in new-onset type 1 diabetes patients. J. Clin. Invest 125, 3285–3296 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Charpentier B. et al. Evidence that antihuman tumor necrosis factor monoclonal antibody prevents OKT3-induced acute syndrome. Transplantation 54, 997–1002 (1992). [DOI] [PubMed] [Google Scholar]
  • 190.Chatenoud L. OKT3-induced cytokine-release syndrome: prevention effect of anti-tumor necrosis factor monoclonal antibody. Transplant. Proc 25, 47–51 (1993). [PubMed] [Google Scholar]
  • 191.Norman DJ, Chatenoud L, Cohen D, Goldman M & Shield CF III Consensus statement regarding OKT3-induced cytokine-release syndrome and human antimouse antibodies. Transplant. Proc 25, 89–92 (1993). [PubMed] [Google Scholar]
  • 192.Herold KC et al. Prevention of autoimmune diabetes with nonactivating anti-CD3 monoclonal antibody. Diabetes 41, 385–391 (1992). [DOI] [PubMed] [Google Scholar]
  • 193.Chatenoud L, Primo J & Bach JF CD3 antibody-induced dominant self tolerance in overtly diabetic NOD mice. J. Immunol 158, 2947–2954 (1997). [PubMed] [Google Scholar]
  • 194. Chatenoud L, Thervet E, Primo J & Bach JF Anti-CD3 antibody induces Long-term remission of overt autoimmunity in nonobese diabetic mice. Proc. Natl Acad. Sci. USA 91, 123–127 (1994). This study shows, in the NOD model, how brief treatment with monolonal CD3 antibody could reverse spontaneous diabetes, after its appearance, and induce specific immune tolerance.
  • 195.Sherry NA et al. Effects of autoimmunity and immune therapy on beta-cell turnover in type 1 diabetes. Diabetes 55, 3238–3245 (2006). [DOI] [PubMed] [Google Scholar]
  • 196.Bach JF Anti-CD3 antibodies for type 1 diabetes: beyond expectations. Lancet 378, 459–460 (2011). [DOI] [PubMed] [Google Scholar]
  • 197. Herold KC et al. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N. Engl. J. Med 346, 1692–1698 (2002). This clinical trial of teplizumab in patients with new-onset T1D shows that a single 14-day course would attenuate the loss of β-cell function.
  • 198.Herold KC et al. A single course of anti-CD3 monoclonal antibody hOKT3gamma1(Ala-Ala) results in improvement in C-peptide responses and clinical parameters for at least 2 years after onset of type 1 diabetes. Diabetes 54, 1763–1769 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Keymeulen B. et al. Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N. Engl. J. Med 352, 2598–2608 (2005). [DOI] [PubMed] [Google Scholar]
  • 200.Herold KC et al. Teplizumab (anti-CD3 mAb) treatment preserves C-peptide responses in patients with new-onset type 1 diabetes in a randomized controlled trial: metabolic and immunologic features at baseline identify a subgroup of responders. Diabetes 62, 3766–3774 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Herold KC et al. Teplizumab treatment may improve C-peptide responses in participants with type 1 diabetes after the new-onset period: a randomised controlled trial. Diabetologia 56, 391–400 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Herold KC et al. Teplizumab: a disease-modifying therapy for type 1 diabetes that preserves beta-cell function. Diabetes Care 46, 1848–1856 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Ramos EL et al. Teplizumab and beta-cell function in newly diagnosed type 1 diabetes. N. Engl. J. Med 389, 2151–2161 (2023). [DOI] [PubMed] [Google Scholar]
  • 204. Herold KC et al. An anti-CD3 antibody, teplizumab, in relatives at risk for type 1 diabetes. N. Engl. J. Med 381, 603–613 (2019). This randomized placebo-controlled clinical trial shows that a single 14-day course of the CD3 antibody teplizumab would delay the onset of T1D in relatives at high risk.
  • 205.Sosenko JM et al. Phenotypes associated with zones defined by area under the curve glucose and C-peptide in a population with islet autoantibodies. Diabetes Care 46, 1098–1105 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.McKinney EF, Lee JC, Jayne DR, Lyons PA & Smith KG T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Long SA et al. Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs β-cell function. Diabetes 61, 2340–2348 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Seelig E et al. The DILfrequency study is an adaptive trial to identify optimal IL-2 dosing in patients with type 1 diabetes. JCI Insight 3, e99306 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Zhang JY et al. Low-dose IL-2 reduces IL-21+ T cell frequency and induces anti-inflammatory gene expression in type 1 diabetes. Nat. Commun 13, 7324 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Rosenzwajg M. et al. Low-dose IL-2 in children with recently diagnosed type 1 diabetes: a phase I/II randomised, double-blind, placebo-controlled, dose-finding study. Diabetologia 63, 1808–1821 (2020). [DOI] [PubMed] [Google Scholar]
  • 211.Putnam AL et al. Expansion of human regulatory T-cells from patients with type 1 diabetes. Diabetes 58, 652–662 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Bluestone JA et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci. Transl. Med 7, 315ra189 (2015). This phase Ib clinical study evaluates the safety, pharmacodynamics and limited information on the efficacy of autologous Treg cells that were expanded ex vivo and administered to indivduals with recent-onset T1D.
  • 213.Dong S. et al. The effect of low-dose IL-2 and Treg adoptive cell therapy in patients with type 1 diabetes. JCI Insight 6, e147474 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Ferreira LMR, Muller YD, BLuestone JA & Tang Q Next-generation regulatory T cell therapy. Nat. Rev. Drug Discov 18, 749–769 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Cui C, Craft J & Joshi NS T follicular helper cells in cancer, tertiary Lymphoid structures, and beyond. Semin. Immunol 69, 101797 (2023). [DOI] [PubMed] [Google Scholar]
  • 216.Linsley PS, Greenbaum CJ, Speake C, Long SA & Dufort MJ B lymphocyte alterations accompany abatacept resistance in new-onset type 1 diabetes. JCI Insight 4, e126136 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Thomas HE et al. Interferon signalling in pancreatic beta cells. Front. Biosci 14, 644–656 (2009). [DOI] [PubMed] [Google Scholar]
  • 218.Mayr A. et al. GAD autoantibody affinity and epitope specificity identify distinct immunization profiles in children at risk for type 1 diabetes. Diabetes 56, 1527–1533 (2007). [DOI] [PubMed] [Google Scholar]
  • 219.Endesfelder D. et al. Time-resolved autoantibody profiling facilitates stratification of preclinical type 1 diabetes in children. Diabetes 68, 119–130 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Achenbach P. et al. Stratification of type 1 diabetes risk on the basis of isLet autoantibody characteristics. Diabetes 53, 384–392 (2004). [DOI] [PubMed] [Google Scholar]
  • 221.Achenbach P. et al. Mature high-affinity immune responses to (pro)insulin anticipate the autoimmune cascade that leads to type 1 diabetes. J. Clin. Invest 114, 589–597 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Schlosser M. et al. In insulin-autoantibody-positive children from the general population, antibody affinity identifies those at high and low risk. Diabetologia 48, 1830–1832 (2005). [DOI] [PubMed] [Google Scholar]
  • 223.Yang ML et al. Citrullination of glucokinase is linked to autoimmune diabetes. Nat. Commun 13, 1870 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Schloot NC et al. Comparison of cytokine ELISpot assay formats for the detection of islet antigen autoreactive T cells. Report of the Third Immunology of Diabetes Society T-cell Workshop. J. Autoimmun 21, 365–376 (2003). [DOI] [PubMed] [Google Scholar]
  • 225.Herold KC et al. Validity and reproducibility of measurement of islet autoreactivity by T-cell assays in subjects with early type 1 diabetes. Diabetes 58, 2588–2595 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.James EA et al. Combinatorial detection of autoreactive CD8+ T cells with HLA-A2 multimers: a multi-centre study by the Immunology of Diabetes Society T Cell Workshop. Diabetologia 61, 658–670 (2018). [DOI] [PubMed] [Google Scholar]
  • 227.Ogura H. et al. Identification and analysis of islet antigen-specific CD8+ T cells with T cell libraries. J. Immunol 201, 1662–1670 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Herold KC et al. Increased T cell proliferative responses to islet antigens identify clinical responders to anti-CD20 monoclonal antibody (rituximab) therapy in type 1 diabetes. J. Immunol 187, 1998–2005 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Brown CT et al. Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS ONE 6, e25792 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Vatanen T. et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 165, 842–853 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Martinov T et al. Programmed death-1 restrains the germinal center in type 1 diabetes. J. Immunol 203, 844–852 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Serr I. et al. miRNA92a targets KLF2 and the phosphatase PTEN signaling to promote human T follicular helper precursors in T1D islet autoimmunity. Proc. Natl Acad. Sci. USA 113, E6659–E6668 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Smith JA, Tso JY, Clark MR, Cole MS & Bluestone JA Nonmitogenic anti-CD3 monoclonal antibodies deliver a partial T cell receptor signal and induce clonal anergy. J. Exp. Med 185, 1413–1422 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Esplugues E et al. Control of TH17 cells occurs in the small intestine. Nature 475, 514–518 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Waldron-Lynch F. et al. Teplizumab induces human gut-tropic regulatory cells in humanized mice and patients. Sci. Transl. Med 4, 118ra112 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Penaranda C, Tang Q & Bluestone JA Anti-CD3 therapy promotes tolerance by selectively depleting pathogenic cells while preserving regulatory T cells. J. Immunol 187, 2015–2022 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Belghith M. et al. TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat. Med 9, 1202–1208 (2003). [DOI] [PubMed] [Google Scholar]
  • 238.Long SA et al. Partial exhaustion of CD8 T cells and clinical response to teplizumab in new-onset type 1 diabetes. Sci. Immunol 1, eaai7793 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Perdigoto AL et al. Treatment of type 1 diabetes with teplizumab: clinical and immunological follow-up after 7 years from diagnosis. Diabetologia 62, 655–664 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Campbell-Thompson M. et al. Network for Pancreatic Ogan Donors with Diabetes (nPOD): developing a tissue biobank for type 1 diabetes. Diabetes Metab. Res. Rev 28, 608–617 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES