ArticlePDF AvailableLiterature Review

Cell-based therapies and other non-traditional approaches for Type 1 diabetes

Authors:

Abstract and Figures

The evolution of Type 1 diabetes (T1D) therapy has been marked by consecutive shifts, from insulin replacement to immunosuppressive drugs and targeted biologics (following the understanding that T1D is an autoimmune disease), and to more disease-specific or patient-oriented approaches such as antigen-specific and cell-based therapies, with a goal to provide efficacy, safety and long-term protection. At the same time, another important paradigm shift from treatment of new onset T1D patients to prevention in high-risk individuals has taken place, based on the hypothesis that therapeutic approaches deemed sufficiently safe may show better efficacy if applied early enough to maintain endogenous β cell function, a concept supported by many preclinical studies. This new strategy has been made possible by capitalizing on a variety of biomarkers that can more reliably estimate the risk and rate of progression of the disease. More advanced ("omic"-based) biomarkers that also shed light on the underlying contributors of disease for each individual will be helpful to guide the choice of the most appropriate therapies, or combinations thereof. In this review, we present current efforts to stratify patients according to biomarkers and current alternatives to conventional drug-based therapies for T1D, with a special emphasis on cell-based therapies, their status in the clinic and potential for treatment and/or prevention. This article is protected by copyright. All rights reserved.
Content may be subject to copyright.
Concise Review: Cell-Based Therapies and Other
Non-Traditional Approaches for Type 1 Diabetes
REMI J. CREUSOT,
a
MANUELA BATTAGLIA,
b
MARIA-GRAZIA RONCAROLO,
c
C. GARRISON FATHMAN
d
Key Words. T cells Cell therapy Autoimmunity Prevention Type 1 diabetes
Immunoregulation Antigen-specific
ABSTRACT
The evolution of Type 1 diabetes (T1D) therapy has been marked by consecutive shifts, from
insulin replacement to immunosuppressive drugs and targeted biologics (following the under-
standing that T1D is an autoimmune disease), and to more disease-specific or patient-oriented
approaches such as antigen-specific and cell-based therapies, with a goal to provide efficacy,
safety, and long-term protection. At the same time, another important paradigm shift from
treatment of new onset T1D patients to prevention in high-risk individuals has taken place,
based on the hypothesis that therapeutic approaches deemed sufficiently safe may show better
efficacy if applied early enough to maintain endogenous bcell function, a concept supported by
many preclinical studies. This new strategy has been made possible by capitalizing on a variety
of biomarkers that can more reliably estimate the risk and rate of progression of the disease.
More advanced (“omic”-based) biomarkers that also shed light on the underlying contributors
of disease for each individual will be helpful to guide the choice of the most appropriate thera-
pies, or combinations thereof. In this review, we present current efforts to stratify patients
according to biomarkers and current alternatives to conventional drug-based therapies for T1D,
with a special emphasis on cell-based therapies, their status in the clinic and potential for treat-
ment and/or prevention. STEM CELLS 2016; 00:000—000
SIGNIFICANCE STATEMENT
This article summarizes the significance of the paradigm shift in the thinking about the treat-
ment of Type 1 diabetes. Current treatment strategies are now directed toward prevention of
disease progression to maintain endogenous beta cell function. The use of immunomodulating
strategies including antigen- and cell-based therapies as well as the need to identify new bio-
markers that allow a measure of disease stage and time to onset of hyperglycemia for selection
of appropriate patients to enroll in prevention trials are discussed.
INTRODUCTION
Type 1 diabetes (T1D), like its more common
Type 2 counterpart, has been rising in preva-
lence and incidence primarily in Western coun-
tries [1, 2] (Fig. 1). Insulin replacement
therapy has been the primary treatment of all
forms of diabetes for almost 100 years, but
inadequate control of its delivery has allowed
a number of complications to markedly dimin-
ish the quality of life of affected individuals,
and contributed to an increasingly intolerable
financial burden. The realization that a subset
of patients presents with an autoimmune form
of insulin-dependent diabetes was made in the
1970s [3]. The initial model suggesting the
potential pathogenesis of this disorder as a
chronic autoimmune disease directed against b
cells was proposed by George Eisenbarth in
the mid-1980s [4]. This understanding led to
subsequent attempts to develop more specific
treatments for this autoimmune form of diabe-
tes, initially with immunosuppressive therapies
that had proven effective in other chronic
autoimmune diseases, including cyclosporine A
(CsA) or anti-thymocyte globulin and predni-
sone [5–7]. Despite initial suggestion of effi-
cacy of CsA, no subsequent study has been
able to confirm these initial results. In addi-
tion, the lack of lasting effects once CsA was
withdrawn and the serious renal toxicity of
the drug severely limited enthusiasm for this
approach.
Subsequent natural history studies have
made the approach to treatment more com-
plex as these studies have demonstrated that
underlying autoimmune responses are present
for varying periods of time, usually years, in
genetically predisposed individuals before the
a
Department of Medicine,
Columbia Center for
Translational Immunology
and Naomi Berrie Diabetes
Center, Columbia University
Medical Center, New York,
USA;
b
Diabetes Research
Institute, IRCCS San Raffaele
Scientific Institute, Milan,
Italy;
c
Division of Stem Cell
Transplantation and
Regenerative Medicine,
Department of Pediatrics,
Stanford University School of
Medicine Stanford, CA, USA;
d
Division of Immunology and
Rheumatology, Department
of Medicine, Stanford
University School of
Medicine Stanford, CA, USA
Correspondence: C. Garrison
Fathman MD, Division of
Rheumatology and Immunology,
Department of Medicine,
Stanford University, 269 Campus
Drive West, Stanford, California
94305, USA. Telephone: 650-
723-7887; Fax: 650-725-1958; e-
mail: cfathman@stanford.edu
Received July 7, 2015; accepted
for publication December 7,
2015; first published online in
STEM CELLS EXPRESS February 3,
2016.
V
CAlphaMed Press
1066-5099/2016/$30.00/0
http://dx.doi.org/
10.1002/stem.2290
This is an open access article
under the terms of the Creative
Commons Attribution-NonCom-
mercial-NoDerivs License, which
permits use and distribution in
any medium, provided the origi-
nal work is properly cited, the
use is non-commercial and no
modifications or adaptations are
made.
STEM CELLS 2016;00:00–00 www.StemCells.com V
C2016 The Authors STEM CELLS published by
Wiley Periodicals, Inc. on behalf of AlphaMed Press
CONCISE REVIEWS
appearance of overt hyperglycemia. Particular major histo-
compatibility class II haplotypes (HLA-DR3/4, HLA-DQ8) confer
the greatest risk as genetic factors [8]. In addition to serving
as a diagnosis tool for T1D in new onset diabetic patients, cir-
culating autoantibodies against bcell proteins (specificity and
quantity) in at-risk individuals, as well as abnormalities in the
oral glucose tolerance test, can generally help predict the risk
of, and time remaining, before the onset of hyperglycemia
[9]. These predictive data have raised the possibility of
attempting therapeutic intervention before the onset of
hyperglycemia in high-risk individuals identified based on bio-
markers like those mentioned above. Prevention of T1D may
represent a viable alternative to an actual cure by perma-
nently blocking the autoimmune response while there are suf-
ficient bcells remaining, and may offer a more cost-effective
approach in the short-term to deal with the alarming rise in
the incidence of disease (Fig. 1). To make the matter even
more complex, many patients may present a spontaneous but
temporary remission after onset, known as the honeymoon
period, possibly reflecting reduced stress on residual bcells
after initial insulin treatment. This honeymoon period may
perhaps represent a sweet window of opportunity (pun
intended) to exploit for the use of intervention therapy.
In this review, we will discuss how our therapeutic arsenal
to fend off this autoimmune disease has greatly diversified
beyond traditional drugs and biologicals to include various
forms of cell therapies, as well as other less conventional
approaches. The field is witnessing a paradigm shift from
immunosuppressive therapies applied after the onset of
hyperglycemia (a time at which bcell function has generally
been irreversibly lost) to prevention strategies attempting to
shut off the autoimmune response and preserve bcell func-
tion in high-risk individuals. This of course entails the use of
reliable biomarkers to identify the most appropriate at-risk
subjects for such intervention trials, and perhaps also guide
the type of therapy that should be employed, paving the way
to more personalized therapies. Current studies using new
techniques of transcriptomics and proteomics [10–13] are
attempting to more precisely stratify those at risk by identify-
ing novel biomarkers that may be superior to those currently
used to define the stage or rate of progression of disease,
and thus help select appropriate subjects to enter into pre-
vention trials. Although the strategies described in this review
have all shown remarkable efficacy in preclinical models, it
should be noted that little or no clinical efficacy data is avail-
able for most of them, whether they are evaluated in the
treatment of recent onset patients after safety has been dem-
onstrated or in prevention studies following safe but ineffec-
tive use in recent onset patients.
ASHIFT FROM TREATMENT TO PREVENTION ISDRIVEN BY
BIOMARKERS GUIDING WHEN AND HOW TO INTERVENE
The progress in identifying the patients who are at “high-risk”
and should be entered into prevention trials has been sup-
ported by an improved understanding of T1D disease proc-
esses that allows screening for at-risk individuals and
stratification of the individual’s risk and time of progression
to the development of hyperglycemia. The first level of
screening is comprised of family history (number of relatives
with T1D and degree of relationship) and HLA haplotype
(HLA-DR3/4 heterozygosy combined with HLA-DQ8 conferring
the highest known risk) [9]. Although these risk factors are
fixed from birth, new relatives may become diagnosed later
and the relative risk re-evaluated. These parameters have
served to enroll young subjects into studies on how environ-
mental factors influence disease progression (e.g., primary
prevention studies examining diet alterations in genetically at-
risk babies with no evidence of autoimmunity, Table 1). These
individuals can be closely and regularly monitored and
undergo a second level of screening consisting of well-
established biomarkers such as circulating autoantibodies to b
cell antigens insulin, GAD65, IA-2, ZnT8, and IGRP [9, 14],
which have served as good predictive tools [15–18] and
enrollment criteria for prevention studies. In vitro immunoas-
says performed on peripheral blood cells including T cell
responses to bcell antigens or identification of diabetogenic
T cells by tetramer staining complete this assessment of the
breadth (how many autoantigens targeted) and amplitude
(antibody titers or frequency of tetramer-positive T cells) of
the autoimmune response [14]. More recently, biomarkers
based on epigenetic changes have been discovered, such as
circulating demethylated insulin DNA [19, 20] and differences
in methylation level at specific CpG sites in immune cells [21].
Increased levels of demethylated insulin DNA in the blood
correlates with the extent of bcell damage [19], while the
extent of insulitis may now be evaluated by refined imaging
techniques [22].
These biomarkers of prediction are important to evaluate
disease risk and rate of progression (indicating with a high
degree of confidence if and approximately when a patient will
progress to overt hyperglycemia unless the course of progres-
sion is altered by treatment), and, therefore, to determine
when to treat. Individuals with a comparable risk level may
experience a different rate of progression, according to their
genetic makeup (other genes besides HLA) and their environ-
ment, which differentially affect mechanisms of immune toler-
ance and pathogenesis. As environmental factors change, the
rate of progression may increase or decrease, placing the sub-
ject at greater risk or slowing down the development of the
Figure 1. T1D incidence has doubled every 20 years. Data for
Finland are from the Finnish National Public Health Institute; data
for Sweden are from the Swedish Childhood Diabetes Registry;
data for Colorado are from the Colorado IDDM Registry, the Bar-
bara Davis Center for Childhood Diabetes, and SEARCH for Diabe-
tes in Youth; data for Germany are a compilation of two reports;
and data from Poland are from Diabetologia 2010;54:508-515.
Reprinted with permission from the Ann N Y Acad Sci
2008;1150:1-13, with additional modifications and permission
from Marian Rewers and Jay Skyler.
2Unconventional approaches to treat Type 1 diabetes
V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press STEM CELLS
disease (Fig. 2). As a result of multiple etiological factors act-
ing in concert, the disease can progress from very fast in the
case of fulminant T1D [23] to very slow in the case of latent
autoimmune diabetes in adults (LADA) [24]. However, the
most aggressive forms of disease, like fulminant T1D, might
not benefit from prevention unless the trigger becomes well
understood.
Because of the heterogeneity of etiological factors that
may control the rate of progression, it is unlikely that patients
stratified as having a similar risk will be equally responsive to
a particular treatment (Table 2). Treating a high-risk patient
with the wrong drug would cost precious time during which b
cells will continue to be destroyed. In prevention studies, as
opposed to new onset cases, more time will be needed
before it can be determined whether the treatment is effec-
tive. Conversely, treating a low-risk patient, who may never
advance to onset, even with the right prevention therapy,
would involve unnecessary costs and risks. Thus, a third level
of screening that is more sophisticated (using novel bio-
markers featured in larger datasets) will be required to help
determine how best to treat each patient by providing clues
as to the underlying defects that need to be acted upon. A
combination of genetic, transcriptomic, and proteomic tests
performed on blood samples will likely be part of such
screening in the future, and extensive research is being con-
ducted to this end. Furthermore, advances in viromics have
enabled the development of sensitive blood tests that can
detect prior exposure to particular viruses, some of which
have long been suspected to play a role as a trigger for the
disease in some individuals [44]. If these tests help confirm a
link between these pathogens (which are not uncommon, and
therefore would not be sufficient to induce T1D), then the
prospect of vaccinating genetically at-risk individuals becomes
possible.
In recent years, it has become clear that combination
therapies, selected to address multiple underlying defects, will
become more prominent in our effort to tackle T1D heteroge-
neity in both prevention and treatment. Such combination
therapies are expected to be effective in larger cohorts of
patients with overlapping defects. Although many of the
immunosuppressive strategies have not been effective when
administered post-hyperglycemia, they might be appropriate
to use at an earlier stage of disease where they may prove
more efficacious. Besides efficacy and safety, the cost will
Table 1. Main clinical trials focused on the prevention of T1D
Prevention trials Drug Type of study
Diet/supplement-based prevention
NCT01055080 (FINDIA) Baby diet alteration Phase 1, primary prevention
NCT00570102 (MIP) Baby diet alteration Phase 2, primary prevention
NCT01115621 (BABYDIET) Baby diet, delayed gluten Phase 1, primary prevention
NCT00179777 (TRIGR) Controlled diet in infants Phase 2, primary prevention
NCT00333554 (NIDDK) Omega-3-fatty acids Phase 2, primary prevention
NCT00141986 (CDA) Vitamin D Phase 1, primary prevention
bcell antigen-based prevention
NCT00004984 (DPT-1) Parenteral or oral insulin Phase 2, secondary prevention
NCT00419562 (NIDDK) Oral insulin Phase 3, secondary prevention
ISRCTN76104595 (Pre-POINT) Oral insulin Phase 1, primary prevention
NCT00654121 (BDR Trial) Subcut. insulin (Actrapid HM) Phase 2, secondary prevention
NCT00223613 (DIPP) Intranasal insulin Phase 3, secondary prevention
NCT00336674 (INIT-II) Intranasal insulin Phase 2, secondary prevention
NCT01122446 (DIAPREV-IT) Diamyd (GAD-Alum) Phase 2, secondary prevention
Combinations of the above
NCT02387164 (DIAPREV-IT2) Diamyd (GAD-Alum 1Vit. D) Phase 2, secondary prevention
Prevention using biological-based immunotherapy
NCT01773707 (NIDDK, TN18) Abatacept (CTLA4-Ig) Phase 2, secondary prevention
NCT01030861 (NIDDK, TN10) Teplizumab (anti-CD3) Phase 2, secondary prevention
Cell-based prevention
CoRD study (Sydney) Umbilical cord blood Phase 1, secondary prevention
Note: Clinical trials are color-shaded based on whether they are ,,or
Figure 2. Rates of T1D disease progression. In T1D, earlier onset
reflects a faster rate of progression through risk levels (repre-
sented by narrower height). The risk level can be evaluated
according to family history and HLA haplotype, which are fixed at
birth, as well as circulating anti-bcell autoantibodies and certain
metabolic measurements, which are dynamic [9]. The rate of pro-
gression (a) takes into account all causes and factors that may
contribute to pathogenesis, including (but not limited to) defec-
tive deletional tolerance, defective immune regulation, defective/
delayed clearance of damaged bcells, viral infection (high tropism
for bcells and/or molecular mimicry), and bcell intrinsic factors
such as susceptibility to infection, apoptosis or dedifferentiation
(Table 2). Progression through these stages may not be linear for
all individuals, as precipitating events during life (depicted by a
thunderbolt) may accelerate the rate, while other environmental
changes may curb it. Furthermore, accumulation of genetic (pre-
fixed) factors may cause an individual to start at an intermediate
risk, but genetic analysis other than HLA haplotype is not yet rou-
tinely performed. Successful prevention of disease will require
more advanced tools to evaluate the risk level, the rate of pro-
gression, and preferably, the nature of the deficiencies that con-
tribute to the rate of progression (Table 2).
Creusot, Battaglia, Roncarolo et al. 3
www.StemCells.com V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press
need to be leveraged against the long-term benefits. Expen-
sive therapies that induce durable tolerance and protection
may, however, save money in the long run. It is unusual for
new drugs to be approved for prevention of disease without
prior testing in new-onset patients: the immunomodulatory
drugs and cell-based therapies described below are no excep-
tion to this rule.
OVERVIEW OF THE CURRENT LANDSCAPE OF NON-CELL
THERAPIES
Cell- or Pathway-Neutralizing Biologics
Regulatory authorities have historically prioritized treatment of
new onset patients because these cases are more pressing, the
risk tolerance greater, and the studies shorter, smaller, and less
expensive. In an attempt to replace the use of globally immuno-
suppressive drugs, a number of promising biologics have been
evaluated in new onset patients, such as anti-CD3 mAb [49–51],
anti-CD20 mAb [52], CTLA4-Ig [53]. These drugs showed signifi-
cant but limited (transient) efficacy in new-onset patients. These
drugs are now being tested in high-risk normoglycemic patients
where they might have a more pronounced and durable effect in
sustaining euglycemia (Table 1). These drugs were deemed safe
enough by the US Food and Drug Administration to be used pro-
phylactically at a dose unlikely to cause serious adverse events.
Current studies using such biologics to prevent disease in high-
risk patients include two major TrialNet studies: TN10 (Clinical-
Trial.gov NCT01030861) using Teplizumab (anti-CD3 mAb) and
TN18 (ClinicalTrial.gov NCT01773707) using Abatacept (CTLA4-Ig).
Many other biologics used to treat other autoimmune diseases
are also being evaluated for T1D. However, these drugs remain
relatively nonspecific and may still carry accrued risks of infections
or malignancies in susceptible subjects.
Low Dose IL-2: A Safer and More Selective Approach?
Because of different sensitivities, it was found that regulatory
T cells (Tregs), a subset of T cells that protects from autoim-
munity, are selectively stimulated by low doses of IL-2, a T
cell growth factor [54]. This new approach is particularly note-
worthy because of its safety profile, based on several pub-
lished studies on the use of low dose IL-2 to treat
inflammatory diseases including chronic graft-versus-host dis-
ease (GvHD) [55–57] and hepatitis C virus-induced vasculitis
[58, 59], and preliminary studies on its use as a potential
treatment of T1D [60]. In addition, preclinical studies in non-
obese diabetic (NOD) mice showed that low dose IL-2 admin-
istered after onset of hyperglycemia restored euglycemia in a
majority of the treated mice [61]. In each instance, low dose
IL-2 therapy was associated with a dose-dependent increase
in the number of circulating Tregs and a marked diminution
of inflammatory cytokine expression in the serum of the
Table 2. Factors contributing to the rate of disease progression
Possible mechanisms
(not mutually exclusive)
Possible causes or
predisposing genes Possible biomarkers
Possible therapeutic
approach to prevent
(and reverse) disease References
Defective deletional tolerance:
higher frequency of
bcell-reactive T cells
(and B cells)
HLA-DR, INS, PTPN22
polymorphism
HLA-DR, selected SNP analysis,
MHC tetramer analysis/ELISPOT
Antigen-specific therapies [8, 25–29]
Defective immune regulation:
reduced number,
responsiveness and/or
function of regulatory
T cells
IL2RA, IL2, CTLA4, IL10,
PTPN22 polymorphism
Vitamin D and/or fiber
deficiency Foxp3
promoter methylation
Treg suppression, TSDR assay,
STAT5 responsiveness
Expanding Tregs and/or
boosting their function
(in vivo or ex vivo);
antigen-specific therapies
[8, 25, 30–36]
Antigen-presenting cells:
hyperactivity under
inflammatory conditions
or defective tolerogenic
properties
Genetic predispositions?
Environmental factors?
Functional characterization
of certain blood cells?
Blockade of specific
costimulatory pathways
or cytokines
[37, 38]
Response of bcells: apoptosis,
stress ( 6generation of
neo-antigens),
de-differentiation or
trans-differentiation
IFIH1 polymorphism,
inflammation, some
unknown genetic
determinants, inability
to cope with excessive
stress
Selected SNPs, circulating
demethylated insulin DNA
(also reflects bcell immune
destruction); impaired
glucose tolerance
Drugs increasing bcell
replication, reducing
bcell stress (imatinib?),
or stabilizing bcell
phenotype
Anti-inflammatory drugs?
[20, 39–41]
Defective/delayed clearance
of damaged bcells / bcell
antigens (disease initiation)
Genetic predispositions? None (too early to detect)? None at the moment [42, 43]
Interaction with microbes:
molecular mimicry
(cross-reactivity with
bcell antigens), immune
deviation or dysregulation
HLA-DR, IFIH1, TLR7/8
polymorphism; Infection
(e.g., enterovirus);
Dysbiosis (imbalanced
microbiome)
Infection history (difficult)
Microbiome profiling
Probiotics? Vaccines?
Anti-inflammatory drugs?
[8, 35, 44–48]
Combinations of genetic and environmental factors contribute to the initiation and progression of T1D disease, leading to various deficiencies at
the level of both immune cells and bcells. Gene-wide association studies have identified some 40 gene polymorphisms each contributing a
small risk to the disease, some of which are listed here. The more deficiencies that exist in a particular individual, the faster the progression is
expected to be. However, different patients are characterized by different combinations of such deficiencies, leading to substantial heterogeneity
in how they progress and how rapidly. Biomarkers that assess not only the risk but also the underlying deficiencies will help inform the choice
of prevention therapies to be applied to more homogeneous cohorts of patients for better efficacy. References are only provided as examples
to illustrate the concepts.
4Unconventional approaches to treat Type 1 diabetes
V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press STEM CELLS
treated mice or patients in the initial short-term safety trial
[60]. An additional dose finding study to determine the optimal
dose of IL-2 required to increase the number and response of
Tregs has been completed in T1D patients (ClinicalTrial.gov
NCT01827735). Subsequently, an efficacy trial has begun in
patients with recent onset T1D (ClinicalTrial.gov NCT01862120).
Once these studies (in Table 3) are completed and the safety
profile confirmed, a move to recruit high-risk patients in low
dose IL-2 studies will be expected.
However, the potential success of low dose IL-2 therapy in
T1D patients rests on two assumptions: (i) Tregs are function-
ally defective and (ii) IL-2 production is impaired. Studies on
whether CD4
1
CD25
1
Tregs are defective in T1D have yielded
conflicting results (decreased frequency [62], decreased func-
tion [63], or normal frequency and function [64]), which may
reflect inadequate identification of Tregs by available markers;
recruitment of patients different in age and disease progres-
sion and differences in experimental conditions. Follow-up
studies using more specific markers (FOXP3
1
CD127
low
and
demethylation of regulatory elements of the FOXP3 gene)
showed that both the frequency and function of Tregs are
normal in the blood of T1D patients, even though a transient
decrease of suppressor activity may occur early after diagnosis
[65], and in a subset of T1D patients [30]. Studies from the
Battaglia lab showed that reduced suppressive function of
Tregs may be restricted to the pancreatic lymph nodes in
patients with long lasting T1D [31]. A defect in IL-2 produc-
tion by total peripheral blood mononuclear cells of patients
with new onset T1D was reported several years ago [66] but
never confirmed as a key immunological feature of T1D
patients. A recent study showed that the T1D-susceptibility
IL2RA haplotype identified by rs12722495 is associated with
decreased signaling via the IL-2 pathway in both memory T
cells and Tregs and that this is linked to diminished Treg func-
tion [32]. However, this phenotype is limited to carriers of
this single nucleotide polymorphism (SNP) and not to all indi-
viduals. Thus, it is likely that this treatment may benefit some
patients more than others, again based on their underlying
defects that contribute to disease.
A Wide Array of Approaches to Reestablish Antigen-
Specific Tolerance
The overall objective of this strategy is to deliver bcell anti-
gens in particular ways such that their presentation in vivo
results in elimination or inactivation of antigen-specific diabe-
togenic T cells, or induction of antigen-specific immunoregula-
tory populations, to confer durable protection from
autoimmunity without compromising the general immunosur-
veillance for infectious agents and malignant cells. The tradi-
tional method has been to administer protein antigens via
tolerogenic routes (mainly oral or intranasal insulin and
GAD65/Alum), but this approach has not produced significant
clinical benefit in recent onset patients [67]. Because of lack
of adverse side effects, these therapies are now being tested
in secondary prevention trials (i.e., in patients with ongoing
autoimmunity evidenced by circulating autoantibodies) (Table
1). It is worth pointing out that oral insulin has also been
tested in a primary prevention trial (in young subject with no
evidence of autoimmunity, Pre-POINT trial, Table 1) and data
suggest that insulin-specific Tregs were induced at the highest
dose [68]. Antigens coupled with apoptotic cells have been
known for several decades to be very tolerogenic and showed
efficacy in preclinical models of T1D [69]. This strategy has
now been tested in patients with multiple sclerosis and was
well tolerated [70]. Massive apoptosis resulting from deple-
tion of B cells and CD8
1
T cells (using a short course of bio-
logics) is accompanied by release of TGF-b, which combined
with exogenous antigens such as GAD65 peptides, supports
the generation of protective Tregs, because CD4
1
T cells are
left untouched and available for conversion [71]. This promis-
ing approach validated in mouse models of T1D and multiple
sclerosis remains to be tested for safety in humans.
A less conventional alternative to protein antigen delivery
lets the body produce specific antigens in cells or sites
Table 3. Main clinical trials using low-dose IL-2 or cell-based therapies in recent onset T1D patients
New onset trials Drug Type of study
Low-dose IL-2
NCT01827735 (DILT1D) Proleukin (IL-2) Phase 1/2, onset <24 months
NCT02265809 (DILfrequency) Aldesleukin (IL-2) Phase 1/2, onset <60 months
NCT01353833 (DF-IL2) Aldesleukin (IL-2) Phase 1/2, onset <24 months
NCT01862120 (DFIL2-Child) IL-2 Phase 2, recent onset
NCT02411253 (DIABIL-2) rhIL-2 Phase 2, recent onset
Cell-based therapies
ISRCTN06128462 (Gdansk) Polyclonal Tregs Phase 1, onset <2 months
NCT01210664 (UCSF) Polyclonal Tregs Phase 1, onset 3-24 months
NCT00445913 (Pittsburgh) Autologous DCs Phase 1, long-term T1D (5y1)
NCT02354911 (Pittsburgh) Autologous DCs Phase 2, new onset <100d
NCT01068951 (Uppsala) MSCs Phase 1, new onset
NCT00690066 (Mesoblast) Prochymal (MSCs) Phase 2, onset 2-20 wks
NCT02057211 (Uppsala) MSCs Phase 2, new onset <3 weeks
NCT01322789 (Sao Paulo) MSCs Phase 1/2, new onset <6 weeks
NCT00305344 (Florida) Umbilical cord blood (UCB) Phase 1/2, post-onset
NCT00989547 (Munich) Umbilical cord blood (UCB) Phase 1, post-onset
NCT01350219 (Tianhe) UCB-derived stem cells Phase 2, post-onset
NCT01996228 (Tianhe) UCB-derived stem cells Phase 1/2, post-onset
NCT00315133 (Sao Paulo) Autologous HSCs Phase 1/2, onset <12 weeks
NCT01285934 (Northwestern) Autologous HSCs Phase 1/2, onset <5 months
Note: Clinical trials are color-shaded based on whether they are ,,or Stem cells used for the generation of new b
cells are not covered here. DCs: dendritic cells; HSCs: hematopoietic stem cells; MSCs: mesenchymal stem/stromal cells; Tregs: regulatory T cells.
Creusot, Battaglia, Roncarolo et al. 5
www.StemCells.com V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press
amenable for tolerance induction following gene transfer [72].
Plasmid DNA encoding autoantigens such as insulin or its
InsB
9-23
immunodominant peptide prevented disease in NOD
mice [73–75] and was given to recent-onset T1D patients in a
phase 1 trial [76]. Data from this trial demonstrated both
safety and diminution of insulin-reactive CD8
1
T cells, thus
tolerogenic DNA vaccines merit consideration for prevention
trials. Delivery of autoantigens by viral vectors used for gene
therapy has also been explored [77, 78]. One legitimate con-
cern when using viral components is the inadvertent activa-
tion of antigen-specific effector T cells that could exacerbate
bcell autoimmunity, especially if expression with ubiquitous
promoters is allowed in professional antigen-presenting cells
(APCs) than can mature and become immunogenic [79]. The
insertion of a microRNA-142 target sequence to abrogate
transgene expression in professional APCs and other hemato-
poietic lineage cells, together with the use of a liver-specific
promoter resulted in a lentiviral vector, which specifically tar-
gets the antigens to hepatocytes [79, 80]. Treatment with
such a vector encoding the InsB
9-23
peptide-induced some
antigen-specific effector T cells but also antigen-specific CD4
1
FoxP3
1
Tregs, which halted islet immune cell infiltration, and
protected mice from T1D [79]. When combined with a single
suboptimal dose of anti-CD3 mAb, it was effective in reversing
hyperglycemia after onset in a Treg-dependent manner [79].
The use of nonintegrating forms of lentiviral vectors will offer
an additional level of safety when implementing such an
approach clinically [81].
So far, antigen-specific therapies for T1D have been
proved efficient in mouse models and to be among the safest
in patients, but evidence for clinical efficacy is lacking. One
possible reason may have to do with the choice of bcell anti-
gens used, which is limited in two aspects: (1) only a single
antigen (mostly insulin or GAD65) is used despite the evi-
dence of epitope spreading reflected by different types of
autoantibodies, and (2) only native antigens are used while it
has become increasingly clear that many diabetogenic T cells
respond to post-translationally modified or processed neo
antigens. Accomplishing long-term antigen-specific tolerance,
whether it is with Tregs or other regulatory cells will require
these issues to be addressed.
CELL-BASED THERAPIES:CURRENT AND FUTURE APPLICATIONS
Cell-based therapies are individualized approaches that cur-
rently involve the transfer of autologous cells that have
immunoregulatory properties and can provide a counterbal-
ance for effector T cells that mediate bcell destruction. While
certain drugs aim at expanding and potentiating Tregs in vivo
(see low dose IL-2 above), cell-based therapies generally
involve Tregs or cells that have the ability to induce or poten-
tiate such immunoregulatory populations in vivo. We will also
discuss the use of different types of stem cells as part of cell-
based therapies to block autoimmune responses, but we will
not cover the generation of new bcells from stem cells for
transplantation, which is reviewed elsewhere [82].
Regulatory T Cells (Tregs)
Several preclinical animal studies have established that the
adoptive transfer of Tregs can prevent various autoimmune
diseases, T1D included. However, only a few studies showed
that Treg cell transfer is efficacious in reverting active dis-
ease and, when it occurred, the transferred cells needed to
be antigen-specific [83–85]. Based on this evidence, Tregs
are now being used in phase 1/2 studies in patients with
autoimmune diseases [86]. Increasing doses of autologous
ex vivo–expanded polyclonal CD4
1
CD25
1
Tregs have been
used safely in newly diagnosed T1D patients [87]. These
studies were instrumental in demonstrating the safety and
feasibility of such a complex approach of personalized medi-
cine, but efficacy has still to be demonstrated in larger
trials.
Based on the data in animal models, to be of any thera-
peutic use, Tregs have to be transferred prior to overt hyper-
glycemia or have to be antigen-specific. It has been, up to
now, difficult to obtain sufficient antigen-specific CD4
1
FoxP3
1
Tregs, but this might be more feasible by inducing
Tregs de novo in vitro. Such an approach has been used in
the context of allogeneic hematopoietic stem cell transplanta-
tion to prevent GvHD where host-specific Tr1 cells were gen-
erated in vitro from donor peripheral blood and transferred
to transplanted hosts [88]. The induction of self-specific Tr1
cells in NOD mice in vivo is feasible and they protect from
diabetes development [89]. However, the generation of
human diabetes-related antigen-specific Tregs in vitro has yet
to be achieved. Studies on adoptive T cell therapy have dem-
onstrated the possibility of engineering T cells using lentivirus,
either by expressing a relevant (bcell antigen-specific) T cell
receptor into polyclonal Tregs [90] or by overexpressing FoxP3
in T cells [91], potentially in antigen-specific T cells that have
been enriched and ex vivo expanded.
Regulatory B Cells (Bregs)
Recently, the IL-10–producing regulatory Bregs have attracted
attention as being altered in autoimmune diseases and thus
represent another potential tool for cell therapy [92]. As with
Tregs, their numbers and function might be compromised in
T1D patients [93], indicating the possibility of using Breg ther-
apy, alone or in concert with Tregs. However, the importance
of antigen-specificity in this case is not clear, and considering
that we are still at the beginning of cell therapy with Tregs,
using Bregs is even more futuristic.
Dendritic Cells
As the most specialized of APCs, dendritic cells (DCs) have
long been a candidate of choice for their ability to engage T
cells through presentation of bcell antigens, and under a tol-
erogenic phenotype, to achieve deletion or inactivation of dia-
betogenic T cells, converting them into Tregs or restimulating
preexisting Tregs. DC infusions have shown remarkable effi-
cacy in numerous preclinical studies, even in the absence of
exogenously provided antigens [94]. The first clinical trial using
autologous DCs in recent onset T1D patients demonstrated
both safety and the potential to induce Bregs [95]. In this
phase 1 study, the first of its kind for the treatment of auto-
immune diseases, the monocyte-derived DCs were locked in a
nonimmunogenic state by silencing important costimulatory
molecules (CD40, CD80, and CD86), but were not provided
exogenous antigen. Autoantigen expression and maturation
stage are two crucial considerations, because immunogenic
DCs expressing bcell antigens could boost autoimmune T cell
6Unconventional approaches to treat Type 1 diabetes
V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press STEM CELLS
responses against bcells. The antigen-specific therapies previ-
ously described rely on the acquisition and presentation of
relevant antigens by tolerogenic APCs that are not that well
characterized but possibly comprising different subsets of DCs
and other types of APCs. In the case of the recent DC trial,
the mechanism of action is not completely understood as no
exogenous antigen was provided, and whether these DCs
could pick up and present relevant autoantigens in vivo
remains unclear. Furthermore, there is evidence that DCs
expressing costimulatory molecules but not inflammatory
cytokines (termed semimature DCs) may induce tolerance as
well [96].
Another phase 1 trial employing tolerogenic DCs pulsed
with citrullinated peptides for the treatment of rheumatoid
arthritis has demonstrated safety as well as immunological
responses reflective of regulation [97], suggesting that provi-
sion of autoantigens may not lead to exacerbated responses
as long as the DCs are maintained tolerogenic, which in this
case was achieved by pretreatment with an NF-jB inhibitor.
An alternative or complementary approach to silencing the
expression of costimulatory genes is the overexpression of tol-
erogenic products for which the list is long and includes
immunoregulatory cytokines, inhibitory ligands, and
metabolism-altering enzymes [98]. A safe and clinically viable
way to overexpress genes and achieve a significant therapeu-
tic outcome is by mRNA electroporation, which has been
used widely for autologous DC therapy in cell therapy of can-
cer [99, 100]. This method of modification allows for coex-
pression of multiple products of interest in the same cell and
at the same time, including relevant antigens for added speci-
ficity if desired [101]. Although transient, expression of genes
in DCs by mRNA is sufficient to induce long-lasting responses
resulting in prevention of T1D in mice [102]. A follow-up trial
of the first safety study of autologous DCs in T1D patients is
set to begin in the near future.
Mesenchymal Stem/Stromal Cells
Mesenchymal stem/stromal cells (MSCs) are endowed with
regenerative and immunosuppressive properties that have
fueled their popularity in cell therapy, yet controversies
remain regarding their name and definition [103]. Although
they can generally suppress immune responses on their own
in a nonspecific manner, they have also been shown to induce
or expand Tregs, including in preclinical T1D studies
[104–106]. Because MSCs are nonprofessional APCs, it is
unclear if and how they specifically interact with Tregs and
diabetogenic T cells, and their effect may be indirect through
inflammation relief [107]. Unlike DCs, MSCs are nonimmuno-
genic, and can provide protection even in an allogeneic host,
which makes them attractive for the clinic [108]. They have
been proven to be well-tolerated in T1D patients whether
they were isolated by bone marrow aspiration [109] or from
adipose tissue [110], and associated with improvement of dis-
ease parameters such as C-peptide preservation. Careful char-
acterization of the phenotype and properties of MSCs used in
cell therapy is crucial to demonstrate consistency between
studies and draw meaningful conclusions, regardless of the
source of the cells, isolation, and culture conditions (Table 4).
Two follow-up clinical trials are currently recruiting in Sweden
and Brazil to demonstrate long-term efficacy (Table 3). All tri-
als so far are being conducted in recently diagnosed T1D
patients using MSCs that are either autologous or from first-
degree relatives. Although no serious side effects have been
reported so far, there remains a concern that slow growing
tumors may appear in the long term in some patients receiv-
ing autologous cells, according to some preclinical studies
[108].
Umbilical Cord Blood
Although this approach is limited to a few individuals who
have banked samples, umbilical cord blood (UCB) is a great
source of abundant MSCs and Tregs, which might work in syn-
ergy when infused into patients. As many important self-
reactive Tregs appear to be released early in life [111], these
Tregs may also include more antigen-specific Tregs of rele-
vance. Autologous UCB infusion was found to be safe but did
not have any significant therapeutic effect despite increased
numbers of Tregs [112, 113] and even with oral docosahexae-
noic acid and vitamin D supplementation [114]. Another
phase 1 trial, also in new onset pediatric subjects, is well
under way in Germany (Table 3). As previously mentioned, it
is possible that therapies involving Tregs would be more
effective when applied prior to disease onset. In that model,
the Cord Reinfusion in Diabetes (CoRD) study, enrolling high-
risk children with banked UCB, was recently initiated in Aus-
tralia and represents the first cell-based therapy used for sec-
ondary prevention of diabetes [115]. A distinct process is
being tested in China, whereby lymphocytes are obtained
from the blood by leukapheresis, “reeducated” ex vivo in con-
tact with UCB-derived stem cells, and then reinfused into the
patient. These studies suggest that this treatment improved
preservation of bcell function without notable adverse
effects, but caution must be exercised in the interpretation of
these studies, which were improperly controlled [116].
Hematopoietic Stem Cells
Perhaps the most drastic of all cell therapies consists of a
major immunological reset with the transient ablation of
Table 4. Phenotype of MSCs used in T1D studies
Source CD11c CD14 CD29 CD31 CD34 CD44 CD45 CD73 CD90 CD105 Reference
Mouse bone marrow 2212 121 1 [104]
Mouse bone marrow 112121[108]
Mouse adipose tissue 2 21211 1 [105]
Human bone marrow 2222111[109]
Human bone marrow 21 21211 1 [*]
Human adipose tissue 211 [110]
Note: Additional characterization may include ability to terminally differentiate (e.g., adipocytes) or to suppress T cell responses. [*] Prochymal
MSCs (NCT00690066).
Creusot, Battaglia, Roncarolo et al. 7
www.StemCells.com V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press
circulating T cells and their replacement with hematopoietic
stem cells (HSCs). Multiple completed studies have involved
autologous HSCs mobilized from peripheral blood and admin-
istered after a nonmyeloablative regimen consisting of cyclo-
phosphamide and anti-thymocyte globulin [117–119]. This
treatment performed in new-onset T1D patients demon-
strated a remarkable ability to normalize glycemia in a major-
ity of the subjects. Independence from insulin lasted between
several months and several years, up to 3-4 years (as
reported in the last meeting of the Immunology of Diabetes
Society), and a larger trial is now enrolling patients (Clinical-
Trial.gov NCT01285934). However, this therapy is fraught with
considerable side effects associated with the nonmyeloabla-
tive regimen [117–119], which make this approach unattrac-
tive in its current form to many prospective patients and
precludes its use for prevention. Furthermore, the contribu-
tion of HSCs in maintaining normoglycemia is unclear as the
immunosuppressive effect of the regimen may account for
part if not all of the therapeutic benefit. Finally, new T cells
(and other immune cells) generated from autologous HSCs
would still carry any inherent genetic defects that may play a
role in disease etiology. Transplantation of bone marrow-
derived allogeneic HSCs with induction of mixed chimerism
has also been tested in a minority of patients with autoim-
munity, including T1D [120]. The use of HSCs in combination
with islet transplant to induce chimerism and immunological
tolerance has been tested in a recent trial at the University of
Miami based on campath-1H and infusion of donor
CD34
1
HSCs (ClinicalTrials.gov NCT00315614), but did not
show any significant benefit. Although allogeneic HSCs from a
compatible and healthy donor may help correct some genetic
abnormalities, this must be preceded by high doses of chemo-
therapy and radiation to ablate the patient’s bone marrow
and is followed by prolonged immunosuppression to prevent
GvHD. The consequent transplant-related morbidity and mor-
tality limit this approach to patients with concomitant hema-
tological malignancies [121]. It should be noted that
preclinical data with purified allogeneic CD34
1
CD90
1
HSCs
showed complete reversion of T1D in the absence of GvHD
[122]. In addition, novel biologicals are under investigation for
use as safer and less toxic drugs to myeloablate the patient’s
bone marrow [123]. Thus, as safer and more effective HSC
transplantation protocols become available, allogeneic HSCs
might also be indicated in T1D. A more detailed review of the
different applications of stem cells (including MSCs and UCB)
to treat T1D can be found elsewhere [124].
In parallel to efforts in generating insulin-producing cells
from stem cells (embryonic stem cells or induced pluripotent
stem cells) [82], there is an expanded interest in growing tis-
sues specialized in tolerance induction, such as thymic tissue
[125, 126]. Although much can be learned from these studies,
the clinical implementation of such advances is elusive,
including where and how to implant the new tissue.
CONCLUSIONS
A variety of original therapeutic strategies for treating or pre-
venting T1D have emerged in the past decade, with the latest
approaches clearly dominated by cell-based therapies. As the
least expensive and most conventional therapies have failed
to deliver efficient and durable protection from diabetogenic
immune responses, testing of more expensive, and individual-
ized therapies has become justified as long as preclinical stud-
ies indicate a strong prospect of durable efficacy achieved
with a minimal number of treatments. Strategies that are
more antigen-specific and less immunosuppressive tend to
have the best safety profile, and their poor efficacy in new
onset patients should not discourage evaluation in prevention
trials in high-risk patients in which they might perform sur-
prisingly well. The enrollment of subjects for prevention stud-
ies should be guided by more refined biomarkers, which may
help the diabetes community to better understand the under-
lying defects behind the autoimmune response in each
patient, and better tailor the treatment type, dose, and tim-
ing. When appropriate, two or more of these therapies may
be combined in order to address multiple defects and benefit
a larger number of patients. A clear advantage of cell-based
therapies is that they can perform multiple tasks. For exam-
ple, one can envision tolerogenic APCs engineered to express
selected bcell antigens (to specifically engage diabetogenic T
cells), additional immunoregulatory ligands or cytokines (to
potentiate T cell deletion or Treg induction), homing mole-
cules (for targeting to inflamed islets or their draining lymph
nodes), anti-inflammatory cytokines (to quench inflammation),
and even growth factors to promote bcell replication.
ACKNOWLEDGMENTS
RJC is currently funded by the National Institutes of Health
(NIH), the Diabetes & Endocrinology Research Center (Colum-
bia University), and the Irving Institute for Clinical and Trans-
lational Research; MB by the Italian Ministry of Health, the
European Community, the Cariplo Foundation, and the Juve-
nile Diabetes Research Foundation (JDRF); MGR by the Italian
Telethon Foundation and JDRF; and CGF by the NIH and JDRF.
AUTHOR CONTRIBUTIONS
All authors wrote and edited the manuscript
REFERENCES
1Diamond Project Group. Incidence and
trends of childhood Type 1 diabetes world-
wide 1990-1999. Diabet Med 2006;23:857-
866.
2Patterson CC, Dahlquist GG, Gyurus E
et al. Incidence trends for childhood type 1
diabetes in Europe during 1989-2003 and
predicted new cases 2005-20: A multicentre
prospective registration study. Lancet 2009;
373:2027-2033.
3Bottazzo GF, Florin-Christensen A,
Doniach D. Islet-cell antibodies in diabetes
mellitus with autoimmune polyendocrine
deficiencies. Lancet 1974;2:1279-1283.
4Eisenbarth GS. Type I diabetes mellitus.
A chronic autoimmune disease. N Engl J Med
1986;314:1360-1368.
5Feutren G, Papoz L, Assan R et al. Cyclo-
sporin increases the rate and length of
remissions in insulin-dependent diabetes of
recent onset. Results of a multicentre
double-blind trial. Lancet. 1986;2:119-124.
6Cyclosporin-induced remission of IDDM
after early intervention. Association of 1 yr
of cyclosporin treatment with enhanced insu-
lin secretion. The Canadian-European
8Unconventional approaches to treat Type 1 diabetes
V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press STEM CELLS
Randomized Control Trial Group. Diabetes
1988;37:1574-1582.
7Eisenbarth GS, Srikanta S, Jackson R
et al. Anti-thymocyte globulin and predni-
sone immunotherapy of recent onset type 1
diabetes mellitus. Diabetes Res 1985;2:271-
276.
8Todd JA. Etiology of type 1 diabetes.
Immunity 2010;32:457-467.
9Ziegler AG, Nepom GT. Prediction and
pathogenesis in type 1 diabetes. Immunity
2010;32:468-478.
10 Kaizer EC, Glaser CL, Chaussabel D et al.
Gene expression in peripheral blood mono-
nuclear cells from children with diabetes.
J Clin Endocrinol Metab 2007;92:3705-3711.
11 Jin Y, She JX. Novel biomarkers in type 1
diabetes. Rev Diabet Stud 2012;9:224-235.
12 Jin Y, Sharma A, Bai S et al. Risk of type
1 diabetes progression in islet autoantibody-
positive children can be further stratified
using expression patterns of multiple genes
implicated in peripheral blood lymphocyte
activation and function. Diabetes 2014;63:
2506-2515.
13 Kallionpaa H, Elo LL, Laajala E et al.
Innate immune activity is detected prior to
seroconversion in children with HLA-
conferred type 1 diabetes susceptibility. Dia-
betes 2014;63:2402-2414.
14 Tooley JE, Herold KC. Biomarkers in type
1 diabetes: Application to the clinical trial
setting. Curr Opin Endocrinol Diabetes Obes
2014;21:287-292.
15 Kulmala P, Savola K, Petersen JS et al.
Prediction of insulin-dependent diabetes mel-
litus in siblings of children with diabetes. A
population-based study. The Childhood Dia-
betes in Finland Study Group. J Clin Invest
1998;101:327-336.
16 LaGasse JM, Brantley MS, Leech NJ
et al. Successful prospective prediction of
type 1 diabetes in schoolchildren through
multiple defined autoantibodies: An 8-year
follow-up of the Washington State Diabetes
Prediction Study. Diabetes Care 2002;25:505-
511.
17 Bingley PJ, Gale EA, European Nicotina-
mide Diabetes Intervention Trial G. Progres-
sion to type 1 diabetes in islet cell antibody-
positive relatives in the European Nicotina-
mide Diabetes Intervention Trial: The role of
additional immune, genetic and metabolic
markers of risk. Diabetologia 2006;49:881-
890.
18 Sosenko JM, Krischer JP, Palmer JP et al.
A risk score for type 1 diabetes derived from
autoantibody-positive participants in the dia-
betes prevention trial-type 1. Diabetes Care
2008;31:528-533.
19 Akirav EM, Lebastchi J, Galvan EM et al.
Detection of beta cell death in diabetes using
differentially methylated circulating DNA.
Proc Natl Acad Sci USA 2011;108:19018-
19023.
20 Herold KC, Usmani-Brown S, Ghazi T
et al. Beta cell death and dysfunction during
type 1 diabetes development in at-risk indi-
viduals. J Clin Invest 2015;125:1163-1173.
21 Rakyan VK, Beyan H, Down TA et al.
Identification of type 1 diabetes-associated
DNA methylation variable positions that pre-
cede disease diagnosis. PLoS Genet 2011;7:
e1002300.
22 Gaglia JL, Harisinghani M, Aganj I et al.
Noninvasive mapping of pancreatic inflamma-
tion in recent-onset type-1 diabetes patients.
Proc Natl Acad Sci USA 2015;112:2139-2144.
23 Shibasaki S, Imagawa A, Hanafusa T. Ful-
minant type 1 diabetes mellitus: A new class
of type 1 diabetes. Adv Exp Med Biol 2012;
771:20-23.
24 Naik RG, Brooks-Worrell BM, Palmer JP.
Latent autoimmune diabetes in adults. J Clin
Endocrinol Metab 2009;94:4635-4644.
25 Cerosaletti K, Buckner JH. Protein tyro-
sine phosphatases and type 1 diabetes:
Genetic and functional implications of PTPN2
and PTPN22. Rev Diabet Stud 2012;9:188-
200.
26 Herold KC, Brooks-Worrell B, Palmer J
et al. Validity and reproducibility of measure-
ment of islet autoreactivity by T-cell assays in
subjects with early type 1 diabetes. Diabetes
2009;58:2588-2595.
27 Rich SS, Concannon P. Role of type 1
diabetes-associated SNPs on autoantibody
positivity in the type 1 diabetes genetics
consortium: Overview. Diabetes Care 2015;
38(Suppl 2):S1-S3.
28 Pugliese A, Zeller M, Fernandez A, Jr.
et al. The insulin gene is transcribed in the
human thymus and transcription levels corre-
lated with allelic variation at the INS VNTR-
IDDM2 susceptibility locus for type 1 diabe-
tes. Nat Genet 1997;15:293-297.
29 Vafiadis P, Bennett ST, Todd JA et al.
Insulin expression in human thymus is modu-
lated by INS VNTR alleles at the IDDM2
locus. Nat Genet 1997;15:289-292.
30 Lawson JM, Tremble J, Dayan C et al.
Increased resistance to CD41CD25hi regula-
tory T cell-mediated suppression in patients
with type 1 diabetes. Clin Exp Immunol
2008;154:353-359.
31 Ferraro A, Socci C, Stabilini 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 2011;60:2903-
2913.
32 Garg G, Tyler JR, Yang JH et al. Type 1
diabetes-associated IL2RA variation lowers IL-
2 signaling and contributes to diminished
CD41CD25 1regulatory T cell function.
J Immunol 2012;188:4644-4653.
33 Schneider A, Rieck M, Sanda S et al. The
effector T cells of diabetic subjects are resist-
ant to regulation via
CD4 1FOXP3 1regulatory T cells. J Immunol
2008;181:7350-7355.
34 de St Groth BF. Regulatory T-cell abnor-
malities and the global epidemic of immuno-
inflammatory disease. Immunol Cell Biol
2012;90:256-259.
35 Adamczak DM, Nowak JK, Frydrychowicz
M et al. The role of Toll-like receptors and
vitamin D in diabetes mellitus type 1--A
review. Scand J Immunol 2014;80:75-84.
36 McClymont SA, Putnam AL, Lee MR
et al. Plasticity of human regulatory T cells in
healthy subjects and patients with type 1
diabetes. J Immunol 2011;186:3918-3926.
37 Mollah ZU, Pai S, Moore C et al. Abnor-
mal NF-kappa B function characterizes
human type 1 diabetes dendritic cells and
monocytes. J Immunol 2008;180:3166-3175.
38 Badami E, Sorini C, Coccia M et al.
Defective differentiation of regulatory
FoxP3 1T cells by small-intestinal dendritic
cells in patients with type 1 diabetes. Diabe-
tes 2011;60:2120-2124.
39 Fisher MM, Watkins RA, Blum J et al.
Elevations in circulating methylated and
unmethylated preproinsulin DNA in new-
onset type 1 diabetes. Diabetes 2015;64:
3867-3872.
40 McGinty JW, Marre ML, Bajzik V et al. T
cell epitopes and post-translationally modi-
fied epitopes in type 1 diabetes. Curr Diabe-
tes Rep 2015;15:90.
41 Talchai C, Xuan S, Lin HV et al. Pancre-
atic beta cell dedifferentiation as a mecha-
nism of diabetic beta cell failure. Cell 2012;
150:1223-1234.
42 Diana J, Simoni Y, Furio L et al. Crosstalk
between neutrophils, B-1a cells and plasma-
cytoid dendritic cells initiates autoimmune
diabetes. Nat Med 2013;19:65-73.
43 Mathis D, Vence L, Benoist C. Beta-cell
death during progression to diabetes. Nature
2001;414:792-798.
44 Xu GJ, Kula T, Xu Q et al. Viral immunol-
ogy. Comprehensive serological profiling of
human populations using a synthetic human
virome. Science 2015;348:aaa0698.
45 Ferreira RC, Guo H, Coulson RM et al. A
type I interferon transcriptional signature
precedes autoimmunity in children geneti-
cally at risk for type 1 diabetes. Diabetes
2014;63:2538-2550.
46 Krogvold L, Edwin B, Buanes T et al.
Detection of a low-grade enteroviral infection
in the islets of langerhans of living patients
newly diagnosed with type 1 diabetes. Diabe-
tes 2015;64:1682-1687.
47 Pane JA, Coulson BS. Lessons from the
mouse: Potential contribution of bystander
lymphocyte activation by viruses to human
type 1 diabetes. Diabetologia 2015;58:1149-
1159.
48 Kostic AD, Gevers D, Siljander H et al.
The dynamics of the human infant gut micro-
biome in development and in progression
toward type 1 diabetes. Cell Host Microbe
2015;17:260-273.
49 Herold KC, Hagopian W, Auger JA et al.
Anti-CD3 monoclonal antibody in new-onset
type 1 diabetes mellitus. N Engl J Med 2002;
346:1692-1698.
50 Herold KC, Gitelman SE, Masharani U
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 2005;54:
1763-1769.
51 Keymeulen B, Vandemeulebroucke E,
Ziegler AG et al. Insulin needs after CD3-
antibody therapy in new-onset type 1 diabe-
tes. N Engl J Med 2005;352:2598-2608.
52 Pescovitz MD, Greenbaum CJ, Krause-
Steinrauf H et al. Rituximab, B-lymphocyte
depletion, and preservation of beta-cell func-
tion. New Engl J Med 2009;361:2143-2152.
53 Orban T, Bundy B, Becker DJ et al. Co-
stimulation modulation with abatacept in
patients with recent-onset type 1 diabetes: A
randomised, double-blind, placebo-controlled
trial. Lancet 2011;378:412-419.
Creusot, Battaglia, Roncarolo et al. 9
www.StemCells.com V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press
54 Yu A, Snowhite I, Vendrame F et al.
Selective IL-2 responsiveness of regulatory T
cells through multiple intrinsic mechanisms
supports the use of low-dose IL-2 therapy in
type 1 diabetes. Diabetes 2015;64:2172-
2183.
55 Koreth J, Matsuoka K, Kim HT et al.
Interleukin-2 and regulatory T cells in graft-
versus-host disease. N Engl J Med 2011;365:
2055-2066.
56 Matsuoka K, Koreth J, Kim HT et al.
Low-dose interleukin-2 therapy restores regu-
latory T cell homeostasis in patients with
chronic graft-versus-host disease. Sci Transl
Med 2013;5:179ra143.
57 Kennedy-Nasser AA, Ku S, Castillo-Caro P
et al. Ultra low-dose IL-2 for GVHD prophy-
laxis after allogeneic hematopoietic stem cell
transplantation mediates expansion of regu-
latory T cells without diminishing antiviral
and antileukemic activity. Clin Cancer Res
2014;20:2215-2225.
58 Saadoun D, Rosenzwajg M, Joly F et al.
Regulatory T-cell responses to low-dose inter-
leukin-2 in HCV-induced vasculitis. N Engl J
Med 2011;365:2067-2077.
59 Oo YH, Mutimer D, Adams DH. Low-
dose interleukin-2 and HCV-induced vasculi-
tis. N Engl J Med 2012;366:1353-1354;
author reply 1354.
60 Hartemann A, Bensimon G, Payan CA
et al. Low-dose interleukin 2 in patients with
type 1 diabetes: A phase 1/2 randomised,
double-blind, placebo-controlled trial. Lancet
Diabetes Endocrinol 2013;1:295-305.
61 Grinberg-Bleyer Y, Baeyens A, You S
et al. IL-2 reverses established type 1 diabe-
tes in NOD mice by a local effect on pancre-
atic regulatory T cells. J Exp Med 2010;207:
1871-1878.
62 Kukreja A, Cost G, Marker J et al. Multi-
ple immuno-regulatory defects in type-1 dia-
betes. J Clin Invest 2002;109:131-140.
63 Lindley S, Dayan C, Bishop A et al. Defec-
tive suppressor function in CD4(1)CD25(1)T-
cells from patients with type 1 diabetes. Dia-
betes 2005;54:92-99.
64 Putnam AL, Vendrame F, Dotta F et al.
CD41CD25 high regulatory T cells in human
autoimmune diabetes. J Autoimmun 2005;24:
55-62.
65 Hughson A, Bromberg I, Johnson B et al.
Uncoupling of proliferation and cytokines from
suppression within the CD41CD251Foxp3 1T-
cell compartment in the 1st year of human type
1 diabetes. Diabetes 2011;60:2125-2133.
66 Kaye WA, Adri MN, Soeldner JS et al.
Acquired defect in interleukin-2 production
in patients with type I diabetes mellitus. N
Engl J Med 1986;315:920-924.
67 Coppieters KT, Harrison LC, von Herrath
MG. Trials in type 1 diabetes: Antigen-
specific therapies. Clin Immunol 2013;149:
345-355.
68 Bonifacio E, Ziegler AG, Klingensmith G
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 2015;313:1541-1549.
69 Prasad S, Xu D, Miller SD. Tolerance
strategies employing antigen-coupled apopto-
tic cells and carboxylated PLG nanoparticles
for the treatment of type 1 diabetes. Rev
Diabet Stud 2012;9:319-327.
70 Lutterotti A, Yousef S, Sputtek A et al.
Antigen-specific tolerance by autologous
myelin peptide-coupled cells: A phase 1 trial
in multiple sclerosis. Sci Transl Med 2013;5:
188ra175.
71 Kasagi S, Zhang P, Che L et al. In vivo-
generated antigen-specific regulatory T cells
treat autoimmunity without compromising
antibacterial immune response. Sci Transl
Med 2014;6:241ra278.
72 Wasserfall CH, Herzog RW. Gene therapy
approaches to induce tolerance in autoim-
munity by reshaping the immune system.
Curr Opin Investig Drugs 2009;10:1143-1150.
73 Bot A, Smith D, Bot S et al. Plasmid vac-
cination with insulin B chain prevents auto-
immune diabetes in nonobese diabetic mice.
J Immunol 2001;167:2950-2955.
74 Chang Y, Yap S, Ge X et al. DNA vaccina-
tion with an insulin construct and a chimeric
protein binding to both CTLA4 and CD40
ameliorates type 1 diabetes in NOD mice.
Gene Ther 2005;12:1679-1685.
75 Solvason N, Lou YP, Peters W et al.
Improved efficacy of a tolerizing DNA vaccine
for reversal of hyperglycemia through
enhancement of gene expression and local-
ization to intracellular sites. J Immunol 2008;
181:8298-8307.
76 Roep BO, Solvason N, Gottlieb PA et al.
Plasmid-encoded proinsulin preserves C-
peptide while specifically reducing proinsulin-
specific CD8(1) T cells in type 1 diabetes. Sci
Transl Med 2013;5:191ra182.
77 Han G, Wang R, Chen G et al. Gene
delivery GAD 500 autoantigen by AAV sero-
type 1 prevented diabetes in NOD mice:
Transduction efficiency do not play important
roles. Immunol Lett 2008;115:110-116.
78 Luth S, Huber S, Schramm C et al.
Ectopic expression of neural autoantigen in
mouse liver suppresses experimental auto-
immune neuroinflammation by inducing
antigen-specific Tregs. J Clin Invest 2008;118:
3403-3410.
79 Akbarpour M, Goudy KS, Cantore A
et al. Insulin B chain 9-23 gene transfer to
hepatocytes protects from type 1 diabetes
by inducing Ag-specific FoxP3 1Tregs. Sci
Transl Med 2015;7:289ra281.
80 Brown BD, Venneri MA, Zingale A et al.
Endogenous microRNA regulation suppresses
transgene expression in hematopoietic line-
ages and enables stable gene transfer. Nat
Med 2006;12:585-591.
81 Annoni A, Goudy K, Akbarpour M et al.
Immune responses in liver-directed lentiviral
gene therapy. Transl Res 2013;161:230-240.
82 Johannesson B, Sui L, Freytes DO et al.
Toward beta cell replacement for diabetes.
EMBO J 2015;34:841-855.
83 Roncarolo MG, Battaglia M. Regulatory
T-cell immunotherapy for tolerance to self
antigens and alloantigens in humans. Nat
Rev Immunol 2007;7:585-598.
84 Tang Q, Henriksen KJ, Bi M et al. In
vitro-expanded antigen-specific regulatory T
cells suppress autoimmune diabetes. J Exp
Med 2004;199:1455-1465.
85 Tarbell KV, Yamazaki S, Olson K et al.
CD25 1CD4 1T cells, expanded with dendri-
tic cells presenting a single autoantigenic
peptide, suppress autoimmune diabetes.
J Exp Med 2004;199:1467-1477.
86 Trzonkowski P, Bacchetta R, Battaglia M
et al. Hurdles in therapy with regulatory T
cells. Sci Transl Med 2015;7:304ps318.
87 Marek-Trzonkowska N, Mysliwiec M,
Dobyszuk A et al. Therapy of type 1 diabetes
with CD4(1)CD25(high)CD127-regulatory T
cells prolongs survival of pancreatic islets -
Results of one year follow-up. Clin Immunol
2014;153:23-30.
88 Bacchetta R, Lucarelli B, Sartirana C et al.
Immunological outcome in haploidentical-HSC
transplanted patients treated with IL-10-
anergized donor T cells. Front Immunol 2014;
5:16.
89 Battaglia M, Stabilini A, Draghici E et al.
Induction of tolerance in type 1 diabetes via
both CD41CD25 1T regulatory cells and T
regulatory type 1 cells. Diabetes 2006;55:
1571-1580.
90 Brusko TM, Koya RC, Zhu S et al. Human
antigen-specific regulatory T cells generated
by T cell receptor gene transfer. PloS One
2010;5:e11726.
91 Allan SE, Alstad AN, Merindol N et al.
Generation of potent and stable human
CD4 1T regulatory cells by activation-
independent expression of FOXP3. Mol Ther
2008;16:194-202.
92 Tedder TF. B10 cells: A functionally
defined regulatory B cell subset. J Immunol
2015;194:1395-1401.
93 Kleffel S, Vergani A, Tezza S et al. Inter-
leukin-10 1regulatory B cells arise within
antigen-experienced CD40 1B cells to main-
tain tolerance to islet autoantigens. Diabetes
2015;64:158-171.
94 Creusot RJ, Giannoukakis N, Trucco M
et al. It’s time to bring dendritic cell therapy
to type 1 diabetes. Diabetes 2014;63:20-30.
95 Giannoukakis N, Phillips B, Finegold D
et al. Phase I (safety) study of autologous tol-
erogenic dendritic cells in type 1 diabetic
patients. Diabetes Care 2011;34:2026-2032.
96 Gordon JR, Ma Y, Churchman L et al.
Regulatory dendritic cells for immunotherapy
in immunologic diseases. Front Immunol
2014;5:7.
97 Benham H, Nel HJ, Law SC et al. Citrulli-
nated peptide dendritic cell immunotherapy
in HLA risk genotype-positive rheumatoid
arthritis patients. Sci Transl Med 2015;7:
290ra287.
98 Schinnerling K, Soto L, Garcia-Gonzalez P
et al. Skewing dendritic cell differentiation
towards a tolerogenic state for recovery of
tolerance in rheumatoid arthritis. Autoimmun
Rev 2015;14:517-527.
99 Kyte JA, Gaudernack G. Immuno-gene
therapy of cancer with tumour-mRNA trans-
fected dendritic cells. Cancer Immunol
Immunother 2006;55:1432-1442.
100 Shurin MR, Gregory M, Morris JC et al.
Genetically modified dendritic cells in cancer
immunotherapy: A better tomorrow? Expert
Opin Biol Ther 2010;10:1539-1553.
101 Bonehill A, Tuyaerts S, Van Nuffel A
et al. Enhancing the T-cell stimulatory
capacity of human dendritic cells by co-
electroporation with CD40L, CD70 and consti-
tutively active TLR4 encoding mRNA. Mol
Ther 2008;16:1170-1180.
102 Creusot RJ, Chang P, Healey DG et al. A
short pulse of IL-4 delivered by DCs electro-
porated with modified mRNA can both pre-
10 Unconventional approaches to treat Type 1 diabetes
V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press STEM CELLS
vent and treat autoimmune diabetes in NOD
mice. Mol Ther 2010;18:2112-2120.
103 Bianco P. “Mesenchymal” stem cells.
Annu Rev Cell Dev Biol 2014;30:677-704.
104 Madec AM, Mallone R, Afonso G et al.
Mesenchymal stem cells protect NOD mice
from diabetes by inducing regulatory T cells.
Diabetologia 2009;52:1391-1399.
105 Bassi EJ, Moraes-Vieira PM, Moreira-Sa
CS et al. Immune regulatory properties of
allogeneic adipose-derived mesenchymal
stem cells in the treatment of experimental
autoimmune diabetes. Diabetes 2012;61:
2534-2545.
106 Kota DJ, Wiggins LL, Yoon N et al. TSG-
6 produced by hMSCs delays the onset of
autoimmune diabetes by suppressing Th1
development and enhancing tolerogenicity.
Diabetes 2013;62:2048-2058.
107 Burr SP, Dazzi F, Garden OA. Mesenchy-
mal stromal cells and regulatory T cells: The
Yin and Yang of peripheral tolerance? Immu-
nol Cell Biol 2013;91:12-18.
108 Fiorina P, Jurewicz M, Augello A et al.
Immunomodulatory function of bone
marrow-derived mesenchymal stem cells in
experimental autoimmune type 1 diabetes.
J Immunol 2009;183:993-1004.
109 Carlsson PO, Schwarcz E, Korsgren O
et al. Preserved beta-cell function in type 1
diabetes by mesenchymal stromal cells. Dia-
betes 2015;64:587-592.
110 Thakkar UG, Trivedi HL, Vanikar AV
et al. Insulin-secreting adipose-derived mes-
enchymal stromal cells with bone marrow-
derived hematopoietic stem cells from auto-
logous and allogenic sources for type 1 dia-
betes mellitus. Cytotherapy 2015;17:940-947.
111 Yang S, Fujikado N, Kolodin D et al.
Immune tolerance. Regulatory T cells gener-
ated early in life play a distinct role in main-
taining self-tolerance. Science 2015;348:589-
594.
112 Haller MJ, Wasserfall CH, Hulme MA
et al. Autologous umbilical cord blood trans-
fusion in young children with type 1 diabetes
fails to preserve C-peptide. Diabetes Care
2011;34:2567-2569.
113 Giannopoulou EZ, Puff R, Beyerlein A
et al. Effect of a single autologous cord blood
infusion on beta-cell and immune function in
children with new onset type 1 diabetes: A
non-randomized, controlled trial. Pediatr Dia-
betes 2014;15:100-109.
114 Haller MJ, Wasserfall CH, Hulme MA
et al. Autologous umbilical cord blood infusion
followed by oral docosahexaenoic acid and
vitamin D supplementation for C-peptide pres-
ervation in children with Type 1 diabetes. Biol
Blood Marrow Transpl 2013;19:1126-1129.
115 Han MX, Craig ME. Research using
autologous cord blood - Time for a policy
change. Med J Aust 2013;199:288-299.
116 Zhao Y, Jiang Z, Zhao T et al. Reversal
of type 1 diabetes via islet beta cell regener-
ation following immune modulation by cord
blood-derived multipotent stem cells. BMC
Med 2012;10:3.
117 Voltarelli JC, Couri CE, Stracieri AB
et al. Autologous nonmyeloablative hemato-
poietic stem cell transplantation in newly
diagnosed type 1 diabetes mellitus. JAMA
2007;297:1568-1576.
118 Couri CE, Oliveira MC, Stracieri AB
et al. C-peptide levels and insulin independ-
ence following autologous nonmyeloablative
hematopoietic stem cell transplantation in
newly diagnosed type 1 diabetes mellitus.
JAMA 2009;301:1573-1579.
119 D’Addio F, Valderrama Vasquez A, Ben
Nasr M et al. Autologous nonmyeloablative
hematopoietic stem cell transplantation in
new-onset type 1 diabetes: A multicenter
analysis. Diabetes 2014;63:3041-3046.
120 Pasquini MC, Voltarelli J, Atkins HL
et al. Transplantation for autoimmune dis-
eases in North and South America: A report
of the Center for International Blood and
Marrow Transplant Research. Biol Blood Mar-
row Transpl 2012;18:1471-1478.
121 Battaglia M. Experiments by nature:
Lessons on type 1 diabetes. Tissue Antigens
2014;83:1-9.
122 Beilhack GF, Landa RR, Masek MA
et al. Prevention of type 1 diabetes with
major histocompatibility complex-compatible
and nonmarrow ablative hematopoietic stem
cell transplants. Diabetes 2005;54:1770-1779.
123 Czechowicz A, Kraft D, Weissman IL
et al. Efficient transplantation via antibody-
based clearance of hematopoietic stem cell
niches. Science 2007;318:1296-1299.
124 Fiorina P, Voltarelli J, Zavazava N. Immu-
nological applications of stem cells in type 1
diabetes. Endocr Rev 2011;32:725-754.
125 Sun X, Xu J, Lu H et al. Directed differ-
entiation of human embryonic stem cells
into thymic epithelial progenitor-like cells
reconstitutes the thymic microenvironment
in vivo. Cell Stem Cell 2013;13:230-236.
126 Parent AV, Russ HA, Khan IS et al. Genera-
tion of functional thymic epithelium from human
embryonic stem cells that supports host T cell
development. Cell Stem Cell 2013;13:219-229.
Creusot, Battaglia, Roncarolo et al. 11
www.StemCells.com V
C2016 The Authors STEM CELLS published by Wiley Periodicals, Inc. on behalf of AlphaMed Press
... [9][10][11] However, some clinical studies have shown limited benefit or no therapeutic value, particularly in diabetic patients. [12][13][14] It is shown that therapy with CD34 + cells, although beneficial in patients with nonischemic cardiomyopathy with normal glucose metabolism, is ineffective in diabetic patients for unknown reasons. 15 Although the exact cause(s) for the inconsistency in the results of clinical studies is unclear, the outcome of cell therapy could ultimately be associated with the number and function of delivered bMSCs. ...
Article
Full-text available
Background Therapy with mesenchymal stem cells remains a promising but challenging approach to critical limb ischemia in diabetes because of the dismal cell survival. Methods and Results Critical limb ischemia in type 2 diabetes mouse model was used to explore the impact of diabetic limb ischemia on the survival of bone marrow mesenchymal stromal cells (bMSCs). Inhibition of intracellular reactive oxygen species was achieved with concomitant overexpression of superoxide dismutase (SOD)‐1 and glutathione peroxidase‐1 in the transplanted bMSCs, and extracellular reactive oxygen species was attenuated using SOD‐3 overexpression and N‐acetylcysteine treatment. In vivo optical fluorescence imaging and laser Doppler perfusion imaging were used to track cell retention and determine blood flow in diabetic ischemic limb, respectively. Survival of the transplanted bMSCs was significantly decreased in diabetic ischemic limb compared with the control. In vitro study indicated that advanced glycation end products, not high glucose, significantly decreased the proliferation of bMSCs and increased their apoptosis associated with increased reactive oxygen species production and selective reduction of SOD‐1 and SOD‐3. In vivo study demonstrated that concomitant overexpression of SOD‐1, SOD‐3, and glutathione peroxidase‐1, or host treatment with N‐acetylcysteine, significantly enhanced in vivo survival of transplanted bMSCs, and improved critical limb ischemia in diabetic mice. Combination of triple antioxidant enzyme overexpression in bMSCs with host N‐acetylcysteine treatment further improved bMSC survival with enhanced circulatory and functional recovery from diabetic critical limb ischemia. Conclusions Simultaneous suppression of reactive oxygen species from transplanted bMSCs and host tissue could additively enhance bMSC survival in diabetic ischemic limb with increased therapeutic efficacy in diabetes.
Article
Background T regulatory type 1 (Tr1) cells are a class of regulatory T cells participating in peripheral tolerance, hence the rationale behind their testing in clinical trials in different disease settings. One of their applications is tolerance induction to allogeneic islets for long‐term diabetes‐free survival. Currently the cellular and molecular mechanisms that promote Tr1‐cell induction in vivo remain poorly understood. Methods We employed a mouse model of transplant tolerance where treatment with G‐CSF/Rapamycin induces permanent engraftment of allogeneic pancreatic islets in C57BL/6 mice via Tr1 cells. The innate composition of graft and spleen cells in tolerant mice was analyzed by flow cytometry. Graft phagocytic cells were co‐cultured with CD4⁺ T cells in vitro to test their ability to induce Tr1‐cell induction. Graft phagocytic cells were depleted in vivo at different time points during G‐CSF/rapamycin treatment, to identify their role in Tr1‐cell induction and consequently in graft survival. Results In the spleen, the site of Tr1‐cell induction, no differences in the frequencies of macrophages or DC were observed. In the graft, the site of antigen uptake, a high proportion of macrophages and not DC was detected in tolerant but not in rejecting mice. Graft‐infiltrating macrophages of G‐CSF/rapamycin‐treated mice had an M2 phenotype, characterized by higher CD206 expression and IL‐10 production, whereas splenic macrophages only had an increased CD206 expression. Graft‐infiltrating cells from G‐CSF/rapamycin‐treated mice induced Tr1‐cell expansion in vitro. Furthermore, Tr1‐cell induction was perturbed upon in vivo depletion of phagocytic cells, early and not late during treatment, leading to graft loss suggesting that macrophages play a key role in tolerance induction mediated by Tr1 cells. Conclusions/interpretation Taken together, in this mouse model of Tr1‐cell induced tolerance to allogeneic islets, M2 macrophages infiltrating the graft upon G‐CSF/rapamycin treatment, are key for Tr1‐cell induction. This work gives mechanistic insight into pharmacologically‐induced Tr1‐cell expansion in vivo in this stringent model of allogeneic transplantation.
Article
Full-text available
Type 1 diabetes (T1D) is a chronic T-cell mediated autoimmune disease characterized by destruction of beta cells. Although new data have better defined the complex etiology underling the interrelation of genetic and environmental factors in the natural history of T1D, relevant pieces of the puzzle still are missing. Genetic predisposition is mainly associated to some histocompatibility leukocyte antigen (HLA) alleles; however, recent data suggest that new as well as still unknown genes might better define the complex multigenetic risk of the disease. In addition to the genetic effects, the concordance in familial aggregation in T1D indicates a pivotal role of environmental factors in the course of the disease, facilitating autoantibodies production. JDRF has recently proposed a new early stage of T1D according to which the detection of two or more autoantibodies in the blood, might describe those children at increased risk of developing T1D during the following years. In contrast to the improvements reached by prediction models, to date primary, secondary and tertiary prevention have still failed to achieve a safe and efficacious intervention strategies. Anyway, the most recent progresses in this field pave the way for future studies, with the aim of preventing T1D in children.
Article
Full-text available
Interest in adoptive T-cell therapies has been ignited by the recent clinical success of genetically-modified T cells in the cancer immunotherapy space. In addition to immune targeting for malignancies, this approach is now being explored for the establishment of immune tolerance with regulatory T cells (Tregs). Herein, we will summarize the basic science and clinical results emanating from trials directed at inducing durable immune regulation through administration of Tregs. We will discuss some of the current challenges facing the field in terms of maximizing cell purity, stability and expansion capacity, while also achieving feasibility and GMP production. Indeed, recent advances in methodologies for Treg isolation, expansion, and optimal source materials represent important strides toward these considerations. Finally, we will review the emerging genetic and biomaterial-based approaches on the horizon for directing Treg specificity to augment tissue-targeting and regenerative medicine.
Article
Full-text available
Colonization of the fetal and infant gut microbiome results in dynamic changes in diversity, which can impact disease susceptibility. To examine the relationship between human gut microbiome dynamics throughout infancy and type 1 diabetes (T1D), we examined a cohort of 33 infants genetically predisposed to T1D. Modeling trajectories of microbial abundances through infancy revealed a subset of microbial relationships shared across most subjects. Although strain composition of a given species was highly variable between individuals, it was stable within individuals throughout infancy. Metabolic composition and metabolic pathway abundance remained constant across time. A marked drop in alpha-diversity was observed in T1D progressors in the time-window between seroconversion and T1D diagnosis, accompanied by spikes in inflammation-favoring organisms, gene functions, and serum and stool metabolites. This work identifies trends in the development of the human infant gut microbiome along with specific alterations that precede T1D onset and distinguish T1D progressors from non-progressors.
Article
Full-text available
Type 1 diabetes (T1D) is an autoimmune disease in which progressive loss of self-tolerance, evidenced by accumulation of auto-antibodies and auto-reactive T cells that recognize diverse self-proteins, leads to immune-mediated destruction of pancreatic beta cells and loss of insulin secretion. In this review, we discuss antigens and epitopes in T1D and the role that post-translational modifications play in circumventing tolerance mechanisms and increasing antigenic diversity. Emerging data suggest that, analogous to other autoimmune diseases such as rheumatoid arthritis and celiac disease, enzymatically modified epitopes are preferentially recognized in T1D. Modifying enzymes such as peptidyl deiminases and tissue transglutaminase are activated in response to beta cell stress, providing a mechanistic link between post-translational modification and interactions with the environment. Although studies of such responses in the at-risk population have been limited, current data suggests that breakdown in tolerance through post-translational modification represents an important checkpoint in the development of T1D.
Article
Full-text available
Improper activation of the immune system contributes to a variety of clinical conditions, including autoimmune and allergic diseases as well as solid organ and bone marrow transplantation. One approach to counteract this activation is through adoptive therapy with regulatory T cells (Tregs). Efforts to manufacture these cells have led to good maunfacturing practice–compliant protocols, and Treg products are entering early clinical trials. Here, we report the stance of the European Union Cooperation in Science and Technology Action BM1305, “Action to Focus and Accelerate Cell-based Tolerance-inducing Therapies—A FACTT,” which identifies hurdles hindering Treg clinical applications in Europe and provides possible solutions.
Article
Full-text available
Elevated ratios of circulating unmethylated to methylated preproinsulin (INS) DNA have been suggested to reflect β-cell death in type 1 diabetes (T1D). We tested the hypothesis that absolute levels (rather than ratios) of unmethylated and methylated INS DNA differ between new-onset T1D subjects and controls, and assessed longitudinal changes in these parameters. We used droplet digital PCR to measure levels of unmethylated and methylated INS DNA in serum from subjects at T1D onset, and at 8 weeks and one year post-onset. Compared to controls, levels of both unmethylated and methylated INS DNA were elevated at T1D onset. At 8 weeks post-onset, methylated INS DNA remained elevated, but unmethylated INS DNA fell. At one year post-onset, both unmethylated and methylated INS DNA returned to control levels. Subjects with obesity, type 2 diabetes (T2D), and autoimmune hepatitis exhibited lower levels of unmethylated and methylated INS compared to T1D subjects at-onset and no differences compared to controls. Our study shows that elevations in both unmethylated and methylated INS DNA occurs in new-onset T1D and that levels of these DNA species change during T1D evolution. Our work emphasizes the need to consider absolute levels of differentially methylated DNA species as potential biomarkers of disease. © 2015 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered.
Article
Full-text available
In animals, immunomodulatory dendritic cells (DCs) exposed to autoantigen can suppress experimental arthritis in an antigen-specific manner. In rheumatoid arthritis (RA), disease-specific anti-citrullinated peptide autoantibodies (ACPA or anti-CCP) are found in the serum of about 70% of RA patients and are strongly associated with HLA-DRB1 risk alleles. This study aimed to explore the safety and biological and clinical effects of autologous DCs modified with a nuclear factor κB (NF-κB) inhibitor exposed to four citrullinated peptide antigens, designated "Rheumavax," in a single-center, open-labeled, first-in-human phase 1 trial. Rheumavax was administered once intradermally at two progressive dose levels to 18 human leukocyte antigen (HLA) risk genotype-positive RA patients with citrullinated peptide-specific autoimmunity. Sixteen RA patients served as controls. Rheumavax was well tolerated: adverse events were grade 1 (of 4) severity. At 1 month after treatment, we observed a reduction in effector T cells and an increased ratio of regulatory to effector T cells; a reduction in serum interleukin-15 (IL-15), IL-29, CX3CL1, and CXCL11; and reduced T cell IL-6 responses to vimentin447-455-Cit450 relative to controls. Rheumavax did not induce disease flares in patients recruited with minimal disease activity, and DAS28 decreased within 1 month in Rheumavax-treated patients with active disease. This exploratory study demonstrates safety and biological activity of a single intradermal injection of autologous modified DCs exposed to citrullinated peptides, and provides rationale for further studies to assess clinical efficacy and antigen-specific effects of autoantigen immunomodulatory therapy in RA. Copyright © 2015, American Association for the Advancement of Science.
Article
Full-text available
The human virome plays important roles in health and immunity. However, current methods for detecting viral infections and antiviral responses have limited throughput and coverage. Here, we present VirScan, a high-throughput method to comprehensively analyze antiviral antibodies using immunoprecipitation and massively parallel DNA sequencing of a bacteriophage library displaying proteome-wide peptides from all human viruses. We assayed over 10(8) antibody-peptide interactions in 569 humans across four continents, nearly doubling the number of previously established viral epitopes. We detected antibodies to an average of 10 viral species per person and 84 species in at least two individuals. Although rates of specific virus exposure were heterogeneous across populations, antibody responses targeted strongly conserved "public epitopes" for each virus, suggesting that they may elicit highly similar antibodies. VirScan is a powerful approach for studying interactions between the virome and the immune system. Copyright © 2015, American Association for the Advancement of Science.
Article
A randomized double-blind placebo-controlled trial was undertaken to determine whether cyclosporin enhances remission of insulin-dependent diabetes mellitus (IDDM) through the 1st yr after diagnosis. Dosage with insulin was minimized with target control of blood glucose levels less than or equal to 7.8 mM (140 mg/dl) before meals. Metabolic control was evaluated by serial determinations of glycosylated hemoglobin levels, and endogenous secretion of insulin was evaluated by determination of the levels of glucagon-stimulated insulin-connecting peptide (CP) in the plasma at 3-mo intervals. A compound definition of remission required a glucagon-stimulated CP level in plasma greater than or equal to 0.6 nM or a non-insulin-receiving state (NIR) in which target control of glycemia was maintained without administration of insulin. A clinical definition of remission required only the NIR state as defined. One hundred eighty-eight patients aged 10-35 yr entered the study within 6 wk of initiation of insulin therapy and within 14 wk of onset of symptoms and were studied for 1 yr. There were no significant differences in metabolic control between the two treatment groups during the study. The anticipated adverse effects of cyclosporin were not more frequent or severe than in other experience with the drug, but histological changes attributable to cyclosporin were present in some kidney biopsies obtained from selected patients after 1 yr. At 1 yr, by the compound definition, 33% of the cyclosporin-group and 21% of the placebo-group patients were in remission, when the corresponding rates for NIR remissions were 24 and 10%.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
Objective: Type 1 diabetes (T1D) arises from the autoimmune destruction of the β-cells of the pancreas, resulting in dependence on exogenously administered insulin for survival. Key biomarkers of the autoimmune process in T1D are the occurrence of autoantibodies directed against β-cells and other antigens. The Type 1 Diabetes Genetics Consortium (T1DGC) assembled collections to 1) discover genes that modify the risk of T1D, 2) conduct phenotyping related to risk, and 3) make available biologic and genetic resources for research. The goal of the T1DGC Autoantibody Workshop was to use T1DGC phenotypic, genotypic, and autoantibody data on affected sibling pair (ASP) families to discover genes accounting for variation in presence of autoantibodies. Research design and methods: The T1DGC provided the working groups with autoantibody and genetic data on 9,976 subjects from 2,321 ASP families. Data were distributed to numerous working groups for analyses of specific autoantibody subsets and targets. Results: Seven groups analyzed the joint autoantibody and genetic data within the ASP families. Six reports are provided in this collection, ranging from candidate gene analyses of selected autoantibodies to evaluation of regions of genetic variants associated with autoimmunity on the collection of autoantibodies. Conclusions: Although selected variants in the available genes remain important genetic predictors for prevalence of T1D, other genes and nongenetic factors are expected to contribute to the initiation of islet autoimmunity, the first step in the pathogenesis of T1D.
Article
Aims To examine incidence and trends of Type 1 diabetes worldwide for the period 1990–1999. Methods The incidence of Type 1 diabetes (per 100 000/year) was analysed in children aged ≤ 14 years from 114 populations in 112 centres in 57 countries. Trends in the incidence of Type 1 diabetes were analysed by fitting Poisson regression models to the dataset. Results A total of 43 013 cases were diagnosed in the study populations of 84 million children. The age‐adjusted incidence of Type 1 diabetes among 112 centres (114 populations) varied from 0.1 per 100 000/year in China and Venezuela to 40.9 per 100 000/year in Finland. The average annual increase in incidence calculated from 103 centres was 2.8% (95% CI 2.4–3.2%). During the years 1990–1994, this increase was 2.4% (95% CI 1.3–3.4%) and during the second study period of 1995–1999 it was slightly higher at 3.4% (95% CI 2.7–4.3%). The trends estimated for continents showed statistically significant increases all over the world (4.0% in Asia, 3.2% in Europe and 5.3% in North America), except in Central America and the West Indies where the trend was a decrease of 3.6%. Only among the European populations did the trend in incidence diminish with age. Conclusions The rising incidence of Type 1 diabetes globally suggests the need for continuous monitoring of incidence by using standardized methods in order to plan or assess prevention strategies.