You are on page 1of 14

Cell Stem Cell

Perspective

Stem Cell Therapies for Treating Diabetes:


Progress and Remaining Challenges
Julie B. Sneddon,1,2,3 Qizhi Tang,4 Peter Stock,4 Jeffrey A. Bluestone,1,5 Shuvo Roy,6,7 Tejal Desai,6,7
and Matthias Hebrok1,3,*
1Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
2Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
3Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
4Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
5Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA 94143, USA
6UCSF-UC Berkeley Joint Ph.D. Program in Bioengineering, University of California, San Francisco, San Francisco, CA 94143, USA
7Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA

*Correspondence: mhebrok@diabetes.ucsf.edu
https://doi.org/10.1016/j.stem.2018.05.016

Restoration of insulin independence and normoglycemia has been the overarching goal in diabetes research
and therapy. While whole-organ and islet transplantation have become gold-standard procedures in
achieving glucose control in diabetic patients, the profound lack of suitable donor tissues severely hampers
the broad application of these therapies. Here, we describe current efforts aimed at generating a sustainable
source of functional human stem cell-derived insulin-producing islet cells for cell transplantation and present
state-of-the-art efforts to protect such cells via immune modulation and encapsulation strategies.

Over the last century, improvements in the synthesis and delivery Clinically Viable Transplantation Strategies for Treating
of recombinant insulin have substantially decreased the Diabetes
morbidity and mortality associated with diabetes mellitus. Whole Pancreas Organ Transplantation
Despite these advances, more than 400 million people across Advances in surgical techniques and refinement of immuno-
the world who are affected by diabetes mellitus continue to suf- suppression have dramatically improved the success of
fer from devastating secondary complications, including dia- pancreas transplantation performed for diabetes mellitus. The
betic nephropathy, retinopathy, and neuropathy. Intensified traditional indication for solid organ pancreas transplant has
metabolic control has reduced or prevented the development been in recipients with type I diabetes (T1D) and end-stage
and progression of secondary complications in two landmark tri- renal disease, and the procedure is most commonly performed
als in patients with type I (Nathan et al., 1993) and type 2 (Holman simultaneously with a kidney transplant (SPK). One-year
et al., 2008; UK Prospective Diabetes Study (UKPDS) Group, allograft success, as defined by insulin independence, is
1998a, 1998b) diabetes mellitus. Unfortunately, the tighter con- approximately 90% at most centers performing this operation.
trol associated with intensified regimens has been limited by Long-term results continue to improve with the evolution of
the inherent risks of hypoglycemia. Excellent metabolic control better immunosuppressive regimens, with five- and ten-year
without the need for exogenous insulin can be achieved with pancreas graft survival rates at 73% and 56%, respectively
beta cell replacement, either through solid organ pancreas trans- (Gruessner and Gruessner, 2016). Marked improvements in
plantation or pancreatic islet transplantation. Both strategies for successful transplantation have increased the indications for
beta cell replacement stabilize or minimize progression of the pancreas transplantation to include pre-uremic T1D recipients
secondary complications associated with diabetes mellitus, with life-threatening diabetes secondary to hypoglycemic
providing stable long-term allograft function as demonstrated unawareness. Type 2 diabetic (T2D) recipients now represent
by insulin independence and normalization of glycated hemoglo- 9% of all SPK recipients, and their early allograft success
bin (HbA1C) levels. is comparable to the T1D SPK recipients (Kandaswamy
Despite the increasing success of both strategies for beta cell et al., 2018).
replacement, broader application of islet and pancreas trans- Pancreas transplantation requires a strong cardiovascular
plantation is severely limited by the number of available donor system to tolerate both the initial procedure as well as the poten-
pancreases and the need for lifelong immunosuppression; as a tial complications associated with transplantation of a fragile or-
result, only a small fraction of people with diabetes mellitus gan containing digestive enzymes. Pancreas transplants involve
can currently benefit from these therapies. Creating an unlimited the intraperitoneal placement of the pancreas in a heterotopic
source of insulin-producing cells from stem cells will permit location. The reconstructed donor pancreas most often receives
widespread application of beta cell replacement to achieve insu- its arterial blood supply from recipient iliac vessels, portal (supe-
lin independence. As this source of beta cells moves closer to rior mesenteric vein) or systemic (iliac vein) venous drainage, and
clinical translation, it is important to review the current state of enteric exocrine drainage of the donor pancreas through an
the art in beta cell replacement, with a focus on successful anastomosis between the donor duodenal segment and the
encapsulation and immune modulation strategies that can be recipient ileum. The technical success of pancreatic transplants
applied to stem cell-derived cells. is around 90%–95%. Early loss of the pancreas allograft is

810 Cell Stem Cell 22, June 1, 2018 ª 2018 Published by Elsevier Inc.
Cell Stem Cell

Perspective

related to thrombosis of the pancreas allograft or leaks of tion regimens that were found to be successful in solid organ
pancreatic enzymes resulting in infection, necessitating removal pancreas transplants (Barton et al., 2012). Novel immunosup-
of the allograft. A technically successful allograft results in pressive regimens involving maintenance based on blocking im-
almost immediate insulin independence. Furthermore, pre- mune costimulation (CTLA-4Ig, Belatacept) or leukocyte adhe-
transplant insulin requirements and body mass index (BMI) do sion (anti-LFA1, Raptiva) have further increased the three-year
not impact the ability to achieve insulin independence, and the insulin independence rates to 70% (Posselt et al., 2010). This
vast majority of pancreas transplant recipients with technically type of immunosuppression is attractive in that these agents
successful allografts achieve insulin independence (Kandasw- specifically target immune cells without toxic effects on beta
amy et al., 2018). Stable function of the pancreas allograft pre- cells or kidneys. Importantly, insulin independence was
vents recurrence of diabetic nephropathy in the simultaneously achieved in several cases with a single infusion of donor islets.
transplanted kidney and can prevent progression of retinopathy A cost comparison between solid organ pancreas transplanta-
and neuropathy (Fioretto et al., 1998). For that reason, solid or- tion and pancreatic islet transplantation has demonstrated com-
gan pancreas transplantation has become the gold standard parable costs to achieve insulin independence (Moassesfar
for beta cell replacement for patients healthy enough to sustain et al., 2016). Unfortunately, the very limited number of pancreatic
this taxing procedure. Still, the widespread application of the islets available for transplantation severely restricts the wide-
technique is limited by the rigors of the procedure as well as spread use of this technology.
the scarce availability of suitable donor pancreases.
Pancreatic Islet Transplantation Stem Cell-Derived Therapies for the Treatment of
Transplantation of isolated islets provides a gentler alternative to Diabetes
whole-organ transplantation. A brief history of the events leading Recent advances in the differentiation of human stem cells to-
to this success is important for understanding future strategies wards pancreatic islet cells now suggest clear and tangible alter-
for beta cell replacement using stem cell-derived beta cells. natives to the more conventional treatment options for T1D and
Although pancreatic islet transplantation had been conducted T2D described above. Remarkable progress has been made
in animal models of diabetes since the early 1980s, successful with regard to the generation of functional beta cells from human
clinical trials in T1D recipients were elusive until the Edmonton stem cell populations over the last decade. The underlying strat-
protocol was published in 2000 (Shapiro et al., 2000). To estab- egy is to closely recapitulate the path that pluripotent stem cells
lish insulin independence in seven consecutive patients, islets take during embryogenesis, from the formation of definitive
were isolated from deceased donor pancreases and infused endoderm, to pancreatic endoderm, to endocrine progenitors,
into the portal vein of the liver. Two to three infusions of islets and finally to pancreatic islet cells (Figure 1) (D’Amour et al.,
from different donors were required to obtain insulin indepen- 2005, 2006; Guo et al., 2013; Kroon et al., 2008; Rezania et al.,
dence, which was initially achieved in all seven patients. The suc- 2011, 2012). More recently, efforts have been placed on gener-
cess was attributed to a regimen that minimized exposure to ating single hormone-positive beta cells capable of glucose-
immunosuppressive agents toxic to beta cells, such as steroids stimulated insulin secretion (GSIS) (Millman et al., 2016; Pagliuca
or lymphodepleting induction therapy. Instead, the immunosup- et al., 2014; Rezania et al., 2014; Russ et al., 2015; Zhu et al.,
pressive regimens consisted of low-dose calcineurin inhibitors 2016). Efforts in several laboratories are focused on generating
(CNIs) to minimize nephron and beta cell toxicity. Despite their fully functional beta and islet cells that could soon replace human
initial insulin independence, all seven patients lost islet function islets as a source for insulin-producing cells. Indeed, clinical tri-
within 2 years. als using stem cell-derived pancreas progenitors capable of
This initial experience revealed some of the problems associ- developing into functional beta and islet cells are currently un-
ated with intraportal infusion of islets, as well as the necessity for derway. Here, we review the current state of generating human
more potent immunosuppression. The requirement for two or embryonic stem cell (hESC)-derived pancreatic islet cell types.
three donor infusions to create an early state of insulin indepen- Optimizing the Stem Cell Niche to Generate Islet-like
dence was likely related to poor engraftment of the donor islets. Structures
The instant blood-mediated inflammatory reaction (IBMIR) and Despite the rapid progress in generating insulin-producing cells
ischemia contributed to islet loss shortly after infusion into the from hESC populations, the formation of fully functional cells
portal system (Eich et al., 2007). The marginal islet mass that sur- perfectly replicating all aspects of endogenous beta cells has re-
vives and engrafts following intraportal infusion is consistent with mained elusive. Considering that beta cells in vivo do not exist in
the finding that insulin independence has been more common in isolation, but rather in the context of intricate, 3D structures of
recipients with low BMIs and lower insulin requirements (Barton the islets of Langerhans, generation of a ‘‘full complement’’ of
et al., 2012; Shapiro et al., 2017). islet cells should be considered beneficial (Figure 1). Indeed,
The problems with marginal mass following islet transplanta- the maintenance of beta cell function has been reported to be
tion are further exacerbated by the alloimmune and recurrent highly dependent on the complex islet cytoarchitecture, with iso-
autoimmune responses against the graft. In fact, significant im- lated beta cells behaving differently from those within intact islets
provements in long-term insulin independence have been (Cabrera et al., 2006; Halban et al., 1982; Ravier et al., 2005; Woj-
achieved by using more potent immunosuppression. Recent in- tusciszyn et al., 2008). In single, isolated beta cells, for instance,
ternational registry data have reported three-year insulin inde- glucose-stimulated insulin secretion is compromised compared
pendence rates following intraportal infusion of islets at nearly to intact islets, due to both elevated basal insulin secretion and
50% (Shapiro et al., 2017). Improvements in long-term insulin in- reduced maximal insulin secretion in response to glucose (Ben-
dependence required the same potent lymphodepleting induc- ninger et al., 2011; Halban et al., 1982). Along with other

Cell Stem Cell 22, June 1, 2018 811


Cell Stem Cell

Perspective

Figure 1. Strategies for Generating Human Pluripotent Stem Cell (hPSC)-Derived Pancreatic Islets for Transplantation
(A) The pancreas is composed of both an exocrine and an endocrine compartment. The latter is composed of small spherical mini-organs called the islets of
Langerhans (inset). Each islet contains insulin-producing beta cells, along with other hormone-producing endocrine cells (alpha, delta, epsilon, and PP cells).
A subset of beta cells are so-called ‘‘hub’’ cells, which orchestrate the coordinated release of insulin across the islet in response to glucose. The endocrine cells do
not exist in isolation, but rather in the context of various types of niche cells, including stromal, neural, endothelial, and perivascular cells.
(B) The generation of beta-like cells from human pluripotent stem cells (hPSCs) involves the promotion of pluripotent cells through a series of intermediate
progenitor stages. At the completion of directed differentiation, the clusters of beta-like cells can be dissociated and reaggregated into islet-like clusters of
defined size and composition. The reaggregation with other endocrine cell types (themselves potentially derived from hPSCs) may lead to improved glucose
homeostatic function, potentially through the specialization of some hPSC-derived beta-like cells into hub cells. Lastly, endocrine cells can be reaggregated
along with niche cells to more closely recapitulate interactions with the local microenvironment that are critical for beta cell function.

mechanisms for coordinating cell-cell communication, gap junc- sition and structure between mouse and human islets have been
tions such as Connexin-36 play an important role in maintaining linked to functional differences. In murine islets, for instance,
beta cell coupling and islet synchronization of insulin oscillations beta-beta intercellular contacts are believed to coordinate homo-
(Benninger et al., 2011; Ravier et al., 2005). As we consider the geneous and synchronous release of insulin, whereas human
methods for generating the most optimally functioning hESC- islets, which lack as many homologous beta cell contacts, display
derived unit for diabetic rescue, it will also be important to asynchronous insulin release following glucose stimulation (Woj-
consider methods for recapitulating endogenous islet structure, tusciszyn et al., 2008). Exciting work is emerging from single-cell
cell-cell contacts, and communication with the islet microenvi- sorting and sequencing that supports the notion of distinct beta
ronment. cell subtypes in human islets (Dorrell et al., 2016).
In rodent models, pancreatic islets typically consist of a beta Another determinant of beta cell function is communication
cell core surrounded by a mantle of alpha, delta, and pancreatic with cells in the islet niche, which is composed of multiple non-
polypeptide (PP) cells, which secrete other hormones important endocrine cell types, including endothelial, perivascular,
for proper glucose homeostasis (Bosco et al., 2010; Schaeffer neuronal, and mesenchymal cells (Hayden et al., 2008; Lammert
et al., 2011). This architectural arrangement favors homologous et al., 2003a; Reinert et al., 2014). In particular, pancreatic
cell-cell contacts and is important for normal islet function; rodent islets are highly vascularized; each beta cell is believed to
models of diabetes exhibit abnormal islet architecture, with alpha, make one or more contacts with an endothelial cell, and together
delta, and PP cells intermingling with beta cells within the islet they form specialized ultrastructural features at their interface
core (Wojtusciszyn et al., 2008). In contrast to rodent islets, hu- (Bonner-Weir and Orci, 1982; Henderson and Moss, 1985).
man islets favor heterologous cell-cell contacts, with more evenly During pancreatic development, endothelial cells are initially re-
distributed endocrine types found across an islet and less cruited into the burgeoning islet by vascular endothelial growth
compartmentalization into peripheral and central compartments factor A (VEGF-A), which is secreted by endocrine cells
(Bosco et al., 2010). The relative proportions of the endocrine sub- (Lammert et al., 2003b). It is the endothelial cells that then syn-
types can also differ between murine and human islets, with the thesize and secrete the components of the islet basement mem-
former dominated by beta cells (80%) and the latter more evenly brane, which is believed to serve as a critical modulator of beta
composed of alpha (40%) and beta (50%) cells (Pan and Wright, cell growth, survival, and function (Lammert et al., 2003a; Niko-
2011), although different proportions have also been noted (Bon- lova et al., 2006; Oberg-Welsh, 2001; Stendahl et al., 2009;
ner-Weir et al., 2015; Kilimnik et al., 2012). Disparities in compo- Wang and Rosenberg, 1999). Work by the Otonkoski group

812 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective

demonstrated the presence of two basement membranes et al., 2016). Furthermore, it is more challenging to create states
around the endothelium and beta cells in human islets (Oton- of insulin independence in patients with higher BMIs and higher
koski et al., 2008; Virtanen et al., 2008). Given the specialized insulin requirements (Balamurugan et al., 2014). Portal infusion
relationship between beta cells and supporting cells in their nat- limits the amount of tissue that can be infused secondary to
ural environment, it stands to reason that recapitulation of some the development of portal hypertension. In addition, isolated is-
elements of the endogenous beta cell niche may lead to lets engraft poorly secondary to IBMIR and ischemia injuries
improved survival and function of hESC-derived beta cells (Eich et al., 2007). Of equal significance, islets infused into the
in vitro. portal vein are not retrievable, and this remains a concern for
The vascular islet niche consists not only of endothelial cells, the clinical translation of any trials using hESC-derived beta cells
but also of mural cells called pericytes. Pericytes wrap around until it has been unequivocally proven that such cells are equiv-
endothelial cell tubes to provide structural support and generate alent to fully mature human islet cells and lack any possibility of
functional, mature blood vessels (Dı́az-Flores et al., 2009) and neoplastic transformation. However, while the risk of teratoma
modulate endothelial cell proliferation, survival, and function formation from undifferentiated stem cells cannot be completely
(Jain, 2003). The role of pericytes in normal islet function is not ignored at present, further optimizing the generation of fully
completely understood, but one study using genetic ablation differentiated islet and beta cells from stem cells should alleviate
suggested that their loss in the adult murine islet results in im- this concern.
pairments in insulin expression and glucose-clearing (Sasson Considering the information gained from solid organ and islet
et al., 2016). transplantation, it is clear that a more optimal site for islet tissue
Given the importance of islet cytoarchitecture for endocrine is necessary. Multiple investigators are optimizing subcutaneous
function, culture systems can be designed to recapitulate ele- or intramuscular sites for islet transplantation to enhance
ments of the intricate 3D structure of the developing islet. Various engraftment and permit removal of the graft in the event of tissue
methods have been used to generate engineered pancreatic dysfunction or transformation (Bertuzzi et al., 2018; Gamble
islets, or so-called ‘‘pseudo-islets,’’ in vitro (Kojima, 2014). Mul- et al., 2018). Similarly, exploring optimal sites that permit retrieval
tiple studies have reported improved function of reaggregated of hESC-derived beta cells will be important for clinical transla-
pseudo-islets, either in vitro or in vivo after transplantation. There tion. Successful pancreas transplantation in T2D patients sug-
is also evidence that beta cells reaggregated with endothelial gests that given an unlimited source of beta cells, cell replace-
progenitor cells demonstrate improved GSIS compared to ment may provide a benefit for the vast majority of people
pseudo-islets composed only of beta cells (Penko et al., 2011). suffering from T2D (Kandaswamy et al., 2018). Of note, trans-
In summary, more work is needed to determine the cellular plantation of any allogeneic cell source requires mitigation of
composition and architectural arrangement of human islets the recipient immune system.
that is optimal for function. Challenges will likely include deci-
sions regarding how to source the desired human niche cells, Immune Modulation in Beta Cell Replacement Therapy
particularly given that there is a lack of truly specific markers The recent success in the use of beta cell replacement to treat
for some of these cell types, and that the supply may be limiting. T1D, combined with advances in the generation of hESC- or
One option is the generation of the niche cells themselves from iPSC-derived beta cells, provides a roadmap for effective treat-
hESCs or induced pluripotent stem cells (iPSCs). Another chal- ment of the disease. However, both the alloimmune and autoim-
lenge is that current techniques for creating pseudo-islets rely mune systems remain major barriers to the widespread adoption
on the property of self-organization among endocrine and non- of cell replacement therapies in T1D and, potentially, T2D. Cur-
endocrine cells; 3D printing of islet tissues may someday be rent immunosuppressive drugs are effective but require lifelong
possible and could instead allow the enforcement of a desired treatment, and in many cases lead to side effects and toxicities
architecture. Future work aimed at incorporating flow in micro- that make the adoption of this treatment strategy limited to
fluidic devices, as has been done in various other systems with only the most severe cases of disease. Here, we focus on new
‘‘organ-on-a-chip’’ approaches, may allow even more sophisti- efforts to understand and control immunity, with the goal of
cated optimization of function while reducing negative effects creating a combinatorial therapy that takes advantage of drugs,
of waste product accumulation. Re-engineering additional com- bioengineering, and gene editing to bring beta cell replacement
ponents of a pancreatic islet with the optimal composition of to the larger diabetes community.
endocrine and non-endocrine niche cells, including vascular Control of Alloimmune Responses against Beta Cells
niche cells and extracellular matrix (ECM) components, may In T1D patients, transplanted allogeneic beta cells face alloim-
lead to an optimally functional beta cell population for transplan- mune-mediated rejection that can eliminate the graft within
tation into patients. days if not suppressed. T cells are the principal drivers of
Applying Lessons Learned from Whole-Organ Pancreas alloimmunity; their activation results from T cell receptor (TCR)
and Islet Transplantation to Stem Cell-Derived recognition of antigenic peptides presented by the major histo-
Beta Cells compatibility complexes (MHCs). The highly polymorphic pro-
There is growing appreciation for the fact that the transplant site teins of the MHC, referred to as human leukocyte antigens
itself influences the functionality and survival of islets. Unlike the (HLAs) in humans, are the major targets of alloimmune attacks.
insulin independence that always occurs following a technically Minor histocompatibility antigens, such as male antigens in
successful pancreas transplant, the marginal mass of engrafted female recipients or mitochondrial antigens, can also trigger
islets associated with infusion through the portal vein often re- rejection responses, although at a slower pace. MHC class I is
quires multiple infusions from different deceased donors (Hering expressed on all cell types, including beta cells, and beta cells

Cell Stem Cell 22, June 1, 2018 813


Cell Stem Cell

Perspective

Table 1. Immune Modulators for Alloimmune and Autoimmune Responses


Drug Effect Use in Transplant Use in T1D Patients
ATG deletes T cells, NK cells induction IS, treatment of rejection does not prevent disease progressiona
(NCT00515099) (Gitelman et al., 2013)
Anti-CD25 deletes activated T cells induction IS does not prevent disease progressiona
(NCT00198146)
Calcineurin inhibitors blocks TCR signaling maintenance IS remission while on IS (Bougnères et al.,
1990; Feutren et al., 1986; Stiller
et al., 1984)
Mycophenolate blocks T and B cell proliferation maintenance IS does not prevent disease progressiona
(NCT00100178)
Steroid anti-inflammatory induction IS; may be used as toxic to beta cells
maintenance, treatment of rejection
Rapamycin blocks mTOR signaling, cell cycle, maintenance IS enhances Treg functiona (NCT02803892,
and metabolism NCT00525889)
CTLA-4Ig blocks T cell costimulation maintenance IS transient halt of islet function declinea
Anti-CD3 partial agonist of TCR; depletes induction IS, treatment of rejection transient halt of islet function declinea
effector T cells (NCT00378508, NCT00627146,
NCT00678886)
LFA3Ig selectively targets memory and no additional benefit when added transient halt of islet function declinea
effector T cells over Tregs to conventional IS (NCT00965458)
Anti-CD20 depletes B cells failed to treat antibody-mediated transient halt of islet function declinea
rejection (Becker et al., 2004; (NCT00279305)
Vo et al., 2008)
BMT causes immunological reset can achieve tolerance using donor transient halt of islet function declinea
BMT in some patients (NCT00821899) (Esmatjes et al., 2010;
Gu et al., 2014)
Treg cell therapy promotes Treg-mediated safe in early phase clinical trials safe in early phase clinical trial
suppression
This table summarizes immune modulatory therapies used in controlling transplant rejection. Many of these drugs have also been explored to induce
diabetes remission in patients with recent onset of type 1 diabetes. ATG, anti-thymocyte globulin; IS, immunosuppressants; BMT, bone marrow trans-
plant.aData from clinical trials with ClinicalTrials.gov identifier number provided in parentheses.

overexpress MHC I in inflamed conditions (Hamilton-Williams stronger induction therapies that deplete T cells, combined
et al., 2003). MHC class II is expressed on antigen presenting with blockade of inflammatory cytokines TNFa and IL-1, lead
cells (APCs) such as B cells, dendritic cells, and macrophages to better long-term islet graft function (Barton et al., 2012).
and can be induced on non-APCs, including beta cells (Foulis Although immunosuppression protects grafts from rejection, it
and Farquharson, 1986; Pujol-Borrell et al., 1987). After trans- globally compromises the patients’ immune systems, leaving
plantation, MHCs released by donor cells are taken up by recip- them vulnerable to infections and malignancies. An ideal
ient APCs, broken down into antigenic fragments, and presented strategy would be to train the immune system to accept the
by recipient MHC on the surface of recipient APC. This normal transplanted graft as self and avoid chronic global immunosup-
process of T cell activation is referred to as the ‘‘indirect’’ pression. Immune tolerance to transplanted tissue has been
pathway in the context of transplantation. This is in contrast to achieved in many preclinical animal models, and potential toler-
the ‘‘direct’’ pathway that is unique to transplantation immu- ance-promoting therapies are currently under development for
nology. In the direct pathway, TCRs on the recipient’s T cells humans.
bind to allogeneic MHC expressed on donor cells, leading to Most commonly used immunosuppressive drugs inhibit both
T cell activation and direct attack of the graft cells. Both direct effector cells and tolerance-promoting immune regulatory
and indirect T cell pathways contribute to graft rejection and mechanisms (Table 1). However, different drugs have varying
often act cooperatively to carry out the attacks (Brennan et al., selectivity in inhibiting anti-graft effector responses versus toler-
2009; Jiang et al., 2004; Makhlouf et al., 2003). ance-promoting regulatory T cells (Tregs) (Furukawa et al.,
The current approach to control transplant rejection is to glob- 2016). For example, the use of CNIs decreases the percentages
ally suppress T cell activation by inhibiting TCR signaling with of Tregs, whereas rapamycin use is associated with an increase
CNIs, prevent T cell proliferation with mycophenolate, and in percentages of Tregs. CTLA-4Ig (Belatacept) inhibits effector
dampen inflammation with steroids. In addition to this triple- T cell activation and clonal expansion but can also impair Treg
drug regimen, transplant patients often receive ‘‘induction’’ ther- homeostasis and function at high doses (Bour-Jordan et al.,
apies, defined as short-term treatments in the peri-transplant 2004; Salomon and Bluestone, 2001). Thus, by selecting immu-
period. Clinical islet transplant experiences have shown that nosuppressive drugs and their dosing, it may be possible to

814 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective

preferentially inhibit effector cells and to leverage the immune pressed recipients of pancreas and kidney transplants (Burke
system’s self-control mechanisms to reduce the burden of et al., 2015). T cell and B cell targeting therapies, such as anti-
immunosuppression. Achieving immune tolerance requires the CD3, Abatacept (a CTLA-4Ig molecule similar to Belatacept
immune system to be aware of the antigen and to actively avoid described above), and anti-CD20, lead to a transient halt in the
activation toward that antigen. Tolerance to self-antigens is decline of beta cell function in some newly diagnosed T1D pa-
achieved through a combination of deletion, cell-intrinsic check- tients (Hagopian et al., 2013; Herold et al., 2002, 2005; Keymeu-
points, and suppression by regulatory mechanisms such as len et al., 2005, 2010; Orban et al., 2011; Pescovitz et al., 2009;
Tregs, IL-10, and transforming growth factor ß (TGFß) signaling. Sherry et al., 2011). Anti-thymocyte globulin, a drug commonly
By blocking immune activation, currently available immunosup- used to induce T cell depletion in organ transplantation, does
pressive drugs also inhibit these tolerance-inducing mecha- not protect against beta cell loss when given at a high dose (Gi-
nisms. Thus, attaining tolerance is very inefficient using currently telman et al., 2013, 2016) and had short-term benefit when given
available immunosuppressive drugs. at a low dose in combination with G-CSF (Haller et al., 2015). In
In the past 2 decades, there have been concerted research ef- contrast, another T cell targeting agent, LFA-3Ig (Alefacept), sta-
forts to develop strategies for inducing transplantation tolerance. bilizes beta cell mass in patients with new-onset disease. A com-
One such strategy is to construct donor-recipient hematopoietic monality among anti-CD3, Abatacept, and Alefacept is that they
chimeras. The presence of donor immune cells will educate the are relatively selective for effector T cells, while sparing and even
recipient immune cells to accept cells of donor origin as self, boosting Tregs (Bluestone et al., 2008; Long et al., 2017; Rigby
thus preventing graft rejection (Kawai and Sachs, 2013). One et al., 2015; Tintle et al., 2011). Thus, their therapeutic efficacy
challenge with this approach is the instability of the mixed may be due to their ability to simultaneously thwart effector
chimeric state, resulting in either the host or the donor immune T cells while promoting regulation. These hints of efficacy in con-
system taking over. When donor cells dominate, the risk of trolling autoimmune attacks on islets provide guidance for
graft-versus-host disease becomes high; when recipient cells designing more effective combination therapies to induce long-
prevail, tolerance towards the donor is often lost. An alternative term tolerance.
strategy is to promote Tregs using adoptive cell therapy (Tang In addition to the broad immunomodulatory strategies
and Bluestone, 2013). A challenge with adoptive Treg therapy described above, there are also ongoing efforts in developing
is the high frequency of donor-reactive effector T cells that can islet antigen-specific therapies. Preclinical data show that it is
overwhelm Treg-mediated suppression. Reduction of donor- possible to target effector cells specific for dominant islet anti-
reactive T cells is needed for Tregs to control the rejection gens using tolerogenic peptide vaccine or nanoparticles. The ef-
responses (Lee et al., 2014; Wells et al., 1999). Emerging preclin- ficacy of such strategies in patients remains to be demonstrated
ical evidence suggests that Treg cell therapy may stabilize he- in clinical trials (Ludvigsson et al., 2012; Wherrett et al., 2011).
matopoietic chimeras; thus, the two therapies may synergize Although many of these studies have shown some effects in
to achieve more reliable and durable tolerance. small patient subsets, and new antigen therapy approaches
Control of Recurrent Autoimmune Attack of are underway in several programs, it will be essential to deter-
Transplanted Beta Cells mine the precise antigen, timing, and method of administration
In T1D patients, islet autoantigen-specific T cells can also attack to maximize and effectively test efficacy. A challenge for anti-
and destroy transplanted beta cells and thus contribute to the gen-specific immunotherapy is the number of islet antigens
rejection of transplanted beta cells (Burke et al., 2015; Stegall that are targeted by the immune system; disarming T cells spe-
et al., 1996). MHC class I expression on islet cells is necessary cific for one or a subset of the islet antigens alone will not likely be
for islet destruction by recurrent autoimmunity (Young et al., sufficient to change the disease course. In addition, the reper-
2004), implicating a role of autoreactive CD8 T cells. Additionally, toires of islet antigen specificities are likely to be distinct in
islet antigens expressed by the transplanted beta cells can be different patients at different disease stages, making it difficult
presented by recipient APCs and activate autoreactive CD4 to know which specificities to target. Approaches that induce
T cells to destroy the graft (Kupfer et al., 2005). These autoreac- dominant antigen-specific regulation are likely to be more effec-
tive T cells can directly kill beta cells, make cytokines toxic to tive and easier to apply to diverse patient populations.
beta cells, and activate innate immune cells such as NK cells Treg cell therapy offers many attractive therapeutic attributes
and macrophages to kill beta cells. with regards to dominant antigen-specific regulation of effector
Strategies for the control of recurrent autoimmune rejection of T cell responses. For example, Tregs control unwanted immune
transplanted beta cells can be adopted from those developed to responses via ‘‘linked suppression’’ and ‘‘infectious tolerance.’’
control autoimmune responses in T1D patients. Extensive In linked suppression, Tregs activated by one antigen can sup-
research efforts have been devoted to the restoration of immune press responses to other antigens presented in the same tissue
self-tolerance in T1D. Interestingly, many of the commonly used microenvironment. In infectious tolerance, Tregs create a tolero-
immunosuppressive drugs for preventing transplant rejection genic tissue microenvironment so that other T cells activated in
are not effective at inducing diabetes remission (Table 1). the vicinity adopt a regulatory instead of an effector cell fate.
Furthermore, the majority of the commonly used immunosup- This allows tolerance to propagate over time beyond the persis-
pressive drugs for preventing alloimmune-mediated transplant tence of the initiating Tregs. These properties ensure that Tregs
rejection can induce short-term diabetes remission with no of limited specificity can exert specific, localized, dominant,
long-term efficacy (Table 1). The ineffectiveness of conventional and durable control of other immune cells. Indeed, a single infu-
drugs at suppressing autoimmune responses is also evident sion of islet antigen-specific Tregs can induce lifelong remission
from recurrent autoimmune attacks against islets in immunosup- of diabetes in autoimmune non-obese diabetic mice (Tang et al.,

Cell Stem Cell 22, June 1, 2018 815


Cell Stem Cell

Perspective

2004). Early phase trials of Treg cell therapy in T1D patients are be needed to silence alloimmune responses against beta cells.
currently underway, demonstrating the feasibility and safety of An efficient way to prevent expression of HLA II genes is to
this approach in human patients (Bluestone et al., 2015; eliminate the master transactivator of HLA class II genes, CIITA
Marek-Trzonkowska et al., 2012). (Chang et al., 1996).
Genetic Engineering of Stem Cell-Derived Beta Cells to Ablating HLA expression on beta cells not only decreases
Prevent Immune Detection/Rejection their alloimmunogenicity, but also reduces autoimmune recog-
Stem cell-derived beta cells are now very close to their endoge- nition of the cells. However, islet autoantigens from the grafted
nous counterparts with regard to functional properties, indi- cells can still be presented by host APCs, leading to activation
cating that the generation of mature beta cells from stem cells of autoreactive T cells, and can injure beta cells by producing
is within grasp. However, issues concerning the prevention of cytotoxic cytokines and activating innate immune cells. More-
immune destruction of such cells by the recipient immune sys- over, beta cells react to inflammation by producing inflamma-
tem remain among the unsolved problems. Most notably, the im- tory cytokines and chemokines themselves, leading to the
mune system of T1D patients that already targets native beta amplification of the immune response against the graft (Eizirik
cells is likely primed to target transplanted stem cell-derived et al., 2009). Much of the beta cell toxicity induced by cytokines
beta cells. Moreover, allogeneic responses to transplanted and the pro-inflammatory response of the beta cells is mediated
hESC-derived beta cells pose a significant obstacle for the im- by the transcription factor NFkB and the signal transducer
plementation of this new technology for both T1D and T2D pa- STAT1 (Eizirik et al., 2009). Controlling NFkB activation by over-
tients. expression of A20 and suppressing STAT1 activation by over-
Current therapeutic strategies described above designed to expression of SOCS1 can be effective in quenching the
promote immune tolerance to transplant antigens and islet cascade of inflammatory responses in beta cells (Chong
self-antigens can be applied to beta cell replacement therapy et al., 2001, 2002; Grey et al., 1999; Solomon et al., 2011).
in diabetic patients. In addition, stem cell technology offers Furthermore, employing other immune modulatory approaches,
further opportunities to engineer beta cells to avoid immune at- such as enhancing immune checkpoints via forced expression
tacks. The advent of novel gene editing tools, most notably the of PDL1, may also be applied to protect beta cells against
CRISPR/Cas9 system (Lin et al., 2014), provides unique oppor- autoreactive T cells.
tunities to disassemble MHC components in hESCs, with the Additional considerations should be given to NK cells that may
goal of preventing presentation of alloantigens by the stem be activated against HLA-ablated stem cell-derived beta cells.
cell-derived cells. Such a strategy would also prevent presenta- NK cell cytotoxicity is regulated by an array of activating and
tion of autoantigens by the stem cell-derived beta cells and inhibitory receptors (Lanier, 2008). NK-mediated killing requires
recognition by memory autoimmune cells in T1D patients. As the engagement of an activating receptor and an absence of
described above, T cell recognition of polymorphic MHC mole- signaling by the inhibitory receptors. Islet beta cells are known
cules is the principal driver of alloimmunity. In T1D patients, to express ligands for NK activating receptors such as Rae1 in
recurrent autoimmune attacks of transplanted beta cells also mice and MICA/B in humans (Hankey et al., 2002; Ogasawara
depend on MHC class I expression on islet cells (Young et al., et al., 2004). Class I HLAs are major ligands for NK inhibitory re-
2004). Thus, eliminating MHC class I and class II expression ceptors. Thus, complete ablation of class I HLA expression on
can potentially reduce alloimmune and autoimmune-mediated stem cell-derived beta cells may render them vulnerable to
rejection of beta cells. NK-mediated killing. HLA-E and G are less polymorphic than
There are six expressed MHC class I genes in the human HLA-A, B, and C and can be expressed in islet endocrine cells
genome: HLA-A, B, C, E, G, and F. HLA-A, B, and C are highly (Cirulli et al., 2006) and both molecules have a strong inhibitory
expressed by most nucleated cells and are most polymorphic effect on NK cells. By keeping HLA-E and G alleles intact while
(thus alloimmunogenic), whereas HLA-E, F, and G are less deleting HLA-A, B, and C, it might be possible to achieve a
polymorphic and have a more restricted expression pattern reduction in T cell responses while maintaining inhibition of
(Shiina et al., 2009). Beta 2 microglobulin (ß2M) facilitates the NK cells.
proper folding and cell surface expression of class I MHC pro- Lastly, it is important to consider graft immune surveillance
teins, and deletion of ß2M is thought to be an efficient way to when designing immunoengineering strategies of stem cell ther-
abolish all HLA I expression. However, experimental data in apies. Reducing immune recognition of stem-cell derived grafts
mice show that the dependency on ß2M for MHC I expression will also impair the efficacy of the immune system to control
is not absolute and ß2M-deficient cells are promptly rejected neoplastic growth and infection of the graft cells. A potential so-
by CD8 T cells recognizing MHC I (Bix and Raulet, 1992; lution for reducing immunogenicity while preserving some im-
Glas et al., 1992). Therefore, individual deletion of HLA I genes mune surveillance would be to retain a single HLA gene in the
may be needed to reduce alloimmunogenicity of beta cells. stem cells while eliminating others. Single HLA-expressing cells
Individual targeting of HLA I genes also offers the opportunity can be more easily matched with recipients to avoid alloimmune
to preserve less polymorphic HLA I alleles, thus ensuring rejection and a bank of stem cells can be constructed such that it
immune surveillance of the graft cells against infections and contains an array of single HLA stem cell lines to allow matching
malignancy transformation. MHC class II expression is more with most of the patient populations. For example, HLA-A2 is ex-
restricted to professional APCs in steady state. Inflammation pressed in 50% of Caucasians and Asians and beta cells derived
can lead to broader expression of HLA II. IFNg and TNF from stem cells that express only HLA-A2 could be used for 50%
together can induce HLA II expression on beta cells (Pujol-Bor- of patients from these ethnic backgrounds. However, the trade-
rell et al., 1987). Thus, abolishing HLA class II expression may off of retaining one HLA is that autoreactive T cells can recognize

816 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective
Figure 2. Cell Encapsulation Strategies for
Islet Transplantation
While encapsulation with expanded polytetra-
fluoroethylene (ePTFE) and alginate have been the
conventional approaches, more recent advances
include the use of microfabricated silicon and
nanotemplated polymer thin films, such as poly-
caprolactone (PCL). A hybrid approach utilizes an
oxygen depot to enhance cell viability. Pros and
cons of the various strategies center around ease
of implantation (injectable or not), ability for
retrieval (if needed), getting oxygen to and insulin
from the cells, and protection provided by the
membrane to cells from attack by the patient’s
immune system.

use of these therapies while reducing or


eliminating the need for immunosuppres-
sion (de Vos et al., 2010; Desai and Shea,
2017; Vaithilingam and Tuch, 2011; Vegas
et al., 2016). An ideal encapsulation de-
vice should (1) provide ample blood sup-
ply to sustain the survival and function of
a beta cell mass sufficient for the mainte-
nance of normoglycemia; (2) be biocom-
patible; (3) provide an immune protective
environment to prevent sensitization and
rejection; (4) allow secreted insulin to
rapidly exit the device for transport via
systemic circulation; and (5) contain any
potentially tumorigenic cells. Achieving
effective glucose homeostasis using
beta cell replacement therapy will require
finding ways of preventing both alloim-
mune and autoimmune responses. Poly-
and attack the beta cells. Therefore, immune modulation will still meric microcapsules have yielded some promising results for
be needed to protect the graft in patients with an underlying allogeneic cell transplantation with immune suppression, but
autoimmune disease. few approaches have been effective for non-immune-sup-
pressed encapsulation due to mechanical rupture of the mem-
Current Status and Future Direction of Cell brane, biochemical instability, and broad membrane pore size
Encapsulation Strategies distributions. Most importantly, few, if any, encapsulation strate-
Despite efforts to develop immune privileged beta cells through gies have been able to protect islets long-term from both an
tolerogenic immunotherapies or genetic manipulation of stem allogeneic and autoimmune response in a spontaneously auto-
cell-derived beta cells, there is likely to be a continued assault immune setting (Desai and Shea, 2017).
on the target tissue by both the alloimmune and autoimmune re- Encapsulation Strategies in the Clinic
sponses. A supportive approach would entail the use of a semi- Despite decades of work on encapsulation strategies, only a
permeable membrane/capsule to protect the transplanted beta handful of technologies have made it to the clinic. Perhaps the
cells from the patient’s immune system while allowing sufficient approach most widely studied and furthest along is based on
mass transfer to maintain cell viability and ensuring favorable in- microencapsulation, in which microcapsules formed from a
sulin secretion kinetics (Figure 2). Moreover, encapsulation thin layer of alginate and with a diameter in the range of 300 to
would allow patient protection from the risk of stem cell-derived 400 mm have been used to encapsulate allogeneic islets. These
beta cell oncogenic transformation. Although the notion of an capsules were delivered intraperitoneally and reduced exoge-
encapsulation strategy that can promote both beta cell survival nous insulin requirements (Basta et al., 2011). Clinical trials
and protection from immunity has been an aspiration for de- have also focused on confirming the safety of alginate microcap-
cades, the optimal device remains yet to be developed, as semi- sules with xenogeneic islets. Specifically, these trials looked at
permeable membranes that protect cells from the immune the safety of xenotransplantation of 10,000 to 20,000 IEQ/kg
system typically have not been sufficiently permeable to the nu- body weight of alginate-encapsulated porcine islets in the perito-
trients necessary to sustain their viability. However, the use of a neal cavity.
suitable encapsulation membrane as a physical barrier between One of the limitations of many encapsulation strategies is the
the recipient and hESC-derived beta cells could enable the safe lack of sufficient oxygen to maintain islet function. To overcome

Cell Stem Cell 22, June 1, 2018 817


Cell Stem Cell

Perspective

this challenge, the company Beta-O2 developed the bAir device observed in the Nanogland device where nanochannels with
to provide exogenous oxygen to transplanted islets. The disc- 3.6 and 5.7 nm pore sizes showed a reduction in glucose diffu-
shaped device consists of a compartment containing islets, sivity by 40% and 25% compared with the molecule in the
encapsulated in an alginate hydrogel slab, adjacent to a gas bulk medium (Sabek et al., 2013). Besides the long diffusion dis-
chamber where cells receive oxygen by diffusion through a tance, solutes also face reduced diffusion as their size ap-
gas-permeable membrane (Barkai et al., 2013). The subcutane- proaches the molecular dimension of the pores. Dechadilok
ously implanted device contains ports that allow daily filling with and Deen reviewed hindered transport theory for both diffusive
oxygen (Neufeld et al., 2013). A case report for this device in a and convective hindrance factors in which uncharged, spherical
single patient reported that islets retained function for the particles travel in the long cylindrical and slit pores of uniform
10-month duration of the study, with modest reduction in exog- cross-section (Dechadilok and Deen, 2006). Depending on the
enous insulin requirements (Ludwig et al., 2013). mode of transport, it is crucial to design immunoprotective mem-
Another strategy that can potentially enhance islet survival af- branes with size exclusion properties for the larger immune
ter transplantation is the enhancement of microvasculature (Gu components (e.g. cytokines, antibodies) while still permitting
et al., 2001). Two devices that have reached clinical trials, the the passage of smaller molecules (e.g. glucose, insulin).
Theracyte device and the Sernova cell pouch (Pepper et al., Although nano-sized pores are ideal to restrict the passage of
2015), aim to pre-vascularize a subcutaneous site prior to admin- immune components, encased cell functions and viability as
istration of the cells through a port. The Theracyte device is well as insulin secretion kinetics could be greatly impacted under
thought to block key immune molecules (Kumagai-Braesch the diffusive transport approach.
et al., 2013; Sörenby et al., 2008) and is composed of a two- Recently, approaches to create controlled nanopores in mate-
membrane pouch. The outer membrane has a 5 mm pore size rials other than silicon have also been explored. A nanoporous
to support cell infiltration and promote angiogenesis throughout thin-film cell encapsulation device made from polycaprolactone
the device. The inner membrane has a pore size diameter of (PCL) was developed using a nanotemplating technique (Nyitray
0.4 mm for immunoisolating the islets adjacent to the vasculature. et al., 2015), whereby well-defined pore sizes are created by
This device was most recently adapted to house stem cells by growing nanorods and then casting a film onto this substrate.
Viacyte (Agulnick et al., 2015; Robert et al., 2018). This system, While still maintaining flexibility, the material is engineered
termed Encaptra, has a single membrane that is immunoisolat- to have precise nanoscale pores and showed cell viability in
ing, while allowing oxygen and nutrients to pass, and it is allogeneic mouse models for up to 90 days. The device was
in a phase I/II clinical trial to assess safety and efficacy also able to support the function of stem cell-derived insulin-pro-
(NCT02239354). In contrast to Encaptra, the Sernova cell pouch ducing cells for up to 6 months (Chang et al., 2017) and block
is not immunoisolating. The device is inserted under the skin for antigen recognition from antigen-specific T cells. The lack of
30 days to enable vascular integration within the device. Subse- foreign body response, in combination with rapid neovasculari-
quently, a series of rods are removed to expose channels that zation around the device, demonstrates its promise for cell
can be filled with transplanted islets. The three-year phase I/II encapsulation.
clinical study using this device recently terminated after recruit- A faster mass transfer of oxygen and nutrients to the encapsu-
ing three patients (NCT01652911) (Gala-Lopez et al., 2016). lated beta cells could be achieved with an intravascular device.
Microfabricated Encapsulation Devices Moreover, convective transport with an applied pressure
Two major challenges for the successful design of next-genera- gradient results in more efficient mass transfer where solutes
tion cell encapsulation devices are providing effective immuno- actively move along with solvent flux (Song et al., 2016), which,
protection and presenting sufficient mass transfer between the in turn, enhances insulin secretion kinetics. In one configuration,
outside environment and the encased islets. As discussed capsules are constructed from silicon semipermeable mem-
above, the semipermeable membranes must exhibit a precisely branes with straight slit pores. In contrast to the previous
controlled pore size to separate soluble inflammatory mediators L-shaped pores, these straight pores exhibit shorter distances
that are on a scale of nanometers in size, while simultaneously for molecules to travel, resulting in higher permeability. This
exhibiting exceptional uniformity in pore size distribution to pro- pore geometry optimizes the tradeoff between selectivity and
vide sufficient immunoprotection. Microfabricated silicon mem- permeability of the membranes. The permeability/selectivity
branes can be used to achieve such levels of high precision con- analysis for ultrafiltration shows that membranes with monodis-
trol over pore sizes, as illustrated by examples like nanoporous persed slit-shaped pores exhibit greater selectivity at a given
biocapsules (Desai et al., 1999, 2004; Leoni and Desai, 2004) value of permeability than membranes with cylindrical pores
and Nanogland (Sabek et al., 2013). The surfaces of silicon mem- for pore size below 100 nm (Kanani et al., 2010). With the rela-
branes can also be selectively grafted with biocompatible poly- tively thin silicon membranes, convection-dominated mass
mer thin films to ensure functional performance over extended transport is advantageous, because it efficiently transports sol-
time periods, making them suitable for biological applications utes such as glucose, oxygen, and insulin between the device
(Li et al., 2011; Melvin et al., 2010; Zhu and Marchant, 2006). and the adjacent vascular system (Song et al., 2017).
The nanoporous biocapsule and Nanogland were designed
with L-shaped pore paths with perpendicular microchannels Conclusions
and parallel nanochannels to the membrane surface. This Rapid advances in device technology, immune modulation, and
L-shaped design effectively prevents diffusion of larger immune generation of functional pancreatic islet cells from human stem
components, but allows hindered diffusion of small molecules cells may soon provide novel therapeutic avenues for the treat-
due to the indirect, long diffusion distance. This effect was ment of diabetic patients. While significant technical hurdles

818 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective

remain in all these areas, the advent of new encapsulation tech- Benninger, R.K.P., Head, W.S., Zhang, M., Satin, L.S., and Piston, D.W. (2011).
Gap junctions and other mechanisms of cell-cell communication regulate
nologies, novel immune modulation approaches, and improved basal insulin secretion in the pancreatic islet. J. Physiol. 589, 5453–5466.
stem cell differentiation protocols paired with gene editing tech-
nologies carry the promise of revolutionizing modern cell therapy Bertuzzi, F., Colussi, G., Lauterio, A., and De Carlis, L. (2018). Intramuscular
islet allotransplantation in type 1 diabetes mellitus. Eur. Rev. Med. Pharmacol.
approaches for both T1D and T2D patients. Sci. 22, 1731–1736.

Bix, M., and Raulet, D. (1992). Functionally conformed free class I heavy chains
ACKNOWLEDGMENTS exist on the surface of beta 2 microglobulin negative cells. J. Exp. Med. 176,
829–834.
J.B.S., Q. T., J.A.B., P.S., and M.H. are part of the UCSF Diabetes Endocri-
nology Research Center (P30 DK063720). S.R. is supported by funds from Bluestone, J.A., Liu, W., Yabu, J.M., Laszik, Z.G., Putnam, A., Belingheri, M.,
the NIH (U01EB025136) and the JDRF Encapsulation Consortium (3-SRA- Gross, D.M., Townsend, R.M., and Vincenti, F. (2008). The effect of costimu-
2015-37-Q-R). M.H. receives support from the NIH (DK105831 and latory and interleukin 2 receptor blockade on regulatory T cells in renal trans-
U01EB025136). plantation. Am. J. Transplant. 8, 2086–2096.

Bluestone, J.A., Buckner, J.H., Fitch, M., Gitelman, S.E., Gupta, S., Hellerstein,
DECLARATION OF INTERESTS M.K., Herold, K.C., Lares, A., Lee, M.R., Li, K., et al. (2015). Type 1 diabetes
immunotherapy using polyclonal regulatory T cells. Sci. Transl. Med. 7,
315ra189.
J. S., Q.T., P.S., and M.H. serve on the scientific advisory board for Encellin,
Inc. T.D. is the founder and serves on the scientific advisory board for Encellin
Bonner-Weir, S., and Orci, L. (1982). New perspectives on the microvascula-
Inc. S.R. is the founder and has an ownership of Silicon Kidney. M.H. is on the ture of the islets of Langerhans in the rat. Diabetes 31, 883–889.
scientific advisory board of Semma, Inc. and holds stock options in Viacyte,
Inc. S.R. holds the US Patent 9,802,158 - Ultrafiltration membrane device, bio- Bonner-Weir, S., Sullivan, B.A., and Weir, G.C. (2015). Human Islet
artificial organ, and related methods. M.H. is an inventor on the US Patent No. Morphology Revisited: Human and Rodent Islets Are Not So Different After
9,850,465 - Generation of Thymic Epithelial Progenitor Cells In Vitro, as well as All. J. Histochem. Cytochem. 63, 604–612.
provisional US Patent applications: Case 2015-203-4 PCT: PCT/US17/26651
(filed 4/07/2017): Controlled induction of human pancreatic progenitors pro- Bosco, D., Armanet, M., Morel, P., Niclauss, N., Sgroi, A., Muller, Y.D., Giovan-
duces functional beta-like cells in vitro; Case 2016-019-4: Application number noni, L., Parnaud, G., and Berney, T. (2010). Unique arrangement of alpha- and
15/747,729 (filed 7/26/16): Generation of human beta cell equivalents from beta-cells in human islets of Langerhans. Diabetes 59, 1202–1210.
pluripotent stem cells in vitro; Case 2018-064-1 PRV: (filed 12/12/2017): The
use of parathyroid gland cells and their secreted factors to promote islet Bougnères, P.F., Landais, P., Boisson, C., Carel, J.C., Frament, N., Boitard, C.,
beta cell engraftments. App 62/597,825. T.D. is an inventor on the Chaussain, J.L., and Bach, J.F. (1990). Limited duration of remission of insulin
patent application PCT/US2016/023808 Entitled: Thin Film Cell Encapsulation dependency in children with recent overt type I diabetes treated with low-dose
Device, Ref. SF2015-176-2. Q.T. and J.A.B. are inventors on the patents cyclosporin. Diabetes 39, 1264–1272.
Regulatory T Cells Suppress Autoimmunity, US Patent #7,722,862,B2
Bour-Jordan, H., Salomon, B.L., Thompson, H.L., Szot, G.L., Bernhard, M.R.,
(issued 5/25/2010) and Expansion of alloantigen-reactive regulatory T cells,
and Bluestone, J.A. (2004). Costimulation controls diabetes by altering the bal-
US Patent #14/382.537 (issued 10/31/2017). Q.T. and P.S. are inventors on ance of pathogenic and regulatory T cells. J. Clin. Invest. 114, 979–987.
the patent application Case 2018-064-1 PRV: (filed 12/12/2017): The use of
parathyroid gland cells and their secreted factors to promote islet beta cell en- Brennan, T.V., Jaigirdar, A., Hoang, V., Hayden, T., Liu, F.-C., Zaid, H., Chang,
graftments. App 62/597,825. J.A.B. is also inventor on the patent CD127 C.K., Bucy, R.P., Tang, Q., and Kang, S.-M. (2009). Preferential priming of al-
expression inversely correlates with Foxp3 and suppresses function of loreactive T cells with indirect reactivity. Am. J. Transplant. 9, 709–718.
CD4+ Tregs.
Burke, G.W., 3rd, Vendrame, F., Virdi, S.K., Ciancio, G., Chen, L., Ruiz, P.,
Messinger, S., Reijonen, H.K., and Pugliese, A. (2015). Lessons From
REFERENCES Pancreas Transplantation in Type 1 Diabetes: Recurrence of Islet Autoimmu-
nity. Curr. Diab. Rep. 15, 121.
Agulnick, A.D., Ambruzs, D.M., Moorman, M.A., Bhoumik, A., Cesario, R.M.,
Payne, J.K., Kelly, J.R., Haakmeester, C., Srijemac, R., Wilson, A.Z., et al. Cabrera, O., Berman, D.M., Kenyon, N.S., Ricordi, C., Berggren, P.-O., and
(2015). Insulin-Producing Endocrine Cells Differentiated In Vitro From Human Caicedo, A. (2006). The unique cytoarchitecture of human pancreatic islets
Embryonic Stem Cells Function in Macroencapsulation Devices In Vivo. has implications for islet cell function. Proc. Natl. Acad. Sci. USA 103,
Stem Cells Transl. Med. 4, 1214–1222. 2334–2339.

Balamurugan, A.N., Naziruddin, B., Lockridge, A., Tiwari, M., Loganathan, G., Chang, C.H., Guerder, S., Hong, S.C., van Ewijk, W., and Flavell, R.A. (1996).
Takita, M., Matsumoto, S., Papas, K., Trieger, M., Rainis, H., et al. (2014). Islet Mice lacking the MHC class II transactivator (CIITA) show tissue-specific
product characteristics and factors related to successful human islet trans- impairment of MHC class II expression. Immunity 4, 167–178.
plantation from the Collaborative Islet Transplant Registry (CITR) 1999-2010.
Am. J. Transplant. 14, 2595–2606. Chang, R., Faleo, G., Russ, H.A., Parent, A.V., Elledge, S.K., Bernards, D.A.,
Allen, J.L., Villanueva, K., Hebrok, M., Tang, Q., and Desai, T.A. (2017). Nano-
Barkai, U., Weir, G.C., Colton, C.K., Ludwig, B., Bornstein, S.R., Brendel, M.D., porous Immunoprotective Device for Stem-Cell-Derived b-Cell Replacement
Neufeld, T., Bremer, C., Leon, A., Evron, Y., et al. (2013). Enhanced oxygen Therapy. ACS Nano 11, 7747–7757.
supply improves islet viability in a new bioartificial pancreas. Cell Transplant.
22, 1463–1476. Chong, M.M., Thomas, H.E., and Kay, T.W. (2001). gamma-Interferon
signaling in pancreatic beta-cells is persistent but can be terminated by over-
Barton, F.B., Rickels, M.R., Alejandro, R., Hering, B.J., Wease, S., Naziruddin, expression of suppressor of cytokine signaling-1. Diabetes 50, 2744–2751.
B., Oberholzer, J., Odorico, J.S., Garfinkel, M.R., Levy, M., et al. (2012).
Improvement in outcomes of clinical islet transplantation: 1999-2010. Dia- Chong, M.M.W., Thomas, H.E., and Kay, T.W.H. (2002). Suppressor of cyto-
betes Care 35, 1436–1445. kine signaling-1 regulates the sensitivity of pancreatic beta cells to tumor ne-
crosis factor. J. Biol. Chem. 277, 27945–27952.
Basta, G., Montanucci, P., Luca, G., Boselli, C., Noya, G., Barbaro, B., Qi, M.,
Kinzer, K.P., Oberholzer, J., and Calafiore, R. (2011). Long-term metabolic and Cirulli, V., Zalatan, J., McMaster, M., Prinsen, R., Salomon, D.R., Ricordi, C.,
immunological follow-up of nonimmunosuppressed patients with type 1 dia- Torbett, B.E., Meda, P., and Crisa, L. (2006). The class I HLA repertoire of
betes treated with microencapsulated islet allografts: four cases. Diabetes pancreatic islets comprises the nonclassical class Ib antigen HLA-G. Diabetes
Care 34, 2406–2409. 55, 1214–1222.

Becker, Y.T., Becker, B.N., Pirsch, J.D., and Sollinger, H.W. (2004). Rituximab D’Amour, K.A., Agulnick, A.D., Eliazer, S., Kelly, O.G., Kroon, E., and Baetge,
as treatment for refractory kidney transplant rejection. Am. J. Transplant. 4, E.E. (2005). Efficient differentiation of human embryonic stem cells to definitive
996–1001. endoderm. Nat. Biotechnol. 23, 1534–1541.

Cell Stem Cell 22, June 1, 2018 819


Cell Stem Cell

Perspective
D’Amour, K.A., Bang, A.G., Eliazer, S., Kelly, O.G., Agulnick, A.D., Smart, N.G., Grey, S.T., Arvelo, M.B., Hasenkamp, W., Bach, F.H., and Ferran, C. (1999).
Moorman, M.A., Kroon, E., Carpenter, M.K., and Baetge, E.E. (2006). Produc- A20 inhibits cytokine-induced apoptosis and nuclear factor kappaB-depen-
tion of pancreatic hormone-expressing endocrine cells from human embryonic dent gene activation in islets. J. Exp. Med. 190, 1135–1146.
stem cells. Nat. Biotechnol. 24, 1392–1401.
Gruessner, A.C., and Gruessner, R.W.G. (2016). Long-term outcome after
de Vos, P., Spasojevic, M., and Faas, M.M. (2010). Treatment of diabetes with pancreas transplantation: a registry analysis. Curr. Opin. Organ Transplant.
encapsulated islets. Adv. Exp. Med. Biol. 670, 38–53. 21, 377–385.
Dechadilok, P., and Deen, W.M. (2006). Hindrance Factors for Diffusion and Gu, Y., Tabata, Y., Kawakami, Y., Balamurugan, A.N., Hori, H., Nagata, N., Sa-
Convection in Pores. Ind. Eng. Chem. Res. 45, 6953–6959. take, A., Cui, W., Qi, M., Misawa, Y., et al. (2001). Development of a new
method to induce angiogenesis at subcutaneous site of streptozotocin-
Desai, T., and Shea, L.D. (2017). Advances in islet encapsulation technologies. induced diabetic rats for islet transplantation. Cell Transplant. 10, 453–457.
Nat. Rev. Drug Discov. 16, 338–350.

Desai, T.A., Chu, W.H., Rasi, G., Sinibaldi-Vallebona, P., Guarino, E., and Fer- Gu, Y., Gong, C., Peng, X., Wei, L., Su, C., Qin, M., Wang, X., and Li, F. (2014).
rari, M. (1999). Microfabricated biocapsules provide short-term immunoisola- Autologous hematopoietic stem cell transplantation and conventional insulin
tion of insulinoma xenografts. Biomed. Microdevices 1, 131–138. therapy in the treatment of children with newly diagnosed type 1 diabetes:
long term follow-up. Chin. Med. J. (Engl.) 127, 2618–2622.
Desai, T.A., West, T., Cohen, M., Boiarski, T., and Rampersaud, A. (2004).
Nanoporous microsystems for islet cell replacement. Adv. Drug Deliv. Rev. Guo, T., Landsman, L., Li, N., and Hebrok, M. (2013). Factors expressed by
56, 1661–1673. murine embryonic pancreatic mesenchyme enhance generation of insulin-pro-
ducing cells from hESCs. Diabetes 62, 1581–1592.
Dı́az-Flores, L., Gutiérrez, R., Madrid, J.F., Varela, H., Valladares, F., Acosta,
E., Martı́n-Vasallo, P., and Dı́az-Flores, L., Jr. (2009). Pericytes. Morphofunc- Hagopian, W., Ferry, R.J., Jr., Sherry, N., Carlin, D., Bonvini, E., Johnson, S.,
tion, interactions and pathology in a quiescent and activated mesenchymal Stein, K.E., Koenig, S., Daifotis, A.G., Herold, K.C., and Ludvigsson, J.;
cell niche. Histol. Histopathol. 24, 909–969. Protégé Trial Investigators (2013). Teplizumab preserves C-peptide in
recent-onset type 1 diabetes: two-year results from the randomized, pla-
Dorrell, C., Schug, J., Canaday, P.S., Russ, H.A., Tarlow, B.D., Grompe, M.T., cebo-controlled Protégé trial. Diabetes 62, 3901–3908.
Horton, T., Hebrok, M., Streeter, P.R., Kaestner, K.H., and Grompe, M. (2016).
Human islets contain four distinct subtypes of b cells. Nat. Commun. 7, 11756. Halban, P.A., Wollheim, C.B., Blondel, B., Meda, P., Niesor, E.N., and Mintz,
D.H. (1982). The possible importance of contact between pancreatic islet cells
Eich, T., Eriksson, O., and Lundgren, T.; Nordic Network for Clinical Islet Trans- for the control of insulin release. Endocrinology 111, 86–94.
plantation (2007). Visualization of early engraftment in clinical islet transplanta-
tion by positron-emission tomography. N. Engl. J. Med. 356, 2754–2755. Haller, M.J., Gitelman, S.E., Gottlieb, P.A., Michels, A.W., Rosenthal, S.M.,
Shuster, J.J., Zou, B., Brusko, T.M., Hulme, M.A., Wasserfall, C.H., et al.
Eizirik, D.L., Colli, M.L., and Ortis, F. (2009). The role of inflammation in insulitis (2015). Anti-thymocyte globulin/G-CSF treatment preserves b cell function in
and beta-cell loss in type 1 diabetes. Nat. Rev. Endocrinol. 5, 219–226. patients with established type 1 diabetes. J. Clin. Invest. 125, 448–455.
Esmatjes, E., Montaña, X., Real, M.I., Blanco, J., Conget, I., Casamitjana, R.,
Hamilton-Williams, E.E., Palmer, S.E., Charlton, B., and Slattery, R.M. (2003).
Rovira, M., Gomis, R., and Marin, P. (2010). Regeneration of insulin production
Beta cell MHC class I is a late requirement for diabetes. Proc. Natl. Acad. Sci.
by autologous bone marrow blood autotransplantation in patients with type 1
USA 100, 6688–6693.
diabetes. Diabetologia 53, 786–789.

Feutren, G., Papoz, L., Assan, R., Vialettes, B., Karsenty, G., Vexiau, P., Du Hankey, K.G., Drachenberg, C.B., Papadimitriou, J.C., Klassen, D.K., Philo-
Rostu, H., Rodier, M., Sirmai, J., Lallemand, A., et al. (1986). Cyclosporin in- sophe, B., Bartlett, S.T., Groh, V., Spies, T., and Mann, D.L. (2002). MIC
creases the rate and length of remissions in insulin-dependent diabetes of expression in renal and pancreatic allografts. Transplantation 73, 304–306.
recent onset. Results of a multicentre double-blind trial. Lancet 2, 119–124.
Hayden, M.R., Patel, K., Habibi, J., Gupta, D., Tekwani, S.S., Whaley-Connell,
Fioretto, P., Steffes, M.W., Sutherland, D.E., Goetz, F.C., and Mauer, M. A., and Sowers, J.R. (2008). Attenuation of endocrine-exocrine pancreatic
(1998). Reversal of lesions of diabetic nephropathy after pancreas transplanta- communication in type 2 diabetes: pancreatic extracellular matrix ultrastruc-
tion. N. Engl. J. Med. 339, 69–75. tural abnormalities. J. Cardiometab. Syndr. 3, 234–243.

Foulis, A.K., and Farquharson, M.A. (1986). Aberrant expression of HLA-DR Henderson, J.R., and Moss, M.C. (1985). A morphometric study of the endo-
antigens by insulin-containing beta-cells in recent-onset type I diabetes melli- crine and exocrine capillaries of the pancreas. Q. J. Exp. Physiol. 70, 347–356.
tus. Diabetes 35, 1215–1224.
Hering, B.J., Clarke, W.R., Bridges, N.D., Eggerman, T.L., Alejandro, R., Bellin,
Furukawa, A., Wisel, S.A., and Tang, Q. (2016). Impact of Immune-Modulatory M.D., Chaloner, K., Czarniecki, C.W., Goldstein, J.S., Hunsicker, L.G., et al.;
Drugs on Regulatory T Cell. Transplantation 100, 2288–2300. Clinical Islet Transplantation Consortium (2016). Phase 3 Trial of Transplanta-
tion of Human Islets in Type 1 Diabetes Complicated by Severe Hypoglycemia.
Gala-Lopez, B.L., Pepper, A.R., Dinyari, P., Malcolm, A.J., Kin, T., Pawlick, Diabetes Care 39, 1230–1240.
L.R., Senior, P.A., and Shapiro, A.M.J. (2016). Subcutaneous clinical islet
transplantation in a prevascularized subcutaneous pouch - preliminary expe- Herold, K.C., Hagopian, W., Auger, J.A., Poumian-Ruiz, E., Taylor, L., Donald-
rience. CellR4 4, e2132. son, D., Gitelman, S.E., Harlan, D.M., Xu, D., Zivin, R.A., and Bluestone, J.A.
(2002). Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus.
Gamble, A., Pepper, A.R., Bruni, A., and Shapiro, A.M.J. (2018). The journey of
N. Engl. J. Med. 346, 1692–1698.
islet cell transplantation and future development. Islets 10, 80–94.

Gitelman, S.E., Gottlieb, P.A., Rigby, M.R., Felner, E.I., Willi, S.M., Fisher, L.K., Herold, K.C., Gitelman, S.E., Masharani, U., Hagopian, W., Bisikirska, B., Do-
Moran, A., Gottschalk, M., Moore, W.V., Pinckney, A., et al.; START Study naldson, D., Rother, K., Diamond, B., Harlan, D.M., and Bluestone, J.A. (2005).
Team (2013). Antithymocyte globulin treatment for patients with recent-onset A single course of anti-CD3 monoclonal antibody hOKT3gamma1(Ala-Ala) re-
type 1 diabetes: 12-month results of a randomised, placebo-controlled, phase sults in improvement in C-peptide responses and clinical parameters for at
2 trial. Lancet Diabetes Endocrinol. 1, 306–316. least 2 years after onset of type 1 diabetes. Diabetes 54, 1763–1769.

Gitelman, S.E., Gottlieb, P.A., Felner, E.I., Willi, S.M., Fisher, L.K., Moran, A., Holman, R.R., Paul, S.K., Bethel, M.A., Matthews, D.R., and Neil, H.A.W.
Gottschalk, M., Moore, W.V., Pinckney, A., Keyes-Elstein, L., et al.; ITN START (2008). 10-year follow-up of intensive glucose control in type 2 diabetes.
Study Team (2016). Antithymocyte globulin therapy for patients with recent- N. Engl. J. Med. 359, 1577–1589.
onset type 1 diabetes: 2 year results of a randomised trial. Diabetologia 59,
1153–1161. Jain, R.K. (2003). Molecular regulation of vessel maturation. Nat. Med. 9,
685–693.
Glas, R., Franksson, L., Ohlén, C., Höglund, P., Koller, B., Ljunggren, H.G.,
and Ka €rre, K. (1992). Major histocompatibility complex class I-specific Jiang, S., Herrera, O., and Lechler, R.I. (2004). New spectrum of allorecogni-
and -restricted killing of beta 2-microglobulin-deficient cells by CD8+ cyto- tion pathways: implications for graft rejection and transplantation tolerance.
toxic T lymphocytes. Proc. Natl. Acad. Sci. USA 89, 11381–11385. Curr. Opin. Immunol. 16, 550–557.

820 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective
Kanani, D.M., Fissell, W.H., Roy, S., Dubnisheva, A., Fleischman, A., and Zyd- Makhlouf, L., Yamada, A., Ito, T., Abdi, R., Ansari, M.J.I., Khuong, C.Q., Winn,
ney, A.L. (2010). Permeability - Selectivity Analysis for Ultrafiltration: Effect of H.J., Auchincloss, H., Jr., and Sayegh, M.H. (2003). Allorecognition and
Pore Geometry. J. Membr. Sci. 349, 405. effector pathways of islet allograft rejection in normal versus nonobese dia-
betic mice. J. Am. Soc. Nephrol. 14, 2168–2175.
Kandaswamy, R., Stock, P.G., Gustafson, S.K., Skeans, M.A., Curry, M.A.,
Prentice, M.A., Fox, A., Israni, A.K., Snyder, J.J., and Kasiske, B.L. (2018). Marek-Trzonkowska, N., Mysliwiec, M., Dobyszuk, A., Grabowska, M., Tech-
OPTN/SRTR 2016 Annual Data Report: Pancreas. Am. J. Transplant. 18 manska, I., Juscinska, J., Wujtewicz, M.A., Witkowski, P., Mlynarski, W., Bal-
(Suppl 1 ), 114–171. cerska, A., et al. (2012). Administration of CD4+CD25highCD127- regulatory
T cells preserves b-cell function in type 1 diabetes in children. Diabetes Care
Kawai, T., and Sachs, D.H. (2013). Tolerance induction: hematopoietic chime- 35, 1817–1820.
rism. Curr. Opin. Organ Transplant. 18, 402–407.
Melvin, M.E., Fissell, W.H., Roy, S., and Brown, D.L. (2010). Silicon induces
Keymeulen, B., Vandemeulebroucke, E., Ziegler, A.-G., Mathieu, C., Kaufman, minimal thromboinflammatory response during 28-day intravascular implant
L., Hale, G., Gorus, F., Goldman, M., Walter, M., Candon, S., et al. (2005). In- testing. ASAIO J. 56, 344–348.
sulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N. Engl.
J. Med. 352, 2598–2608. €rtler, M., Pagliuca, F.W., and Melton,
Millman, J.R., Xie, C., Van Dervort, A., Gu
D.A. (2016). Generation of stem cell-derived b-cells from patients with type 1
Keymeulen, B., Walter, M., Mathieu, C., Kaufman, L., Gorus, F., Hilbrands, R., diabetes. Nat. Commun. 7, 11463.
Vandemeulebroucke, E., Van de Velde, U., Crenier, L., De Block, C., et al.
Moassesfar, S., Masharani, U., Frassetto, L.A., Szot, G.L., Tavakol, M., Stock,
(2010). Four-year metabolic outcome of a randomised controlled CD3-anti-
P.G., and Posselt, A.M. (2016). A Comparative Analysis of the Safety, Efficacy,
body trial in recent-onset type 1 diabetic patients depends on their age and
and Cost of Islet Versus Pancreas Transplantation in Nonuremic Patients With
baseline residual beta cell mass. Diabetologia 53, 614–623.
Type 1 Diabetes. Am. J. Transplant. 16, 518–526.
Kilimnik, G., Jo, J., Periwal, V., Zielinski, M.C., and Hara, M. (2012). Quantifica- Nathan, D.M., Genuth, S., Lachin, J., Cleary, P., Crofford, O., Davis, M., Rand,
tion of islet size and architecture. Islets 4, 167–172. L., and Siebert, C.; Diabetes Control and Complications Trial Research Group
(1993). The effect of intensive treatment of diabetes on the development and
Kojima, N. (2014). In vitro reconstitution of pancreatic islets. Organogenesis progression of long-term complications in insulin-dependent diabetes melli-
10, 225–230. tus. N. Engl. J. Med. 329, 977–986.
Kroon, E., Martinson, L.A., Kadoya, K., Bang, A.G., Kelly, O.G., Eliazer, S., Neufeld, T., Ludwig, B., Barkai, U., Weir, G.C., Colton, C.K., Evron, Y., Balyura,
Young, H., Richardson, M., Smart, N.G., Cunningham, J., et al. (2008). Pancre- M., Yavriyants, K., Zimermann, B., Azarov, D., et al. (2013). The efficacy of an
atic endoderm derived from human embryonic stem cells generates glucose- immunoisolating membrane system for islet xenotransplantation in minipigs.
responsive insulin-secreting cells in vivo. Nat. Biotechnol. 26, 443–452. PLoS ONE 8, e70150.
Kumagai-Braesch, M., Jacobson, S., Mori, H., Jia, X., Takahashi, T., Werner- Nikolova, G., Jabs, N., Konstantinova, I., Domogatskaya, A., Tryggvason, K.,
son, A., Flodström-Tullberg, M., and Tibell, A. (2013). The TheraCyte device Sorokin, L., Fa €ssler, R., Gu, G., Gerber, H.P., Ferrara, N., et al. (2006). The
protects against islet allograft rejection in immunized hosts. Cell Transplant. vascular basement membrane: a niche for insulin gene expression and Beta
22, 1137–1146. cell proliferation. Dev. Cell 10, 397–405.

Kupfer, T.M., Crawford, M.L., Pham, K., and Gill, R.G. (2005). MHC-mis- Nyitray, C.E., Chang, R., Faleo, G., Lance, K.D., Bernards, D.A., Tang, Q., and
matched islet allografts are vulnerable to autoimmune recognition in vivo. Desai, T.A. (2015). Polycaprolactone Thin-Film Micro- and Nanoporous Cell-
J. Immunol. 175, 2309–2316. Encapsulation Devices. ACS Nano 9, 5675–5682.

Lammert, E., Cleaver, O., and Melton, D. (2003a). Role of endothelial cells in Oberg-Welsh, C. (2001). Long-term culture in matrigel enhances the insulin
early pancreas and liver development. Mech. Dev. 120, 59–64. secretion of fetal porcine islet-like cell clusters in vitro. Pancreas 22, 157–163.

Lammert, E., Gu, G., McLaughlin, M., Brown, D., Brekken, R., Murtaugh, L.C., Ogasawara, K., Hamerman, J.A., Ehrlich, L.R., Bour-Jordan, H., Santamaria,
Gerber, H.P., Ferrara, N., and Melton, D.A. (2003b). Role of VEGF-A in vascu- P., Bluestone, J.A., and Lanier, L.L. (2004). NKG2D blockade prevents autoim-
larization of pancreatic islets. Curr. Biol. 13, 1070–1074. mune diabetes in NOD mice. Immunity 20, 757–767.

Lanier, L.L. (2008). Up on the tightrope: natural killer cell activation and inhibi- Orban, T., Bundy, B., Becker, D.J., DiMeglio, L.A., Gitelman, S.E., Goland, R.,
tion. Nat. Immunol. 9, 495–502. Gottlieb, P.A., Greenbaum, C.J., Marks, J.B., Monzavi, R., et al.; Type 1 Dia-
betes TrialNet Abatacept Study Group (2011). Co-stimulation modulation
Lee, K., Nguyen, V., Lee, K.-M., Kang, S.-M., and Tang, Q. (2014). Attenuation with abatacept in patients with recent-onset type 1 diabetes: a randomised,
of donor-reactive T cells allows effective control of allograft rejection using reg- double-blind, placebo-controlled trial. Lancet 378, 412–419.
ulatory T cell therapy. Am. J. Transplant. 14, 27–38.
Otonkoski, T., Banerjee, M., Korsgren, O., Thornell, L.-E., and Virtanen, I.
Leoni, L., and Desai, T.A. (2004). Micromachined biocapsules for cell-based (2008). Unique basement membrane structure of human pancreatic islets: im-
sensing and delivery. Adv. Drug Deliv. Rev. 56, 211–229. plications for beta-cell growth and differentiation. Diabetes Obes. Metab. 10
(Suppl 4 ), 119–127.
Li, L., Marchant, R.E., Dubnisheva, A., Roy, S., and Fissell, W.H. (2011). Anti- €rtler, M., Segel, M., Van Dervort, A., Ryu, J.H.,
Pagliuca, F.W., Millman, J.R., Gu
biofouling Sulfobetaine Polymer Thin Films on Silicon and Silicon Nanopore
Peterson, Q.P., Greiner, D., and Melton, D.A. (2014). Generation of functional
Membranes. J. Biomater. Sci. Polym. Ed. 22, 91–106.
human pancreatic b cells in vitro. Cell 159, 428–439.
Lin, S., Staahl, B.T., Alla, R.K., and Doudna, J.A. (2014). Enhanced homology- Pan, F.C., and Wright, C. (2011). Pancreas organogenesis: from bud to plexus
directed human genome engineering by controlled timing of CRISPR/Cas9 de- to gland. Dev. Dyn. 240, 530–565.
livery. eLife 3, e04766.
Penko, D., Mohanasundaram, D., Sen, S., Drogemuller, C., Mee, C., Bonder,
Long, S.A., Thorpe, J., Herold, K.C., Ehlers, M., Sanda, S., Lim, N., Linsley, C.S., Coates, P.T.H., and Jessup, C.F. (2011). Incorporation of endothelial pro-
P.S., Nepom, G.T., and Harris, K.M. (2017). Remodeling T cell compartments genitor cells into mosaic pseudoislets. Islets 3, 73–79.
during anti-CD3 immunotherapy of type 1 diabetes. Cell. Immunol. 319, 3–9.
Pepper, A.R., Pawlick, R., Gala-Lopez, B., MacGillivary, A., Mazzuca, D.M.,
Ludvigsson, J., Krisky, D., Casas, R., Battelino, T., Castaño, L., Greening, J., White, D.J.G., Toleikis, P.M., and Shapiro, A.M.J. (2015). Diabetes Is Reversed
Kordonouri, O., Otonkoski, T., Pozzilli, P., Robert, J.-J., et al. (2012). GAD65 in a Murine Model by Marginal Mass Syngeneic Islet Transplantation Using a
antigen therapy in recently diagnosed type 1 diabetes mellitus. N. Engl. J. Subcutaneous Cell Pouch Device. Transplantation 99, 2294–2300.
Med. 366, 433–442.
Pescovitz, M.D., Greenbaum, C.J., Krause-Steinrauf, H., Becker, D.J., Gitel-
Ludwig, B., Reichel, A., Steffen, A., Zimerman, B., Schally, A.V., Block, N.L., man, S.E., Goland, R., Gottlieb, P.A., Marks, J.B., McGee, P.F., Moran,
Colton, C.K., Ludwig, S., Kersting, S., Bonifacio, E., et al. (2013). Transplanta- A.M., et al.; Type 1 Diabetes TrialNet Anti-CD20 Study Group (2009). Rituxi-
tion of human islets without immunosuppression. Proc. Natl. Acad. Sci. USA mab, B-lymphocyte depletion, and preservation of beta-cell function.
110, 19054–19058. N. Engl. J. Med. 361, 2143–2152.

Cell Stem Cell 22, June 1, 2018 821


Cell Stem Cell

Perspective
Posselt, A.M., Szot, G.L., Frassetto, L.A., Masharani, U., Tavakol, M., Amin, R., Shiina, T., Hosomichi, K., Inoko, H., and Kulski, J.K. (2009). The HLA genomic
McElroy, J., Ramos, M.D., Kerlan, R.K., Fong, L., et al. (2010). Islet transplan- loci map: expression, interaction, diversity and disease. J. Hum. Genet.
tation in type 1 diabetic patients using calcineurin inhibitor-free immunosup- 54, 15–39.
pressive protocols based on T-cell adhesion or costimulation blockade. Trans-
plantation 90, 1595–1601. Solomon, M., Flodström-Tullberg, M., and Sarvetnick, N. (2011). Beta-cell
specific expression of suppressor of cytokine signaling-1 (SOCS-1) delays
Pujol-Borrell, R., Todd, I., Doshi, M., Bottazzo, G.F., Sutton, R., Gray, D., Adolf, islet allograft rejection by down-regulating Interferon Regulatory Factor-1
G.R., and Feldmann, M. (1987). HLA class II induction in human islet cells by (IRF-1) signaling. Transpl. Immunol. 24, 181–188.
interferon-gamma plus tumour necrosis factor or lymphotoxin. Nature 326,
304–306. Song, S., Faleo, G., Yeung, R., Kant, R., Posselt, A.M., Desai, T.A., Tang, Q.,
and Roy, S. (2016). Silicon nanopore membrane (SNM) for islet encapsulation
€ldenagel, M., Charollais, A., Gjinovci, A., Caille, D., Söhl, G.,
Ravier, M.A., Gu and immunoisolation under convective transport. Sci. Rep. 6, 23679.
Wollheim, C.B., Willecke, K., Henquin, J.-C., and Meda, P. (2005). Loss of con-
Song, S., Blaha, C., Moses, W., Park, J., Wright, N., Groszek, J., Fissell, W.,
nexin36 channels alters beta-cell coupling, islet synchronization of glucose-
Vartanian, S., Posselt, A.M., and Roy, S. (2017). An intravascular bioartificial
induced Ca2+ and insulin oscillations, and basal insulin release. Diabetes
pancreas device (iBAP) with silicon nanopore membranes (SNM) for islet
54, 1798–1807.
encapsulation under convective mass transport. Lab Chip 17, 1778–1792.
Reinert, R.B., Cai, Q., Hong, J.-Y., Plank, J.L., Aamodt, K., Prasad, N., Ara- Sörenby, A.K., Kumagai-Braesch, M., Sharma, A., Hultenby, K.R., Wernerson,
mandla, R., Dai, C., Levy, S.E., Pozzi, A., et al. (2014). Vascular endothelial A.M., and Tibell, A.B. (2008). Preimplantation of an immunoprotective device
growth factor coordinates islet innervation via vascular scaffolding. Develop- can lower the curative dose of islets to that of free islet transplantation: studies
ment 141, 1480–1491. in a rodent model. Transplantation 86, 364–366.
Rezania, A., Riedel, M.J., Wideman, R.D., Karanu, F., Ao, Z., Warnock, G.L., Stegall, M.D., Lafferty, K.J., Kam, I., and Gill, R.G. (1996). Evidence of recurrent
and Kieffer, T.J. (2011). Production of functional glucagon-secreting a-cells autoimmunity in human allogeneic islet transplantation. Transplantation 61,
from human embryonic stem cells. Diabetes 60, 239–247. 1272–1274.

Rezania, A., Bruin, J.E.J., Riedel, M.J.M., Mojibian, M., Asadi, A., Xu, J., Stendahl, J.C., Kaufman, D.B., and Stupp, S.I. (2009). Extracellular matrix in
Gauvin, R., Narayan, K., Karanu, F., O’Neil, J.J.J., et al. (2012). Maturation pancreatic islets: relevance to scaffold design and transplantation. Cell Trans-
of human embryonic stem cell-derived pancreatic progenitors into functional plant. 18, 1–12.
islets capable of treating pre-existing diabetes in mice. Diabetes 61,
2016–2029. Stiller, C.R., Dupré, J., Gent, M., Jenner, M.R., Keown, P.A., Laupacis, A., Mar-
tell, R., Rodger, N.W., von Graffenried, B., and Wolfe, B.M. (1984). Effects of
Rezania, A., Bruin, J.E., Arora, P., Rubin, A., Batushansky, I., Asadi, A., cyclosporine immunosuppression in insulin-dependent diabetes mellitus of
O’Dwyer, S., Quiskamp, N., Mojibian, M., Albrecht, T., et al. (2014). Reversal recent onset. Science 223, 1362–1367.
of diabetes with insulin-producing cells derived in vitro from human pluripotent
stem cells. Nat. Biotechnol. 32, 1121–1133. Tang, Q., and Bluestone, J.A. (2013). Regulatory T-cell therapy in transplanta-
tion: moving to the clinic. Cold Spring Harb. Perspect. Med. 3, a015552.
Rigby, M.R., Harris, K.M., Pinckney, A., DiMeglio, L.A., Rendell, M.S., Felner,
Tang, Q., Henriksen, K.J., Bi, M., Finger, E.B., Szot, G., Ye, J., Masteller, E.L.,
E.I., Dostou, J.M., Gitelman, S.E., Griffin, K.J., Tsalikian, E., et al. (2015). Ale-
McDevitt, H., Bonyhadi, M., and Bluestone, J.A. (2004). In vitro-expanded an-
facept provides sustained clinical and immunological effects in new-onset
tigen-specific regulatory T cells suppress autoimmune diabetes. J. Exp. Med.
type 1 diabetes patients. J. Clin. Invest. 125, 3285–3296.
199, 1455–1465.
Robert, T., De Mesmaeker, I., Stangé, G.M., Suenens, K.G., Ling, Z., Kroon, Tintle, S., Shemer, A., Suárez-Fariñas, M., Fujita, H., Gilleaudeau, P., Sullivan-
E.J., and Pipeleers, D.G. (2018). Functional Beta Cell Mass from Device- Whalen, M., Johnson-Huang, L., Chiricozzi, A., Cardinale, I., Duan, S., et al.
Encapsulated hESC-Derived Pancreatic Endoderm Achieving Metabolic Con- (2011). Reversal of atopic dermatitis with narrow-band UVB phototherapy
trol. Stem Cell Reports 10, 739–750. and biomarkers for therapeutic response. J. Allergy Clin. Immunol. 128,
583–593, e1–4.
Russ, H.A., Parent, A.V., Ringler, J.J., Hennings, T.G., Nair, G.G., Shveygert,
M., Guo, T., Puri, S., Haataja, L., Cirulli, V., et al. (2015). Controlled induction UK Prospective Diabetes Study (UKPDS) Group (1998a). Intensive blood-
of human pancreatic progenitors produces functional beta-like cells in vitro. glucose control with sulphonylureas or insulin compared with conventional
EMBO J. 34, 1759–1772. treatment and risk of complications in patients with type 2 diabetes (UKPDS
33). Lancet 352, 837–853.
Sabek, O.M., Ferrati, S., Fraga, D.W., Sih, J., Zabre, E.V., Fine, D.H., Ferrari,
M., Gaber, A.O., and Grattoni, A. (2013). Characterization of a nanogland for UK Prospective Diabetes Study (UKPDS) Group (1998b). Effect of intensive
the autotransplantation of human pancreatic islets. Lab Chip 13, 3675–3688. blood-glucose control with metformin on complications in overweight patients
with type 2 diabetes (UKPDS 34). Lancet 352, 854–865.
Salomon, B., and Bluestone, J.A. (2001). Complexities of CD28/B7: CTLA-4
costimulatory pathways in autoimmunity and transplantation. Annu. Rev. Vaithilingam, V., and Tuch, B.E. (2011). Islet transplantation and encapsula-
Immunol. 19, 225–252. tion: an update on recent developments. Rev. Diabet. Stud. 8, 51–67.

Sasson, A., Rachi, E., Sakhneny, L., Baer, D., Lisnyansky, M., Epshtein, A., and Vegas, A.J., Veiseh, O., Gu €rtler, M., Millman, J.R., Pagliuca, F.W., Bader, A.R.,
Landsman, L. (2016). Islet pericytes are required for beta-cell maturity. Dia- Doloff, J.C., Li, J., Chen, M., Olejnik, K., et al. (2016). Long-term glycemic con-
betes 65, 3008–3014. trol using polymer-encapsulated human stem cell-derived beta cells in im-
mune-competent mice. Nat. Med. 22, 306–311.
Schaeffer, M., Hodson, D.J., Lafont, C., and Mollard, P. (2011). Endocrine cells
Virtanen, I., Banerjee, M., Palgi, J., Korsgren, O., Lukinius, A., Thornell, L.-E.,
and blood vessels work in tandem to generate hormone pulses. J. Mol. Endo-
Kikkawa, Y., Sekiguchi, K., Hukkanen, M., Konttinen, Y.T., and Otonkoski, T.
crinol. 47, R59–R66.
(2008). Blood vessels of human islets of Langerhans are surrounded by a dou-
ble basement membrane. Diabetologia 51, 1181–1191.
Shapiro, A.M., Lakey, J.R., Ryan, E.A., Korbutt, G.S., Toth, E., Warnock, G.L.,
Kneteman, N.M., and Rajotte, R.V. (2000). Islet transplantation in seven pa- Vo, A.A., Lukovsky, M., Toyoda, M., Wang, J., Reinsmoen, N.L., Lai, C.-H.,
tients with type 1 diabetes mellitus using a glucocorticoid-free immunosup- Peng, A., Villicana, R., and Jordan, S.C. (2008). Rituximab and intravenous im-
pressive regimen. N. Engl. J. Med. 343, 230–238. mune globulin for desensitization during renal transplantation. N. Engl. J. Med.
359, 242–251.
Shapiro, A.M.J., Pokrywczynska, M., and Ricordi, C. (2017). Clinical pancre-
atic islet transplantation. Nat. Rev. Endocrinol. 13, 268–277. Wang, R.N., and Rosenberg, L. (1999). Maintenance of beta-cell function and
survival following islet isolation requires re-establishment of the islet-matrix
Sherry, N., Hagopian, W., Ludvigsson, J., Jain, S.M., Wahlen, J., Ferry, R.J., relationship. J. Endocrinol. 163, 181–190.
Jr., Bode, B., Aronoff, S., Holland, C., Carlin, D., et al.; Protégé Trial Investiga-
tors (2011). Teplizumab for treatment of type 1 diabetes (Protégé study): 1-year Wells, A.D., Li, X.C., Li, Y., Walsh, M.C., Zheng, X.X., Wu, Z., Nuñez, G., Tang,
results from a randomised, placebo-controlled trial. Lancet 378, 487–497. A., Sayegh, M., Hancock, W.W., et al. (1999). Requirement for T-cell apoptosis

822 Cell Stem Cell 22, June 1, 2018


Cell Stem Cell

Perspective
in the induction of peripheral transplantation tolerance. Nat. Med. 5, Young, H.Y., Zucker, P., Flavell, R.A., Jevnikar, A.M., and Singh, B.
1303–1307. (2004). Characterization of the role of major histocompatibility complex
in type 1 diabetes recurrence after islet transplantation. Transplantation
Wherrett, D.K., Bundy, B., Becker, D.J., DiMeglio, L.A., Gitelman, S.E., Go- 78, 509–515.
land, R., Gottlieb, P.A., Greenbaum, C.J., Herold, K.C., Marks, J.B., et al.;
Type 1 Diabetes TrialNet GAD Study Group (2011). Antigen-based therapy Zhu, J., and Marchant, R.E. (2006). Dendritic saccharide surfactant polymers
with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset as antifouling interface materials to reduce platelet adhesion. Bio-
type 1 diabetes: a randomised double-blind trial. Lancet 378, 319–327. macromolecules 7, 1036–1041.

Wojtusciszyn, A., Armanet, M., Morel, P., Berney, T., and Bosco, D. (2008). In- Zhu, S., Russ, H.A., Wang, X., Zhang, M., Ma, T., Xu, T., Tang, S., Hebrok, M.,
sulin secretion from human beta cells is heterogeneous and dependent on cell- and Ding, S. (2016). Human pancreatic beta-like cells converted from fibro-
to-cell contacts. Diabetologia 51, 1843–1852. blasts. Nat. Commun. 7, 10080.

Cell Stem Cell 22, June 1, 2018 823

You might also like