To investigate whether fecal microbial transplantation (FMT) from either allogenic (individuals with type 1 diabetes and a highly preserved beta cell fraction) or autologous (own) donor, administered every 8 weeks during 6 months through a small…
ID
Source
Brief title
Condition
- Glucose metabolism disorders (incl diabetes mellitus)
Synonym
Research involving
Sponsors and support
Intervention
Outcome measures
Primary outcome
residual beta cell function
Residual beta cell function will be measured by stimulated C-peptide response
upon mixed-meal tolerance (MMTT) area under the curve (AUC0-120min) at 0, 2, 6,
9 and 12 months, using a 2 hour (-10, 0, 15, 30, 45, 60, 90, 120 min) mixed
meal (MMT) test at 6 ml per kg body weight (max 360 ml per MMT of Sustacal
Boost Nutritional Drink, Nestle HS, Switzerland: 33% carbohydrates, 57% fat and
15% protein). An AUC0-120 of plasma C-peptide upon the Boost mixed meal
tolerance test (MMTT)) is then calculated.
Secondary outcome
Changes in immunologic tone
II Immunologic parameters: In fresh whole blood samples, detailed multicolor
flow cytometry is performed to characterize circulating immune cell fractions
and specifically measure T-cell exhaustion. This includes monitoring of general
leukocyte composition (monocyte/T/B/NK), granulocytes (Neu/Eo/Baso),
particularly focusing at changes in the CD4, CD8 T cell and Treg compartments.
Naturally occurring (nTreg) and induced regulatory T cells (tTreg) are analysed
by surface and intracellular staining (CD25, CD127, CD122, FOXP3, IL-10, Ki67,
CTLA-4, GITR, LAG-3, CD49b, ICOS and CD39). Detailed analyses of T-cell subsets
allow quantification of naïve and memory subsets (using CD45RA, CCR7 and CD95),
subsets of antigen-experienced T cells such as Th1, Th2, Th17, Tfh or Trm
(using CXCR5, CCR4, CCR6, CXCR3 and CD103) and T cell exhaustion (using CD57,
PD-1, Tim3 and CD69). This allows definition of more than 100 different cell
subsets, approaching the analyzing resolution of the more expensive and less
sensitive mass-spectrometry (CyTOF). Such analyses provide not only important
information regarding the therapy induced changes but also allow comparisons of
the results with trials testing other therapeutic approaches. We will collect
pax-gene tubes to extract mRNA from whole blood. These measurements will be
performed at the LUMC lab of Prof Roep, who is an expert in blood T cell tests
in autoimmune diseases (15). Buffy coats will be stored for HLA and/or
epigenetic analyses.
We will use RNA seq on whole blood stored in a PAX-gene tube to measure
expression patterns and we will use a machine learning algorithm to pinpoint
which immune pathways are differentially expressed by FMT.
Effect on intestinal gut microbiota composition upon multiple allogenic fecal
infusions
To assess therapy specificity morning stool samples will be collected -6, 0, 6
and 12 months in the study to determine microbiota composition. Samples will be
taken by collection on toilet paper (by patient him/herself wearing gloves),
divided over 3 eppendorfs and directly frozen in fridge at home (-20C). Samples
will be transported to AMC on icepacks. At the AMC, all samples will be stored
at -80C. Fecal analysis will be done by 16s microbiota sequencing at AMC
microbiota core center.
Effect on intestinal gut microbiota metabolites upon multiple allogenic fecal
infusions
To assess the effect of the FMTs on microbial metabolite composition, we will
store plasma obtained by vena puncture also performed for the immunological
analyses, and we will ask participants to collect second void urine samples in
the morning.
Glycemic control and basic biochemistry
To investigate overt effects of the interventions on glycemic control we will
collect fasting blood for determination of glucose, HbA1c, lipid spectrum and
eGFR. We will also read-out participants continuous glucose monitoring device
for their time in range and hyper- and hypo glycemic episodes.
Questionares
At each study visit the following questionnaires will be taken
• Intercurrent illnesses, hypoglycemic episodes, insulin dosages, new
medication
• Hypo-awareness
• Gastro-intestinal complaints
• Dietary intake lists online (via mijn.voedingscentrum.nl/nl/eetmeter)
Background summary
The incidence of Type 1 diabetes mellitus (T1D) has tripled in the last thirty
years, and T1D is associated with a lifelong increase of considerable morbidity
and mortality compared to healthy subjects. In fact, T1D diagnosed in childhood
leads to an almost 20 year loss of life-expectancy, more than most childhood
cancers. Notwithstanding decades of intensive research in animals, the
environmental factors driving T1D are still unknown and therapeutic strategies
have invariably failed to halt disease progression.
As the increased T1D incidence is primarily observed in subjects who are not
genetically predisposed, environmental factors including altered diet,
antibiotic use as well as mode of birth have been suggested to play a role, and
these factors have invariably been linked to changes in the gut microbiome.
Indeed, an altered composition of the fecal microbiota composition was observed
in adolescent T1D patients. Moreover, an increased amount of pathogenic
bacterial species has been observed in fecal samples of T1D patients at the
time of diagnosis. Interestingly, this altered fecal microbiota is already
present before the clinical onset of T1D and is related to islet autoantibodies.
Interestingly, non-obese diabetic (NOD) mouse studies suggested that
interaction of intestinal microbes with the innate immune system is a critical
factor in developing T1DM [16], most likely by inducing an altered T-helper
cell type 17 (Th17) population in the small-intestinal lamina propria. One of
the current hypotheses linking the gut microbiome to immunological tone is
production of microbial metabolites such as the short-chain fatty acids
(SCFAs). Production of these compounds is indeed altered in T1D, and the best
known SCFA butyrate is known to stabilize T-cell function in mice. Furthermore,
irritation of the pancreatic duct by microbiota in the proximal gut may
contribute to beta cell inflammation. By introducing beneficial fecal
microbiota to the proximal gut, the organisms that alter immunological tone and
irritate the pancreatic duct may be attenuated, resulting in improved beta cell
function and restoration. Thus, intestinal microbiota, their metabolites and
their associated gut immune system alterations, may either promote or protect
from beta cell autoimmunity. We hypothesize that if one is able to shape the
(small) intestinal microbiota with fecal microbiota transplantation (FMT) it
may be possible to stabilize or even reverse β-cell destruction, reducing
exogenous insulin needs and subsequently associated complications in T1D.
FMT is a promising treatment for T1D, not only because of potential efficacty,
but also because it is a safe procedure, that in our institute has been
performed >500 times without any serious adverse events. In short, a fecal
microbiota suspension is delivered through a duodenal tube after large bowel
lavage. This procedure is usually repeated 3 times with a 2-month interval.
Using an extensive screening protocol for infectious agents in accordance with
European guidelines, to date no infections attributable to the procedure have
been recorded in our group. The FMT procedure itself is tasteless and odorless
and in general generates few side-effects outside of the discomfort of placing
a duodenal tube.
Based on this notion, we initiated in 2013 a randomized pilot trial using
repetitive donor (healthy donor) vs. autologous (own) FMT on residual β-cell
function in new-onset T1D (DIMID trial, please see attached manuscript). Newly
diagnosed male/female patients with T1D were included and randomized (figure
1a,b). Moreover, healthy aged matched males/females were used as donors.
Surprisingly, autologous FMT in 10 new onset T1D subjects had a significant
(p<0.01) effect on preserving residual β-cell function as determined by
Sustacal Boost (Nestle HS) stimulated C-peptide AUC0-120min response after 12
months, whereas donor FMT in 10 new onset T1D had a less obvious beneficial
effect and showed overall a similar β-cell decline as seen in other trials with
placebo use. We have found several changes induced by both donor and autologous
FMT on gut microbiome composition and identified several bacterial strains and
plasma metabolites and T-cell signatures that predicted response to FMT.
The immunological consequence of flooding the small intestine with large bowel
microbiota by FMT may be an important immunological event. Based on our pilot
data, we therefore formulate a paradigm shift, in which we hypothesize that the
molecular mimicry against microbiome-associated antigens that drives T1D can be
exhausted by challenging the immune system though FMT administered in the
duodenum. Exhausted T-cells are ineffective T-cells that express high levels of
the so-called check-point proteins that inhibit immunological responses. Slow
progression of T1D is linked to more exhausted CD8 cells in infiltrated islets
[23], while increased circulating exhausted T cells predicted response to
anti-CD3 therapy in T1D.
In most people with longstanding T1D the remaining residual β-mass eventually
stabilizes, for reasons that are still not understood [39]. In fact, T1D is
even considered a relapsing remitting-disease, in which β-cell proliferation
increases over time. Therefore, even in longstanding T1D the residual β-cell
function may be improved. We hypothesize that the apparent anti-diabetic effect
of autologous FMT can be improved further if an FMT from individuals with a
proven durable preserved beta cell function is used, as these individuals
presumably carry a gut microbioma that best induces immuno-tolerance.
Therefore, we will compare in long-standing T1D autologous FMT to donor FMT
from T1D with preserved β-cell function, presuming that both treatments improve
residual β-cell function and immune cell activities.
Our aim is to use FMTs to shift the *permanent honeymoon-phase* from a rare to
the default phenotype. This will be, if achieved, a major breakthrough for the
treatment of T1D.
Study objective
To investigate whether fecal microbial transplantation (FMT) from either
allogenic (individuals with type 1 diabetes and a highly preserved beta cell
fraction) or autologous (own) donor, administered every 8 weeks during 6 months
through a small intestinal tube, restores residual beta cell function up until
12 months after intervention in patients with longstanding type 1 diabetes.
Study design
This is a double blind randomised clinical trial. Patients will be treated with
infusion of allogenic or autologous feces by duodenal tube after bowel lavage.
Type 1 diabetes mellitus patients will be randomized to the following 2
treatment arms:
1. Multiple allogenic Type 1 diabetes donor fecal infusions at 0, 8 and 16
weeks.
2. Multiple autologous (own) feces infusions at 0, 8 and 16 weeks.
6 months before initial treatment, at baseline, 6 and 12 months residual beta
cel function, microbial composition, and immune cel function will be
characterised.
Intervention
Patients will be treated with infusion of allogenic or autologous feces by
duodenal tube after bowel lavage.
Type 1 diabetes mellitus patients will be randomized to the following 2
treatment arms:
1. Multiple allogenic Type 1 diabetes donor fecal infusions at 0, 8 and 16
weeks.
2. Multiple autologous (own) feces infusions at 0, 8 and 16 weeks.
Study burden and risks
Participant may benefit in terms of helping to further unravel the relation
between the gut microbiome and residual beta cell function. The DIMID study was
very promising, suggesting that manipulation of the gut microbiome may indeed
preserve the residual beta
cell mass. This study will investigate if FMT is also beneficial in the setting
of longstanding type 1 diabetes. Participants may benefit from an increased
redisual beta cell function, which is associated with a lower risk of diabetic
complications and hypoglycemia. In the long therm, the FMT procedure may be
refined to a probiotic formation or a combination of potent microbial
metabolites and antigens that induce immunotolerance. Therefore, the risks
described below outweigh the potential gain from this study.
For the mixed meal test, morning long- and short acting insulin must be
withheld. This carries a small risk of hypo- and hyperglycemia. The study
participants will be carefully instructed and no major risk is expected in the
context of this procedure, especially given the current use of continuous
glucose-monitoring technologies. The placing of the intravenous cannula in our
study can be an unpleasant experience for the subjects and may result in (self
limiting) bruising.
Gastroscopy is a procedure associated with discomfort, but when participants
are well fasted is very safe. The required fasting is associated with a small
risk of hypoglycemia, for which participants will be adequately instructed. In
the age of continuous glucose monitoring this risk is also very low.
In our centre, FMT procedures have not been associated with major adverse
events, and donors are extensively screened to mitigate the risk of potential
infections by the FMT procedure.
Meibergdreef 9
Amsterdam 1105 AZ
NL
Meibergdreef 9
Amsterdam 1105 AZ
NL
Listed location countries
Age
Inclusion criteria
-Patients with >5 years type 1 diabetes
-Aged 18-65 years
-BMI 18-30 kg/m2
-Male/females
-No concomitant medication except insulin
Exclusion criteria
-Inability to provide written informed consent
-Evidence for absent residual beta cel function (undetectable C-peptide)
-Antibiotics use in the last 3 months and proton-pump inhibitor use
-Evidence for compromised immunity
-Second auto-immune disease (i.e. coeliac disease, hyper- or hypothyroidism,
inflammatory bowel disease)
Design
Recruitment
Followed up by the following (possibly more current) registration
No registrations found.
Other (possibly less up-to-date) registrations in this register
No registrations found.
In other registers
Register | ID |
---|---|
CCMO | NL74454.018.20 |