Proceedings of the 34th Meeting
Working Group on Prolamin Analysis
and Toxicity (PWG)
Edited by Peter Koehler
Esslingen, December 2020
Preface
The 34th meeting of the Working Group on Prolamin Analysis and Toxicity (PWG) was
different from all previous PWG meetings. Planned as a face-to-face-meeting hosted by
Dr. Schaer in Postal, Italy, the global Covid 19 pandemic made this type of meeting
impossible. Due to the flexibility of the hosts Jacqueline Pante and Fabiana Saorin, it
was possible to hold the meeting as an online event with a small core group of the Italian
PWG-members present in Postal. Carlo Catassi was among them and spontaneously
volunteered to be the on-site moderator of the meeting and except from the beginning
and the end of the meeting, he accompanied the participants through the scientific
programme. All other group members and the audience participated online. Apart from
the group members, the audience comprised an invited speaker, guests from academia,
industry, and international coeliac societies. Representatives from cereal starch
producers, producers of gluten-free foods, as well as manufacturers of kits for gluten
analysis participated from industry. In spite of the online format, the audience was very
interested, and the presentations were lively discussed.
Analytical and clinical work in the field of CD, non-coeliac gluten/wheat sensitivity
(NCGS/NCWS), gluten and amylase-trypsin inhibitors done by PWG members were
presented in seven talks and discussed at the two meeting sessions. The symposium “Triggers and drivers of coeliac disease” comprised four presentations of PWG
members and one invited speaker and highlighted the main drivers that are responsible
for triggering CD. In addition, one presentation focussed on regulatory aspects of gluten
analysis and labelling. In particular, the issue of quantitating gluten in fermented foods
such as beer by competitive ELISA was an important part of this talk.
I would like to express my thanks to all participants of the meeting for their active
contributions and the discussions that resulted thereof. I am in particular grateful to
Fabiana Saorin and Jacqueline Pante for their flexibility and dedication, and Carlo
Catassi for being the moderator in Postal. This made the meeting a success in spite of
the general restrictions due to the corona pandemic. Finally, I express my gratitude to
all friends, colleagues, sponsors and participants for their inspiration and ongoing
support of the PWG and the meeting.
Esslingen, December 2020, Peter Koehler
Executive Summary
Twelve presentations covered aspects related to gluten, coeliac disease (CD) and other
relevant hypersensitivities, amylase-trypsin inhibitors (ATI) as well as legal issues.
Most authors have sent abstracts that are compiled in this proceedings book. Starting
with analytical aspects of gluten, the programme included breeding for low occurrence
of CD-active epitopes, and genetic modification to reduce or eliminate ATI from wheat.
Furthermore, therapies of CD were reviewed, the pathomechanism of CD and the
gluten-free diet were covered and reasons for the onset of CD from several perspectives
were presented.
Analytical session
Four presentations were given in this session. One presentation was about the analysis
of gluten to detect coeliac disease-relevant epitopes by LC-MS/MS. Linked to this topic
was a report on activities for producing novel reference materials for gluten analysis. A
main point in this presentation was, that the PWG would take the lead in preparing a
new gliadin reference material, and that an isolated protein preparation is preferred over
flour because of limited stability of flour. Another talk described ongoing research from
the Wageningen group on the generation of wheat with low occurrence of CD-active
epitopes and this included traditional breeding strategies as well as the CRISPR/Cas9
approach. Finally, RNAi and CRISPR/Cas9 approaches were reported to reduce or
eliminate ATI from wheat.
Clinical session
This session included three presentations and started with a comprehensive overview on
approaches for the therapy of CD. The second talk dealt with the immunophenotypic
and functional changes occurring in the gut biopsies of pediatric patients with potentialor
acute CD and showed a direct correlation between the number of TCRγδ+ cells and
the serum levels of anti-TG2 in CD patients. The last presentation of this session was
on the measurement of contaminating gluten in the daily diet of CD children following
a gluten-free diet. It was found that in all investigated children the daily gluten intake
was always well below the safety threshold of 10 mg/day.
Symposium: Triggers and drivers of coeliac disease
Four presentations of the symposium covered the currently relevant triggers of CD
starting with the relevance of specific gluten peptides and continuing with the role of
viruses and bacteria. A very interesting finding is that specific bacteria produce peptides
with amino acid sequences very similar to gluten peptides. The last presentation on T
cell immunology in CD showed gluten-specific, HLA-DQ restricted CD4+ T cells as
the critical checkpoint and effector cell in CD with a concurrent importance in the innate
gluten-induced responses in CD.
Analytical research reports
Analysis of gluten to detect coeliac disease relevant
epitopes
Marie-Christin Lay1, Katharina A. Scherf1,2
1 Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Functional Biopolymer Chemistry, Freising, Germany
2 Karlsruhe Institute of Technology (KIT), Institute of Applied Biosciences, Department of Bioactive and Functional Food Chemistry, Karlsruhe, Germany
Abstract
Coeliac disease (CD) is a chronic immune-mediated inflammatory disease of the small
intestine, triggered by the ingestion of gluten. So far, 38 T-cell epitopes consisting of
nine amino acids each are known that provoke the adaptive immune reaction in CD
patients.
The aim of the project is the development of a comprehensive liquid chromatography
tandem mass spectrometry (LC-MS/MS) method to detect peptides with at least one
CD-active epitope in different wheat flours and products.
Based on the T-cell epitopes, 27 epitopes occurring in wheat proteins were identified by
reversing the transglutaminase-mediated deamidation of specific glutamine residues. In
a second step, the wheat proteins containing these epitopes were identified.
Isolated wheat gluten samples and wheat flours were prepared according to a bottom-up
proteomics workflow using chymotrypsin and trypsin for enzymatic digestion. The
generated peptides were analysed with a tripleTOF-LC-MS/MS system. Based on the
about 1900 identified peptides containing at least one CD-active epitope, marker
peptides were selected. These marker peptides were analysed with a QTRAP-LCMS/
MS system in the flours of ten different common wheat cultivars and one
commercial wheat flour.
The first results of the relative quantification of the marker peptides in the different
common wheat flours showed differences between the samples. Especially the cultivar
Winnetou showed higher amounts of peptides belonging to the -gliadin proteins
compared to the other cultivars. The highest relative proportion of the peptides belonged
to peptides of the -gliadin fraction. To interpret these results, it should be taken into
account that also the ratio of the -gliadin fraction within gluten proteins and of the -
gliadin peptides within the marker peptides are the highest.
The next step of the project will be the absolute quantification of the marker peptides in
a large sample set of different common wheat, emmer, einkorn, spelt and durum wheat
cultivars.
Wheat lines with specific ATI genes silenced by RNAi
and CRISPR-Cas9 for the understanding of their role in
Non Celiac Wheat Sensitivity
Stefania Masci1, Francesco Camerlengo1, Stefano D’Amico2, Sandra Denery-Papini3,
Angela Doherty4, Arianna Frittelli1, Shahidul Islam5, Raviraj M. Kalunke1, Domenico
Lafiandra1, Colette Larré3, Roberta Lupi3, Wujun Ma5, Damiano Martignago4,
Francesco Sestili1, Caroline Sparks4, Silvio Tundo1
1 DAFNE, University of Tuscia, 01100 Viterbo, Italy
2 IANF, AGES, 1220 Vienna, Austria
3 INRAE UR1268 BIA, 44000 Nantes, France
4 Rothamsted Research, Harpenden, United Kingdom
5 Australia China Centre for Wheat Improvement, Murdoch University, Australia
Abstract
Non Celiac Wheat Sensitivity (NCWS) is gaining a great importance, due to its
diffusion, but the real culprit has not been ascertained yet. Among the most likely
candidates, there are alpha-amylase/trypsin inhibitors (ATI) that are involved also in
some respiratory allergies. These latter polypeptides are a group of exogenous protease
inhibitors, which are encoded by a multigene family dispersed over several
chromosomes in durum and bread wheat. With the aim of obtaining wheat genotypes
showing a lower amount of these proteins, we have pursued this goal by using two
methods, RNAi and CRISPR-Cas9 silencing. Silencing by RNAi interference that is
considered a totally transgenic technique has been carried out on the bread wheat
cultivar Bobwhite on the three ATI genes CM3, CM16 and 0.28, whereas the durum
wheat cultivar Svevo has been silenced by CRISPR/Cas9 by using a multiplexing
strategy to edit the two ATI genes CM3 and CM16, and a marker-free approach, that
makes these latter lines potentially non-transgenic.
RNAi silenced lines do not show differences in terms of yield, have an effective decrease
of the target genes, but also a range of pleiotropic effects, including a higher trypsin
inhibition and a strong decrease in HMW-GS accumulation. They generate a lower
reaction when tested with sera of patients allergic to wheat, accounting for the important
role of the three target proteins in wheat allergies.
As regards the genome-edited lines, they also show absence of the target genes, but also
the activation of the ATI 0.28 pseudogene present in durum wheat, as a pleiotropic
effect.
The development of wheats accumulating a lower amount of ATI, not only allows to
use them as a basis for the production of varieties with a lower impact on adverse
reaction, but also to test if these proteins are actually implicated in those pathologies for
which the triggering factor has not been established yet, as is the case for NCWS.
Wheat modified for low occurrence of CD epitopes
Marinus J. M. (René) Smulders
Plant Breeding, Wageningen University & Research, Wageningen, The Netherlands
Abstract
In wheat, coeliac disease (CD) epitopes occur mostly in gliadins, while the baking
quality is determined predominantly by glutenins. Thus, removing and mutating gliadins
can be used to lower the immunogenicity of wheat. Nevertheless, plant breeding cannot
generate bread wheat that is safe for coeliac disease patients while retaining baking
quality solely by combining natural or randomly induced mutations in gliadins and
glutenins, due to the large number of genes in these gene families that have to be
modified or removed, and because these genes are closely linked in clustered loci on the
three genomes of bread wheat. Combining recently developed biotechnological
approaches with classical breeding now offers the opportunity to change this situation.
A good starting point would be to use lines with deletions of some of the major gliadin
loci. Deletion lines already exist, but lines with smaller, more focussed chromosomal
deletions may be generated using -irradiation or fast neutrons, and these may have
improved performance as a crop. CD-safe barley has been produced by combining such
chromosomal deletions.
Next, various approaches may be used to lower the expression of the other gliadin genes.
One interesting approach is to introduce the recessive, low-prolamin mutation lys3a.
Other approached that may be used are RNAi of gliadin gene families, or RNAi of
DEMETER, a gene necessary for activation of storage protein genes during wheat
endosperm development. However, as the RNAi construct must remain present, this
classifies as a GM approach.
Subsequently, the CD epitopes in the remaining gliadins that are expressed may be
modified using gene editing with CRISPR/Cas, or by base editing. As the major epitopes
are well known, the targets are clear. The challenge is to deal with the multiple loci that
need to be modified, but recent improvements in the regeneration of wheat make this
more realistic. Selection and screening must initially be done at DNA and protein level
and confirmed with T cell tests, etc. However, once a clustered locus on a chromosome
is devoid of major epitopes, it can be combined with other hypoallergenic loci through
regular crossing and selecting, and these loci may be tracked in a breeding programme
using linked molecular markers.
Thus, in the countries where targeted mutagenesis using gene editing is not considered
as genetic modification, developing coeliac-safe wheat now appears doable. An
intermediate product will be low-gluten wheat varieties.
Looking beyond PWG-gliadin at future reference
materials for gluten
Katharina Scherf
Karlsruhe Institute of Technology (KIT), Institute of Applied Biosciences, Department
of Bioactive and Functional Food Chemistry, Karlsruhe, Germany
Abstract
Reference materials are essential to validate analytical methods, calibrate instruments,
verify laboratory performance, ensure quality control and estimate uncertainty. There is
no certified reference material available for gluten, mainly because its protein
composition is very complex and depends on genetic and environmental variability.
Prolamin Working Group (PWG)-gliadin was isolated from a mixture of 28
representative European wheat varieties and the material is purified, homogeneous,
completely soluble in 60% ethanol, stable, well-characterised and used to calibrate
ELISA test kits and other methods for gluten detection. However, as the supply is
limited, a new reference material for gluten needs to be developed.
The first discussions among the gluten reference material team concluded that a blend
of five or more varieties of wheat, rye or barley from different continents appears to be
most suitable to cover the genetic variability while evening out year-to-year variations
of protein content and composition. The varieties should be widely used and milled to
white flour for reasons of stability. Concerning long-term stability, the PWG continues
to support the use of protein isolates, because PWG-gliadin has been stable for almost
20 years now when kept frozen at -80 °C. Lab-scale experiments using five varieties of
wheat including Akteur (Germany), Carberry (Canada), Mv Magvas (Hungary), Yitpi
(Australia) and Yumai-34 (China), as well as their blend, confirmed that gluten and
gliadin isolates can be prepared while keeping the original protein composition of the
flour. Further experiments based on ELISA, gel electrophoretic and chromatographic
techniques are currently underway to select five suitable rye and barley varieties each
from a collection of 123 barley and 57 rye varieties.
Based on its expertise and previous work with the PWG-gliadin reference material, the
PWG will take the lead in the production of a new reference material for gluten and
provide the new material to all stakeholders. The PWG agreed that a new gliadin isolate
seems to be most appropriate for various applications such as clinical research and
compliance monitoring of gluten-free products.
5. Clinical research reports
Update on clinical studies for the pharmacological
treatment of coeliac disease
Detlef Schuppan1,2
1 Institute of Translational Immunology, Center for Celiac and Small Intestinal
Diseases, Food Allergy and Autoimmunity, University Medical Center, Mainz,
Germany
2 Div. of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical
School, Boston, USA
Abstract
Coeliac disease (CD) is a small intestinal inflammatory condition that affects up to 2%
of most populations worldwide (1). CD is triggered and maintained by the ingestion of
gluten from wheat and related grains and occurs only in carriers of the human leukocyte
antigens (HLA) DQ2 or DQ8, a necessary but not sufficient genetic precondition for the
manifestation of CD. Classical symptoms of CD include diarrhea, weight loss, and
malnutrition, but CD often manifests with nonspecific or atypical symptoms, or
combined with autoimmune diseases that share the same major genetic predisposition
as CD, HLA DQ2 or DQ8 (2-5).
The currently only treatment for CD is the life-long adherence to a strict gluten-free diet
(GFD), i.e., complete avoidance of even traces of gluten in the diet, but maintaining the
GFD poses significant practical and social challenges. Moreover, some patients are
exquisitely sensitive to hidden traces of gluten, and up to 50% of celiacs do not show
complete mucosal healing after one year of the GFD (6,7). Therefore, there is a need for
an efficient (supportive) pharmacological therapy as add-on to the GFD to prevent
complications, such as consequences of malabsorption and possibly autoimmunity and
malignancy that can be due to continuous (minor) gluten ingestion (8).
Several pharmacological therapies to prevent gluten induced mucosal damage in
patients with CD, and some made it towards phase 1b-3 clinical dose finding and
efficacy studies, and the best study design for phase 2 is the gluten challenge in patients
in remission and histological readout, to be complemented by patient related outcome
measures (9). The most prominent or promising therapeutic approaches are shortly
discussed:
1. Oral proteases that cleave immunogenic gluten peptides that otherwise escape
intestinal digestion: While a phase 2a study showed promise for the combination
of the barley germinating seed glutamine specific with a microbial proline specific
endoprotease (Alvine), the real life phase 3 study failed (10,11). Currently, a phase
2 study with a synthetic glutenase (Kuma030, Takeda), with a more than 100fold higher in vitro activity to degrade immunogenic gluten peptides, is ongoing (12).
A major challenge for enzyme therapy remains: to secure rapid and complete
enzymatic digestion of immunogenic gluten peptides that are embedded in a
complex food matrix. This must occur within the stomach and proximal small
intestine before these peptides reach the mucosal immune system of the small
intestine (13).
2. Blocking antibody to IL-15: This based on a prominent role of epithelial and
immune cell derived IL-15 as a driver of intestinal inflammation in CD. However,
using the antibody AMG 714 (Amgen/Provention Bio) a phase 2 study showed no
protection from mucosal damage, although patient symptoms improved (14). An
IL-15 antibody that has a much higher potency to block the IL-15 receptor
(Calypso) will be tested in a phase 1b study in 2021.
3. Tolerizing therapies: The aim is to induce tolerance to gluten in CD patients. While
a long lasting development of a tolerizing vaccine, utilizing 3 major gluten
antigenic epitopes finally failed (Nexvac (15)], the encapsulation of immunogenic
gluten in PLGA nanoparticles that target myeloid cells to induce intestinal
tolerance showed efficacy in a CD mouse model in vitro (16), and signs of efficacy
in a phase 1b study in patients (TAK-101, Takeda). Another approach employs
immunogenic gluten peptide-loaded and liver sinusoidal endothelial cell targeted
nanoparticles that effectively dampened experimental multiple sclerosis. This has
effectively suppressed the development of Multiple Sclerosis in Mice (17). A
phase 1b study is planned for 2021.
4. Transglutaminase (TG) inhibitors: The aim is to block the conversion of gluten
peptides that exhibit with low immunogenicity to deamidated gluten peptides that
are optimally presented on HLA-DQ2 or -DQ8 to induce intestinal T cell activation
and inflammation (18-20). This approach has shown proof-of-concept in in vitro
systems and in an IL-15 transgenic celiac mouse model, where a general
transglutaminase inhibitor attenuated gluten-induced T cell activation (21). A
phase 2 clinical study using a highly specific oral inhibitor of TG2, the CD-specific
transglutaminase, in 160 CD patients in remission and challenged with gluten for
6 weeks will be published soon.
In conclusion, several pharmacological studies aiming at protection of CD patients from
gluten induced intestinal inflammation have been developed and are currently being
assessed in clinical efficacy studies. CD therapeutics have become a “hot field” in view
of, e.g., the saturated area of therapeutics for chronic inflammatory bowel disease. The
aim is to abolish the detrimental effect of up to ~3-5 grams of daily gluten exposure,
equivalent to 15-30% of normal gluten consumption. Drugs that mainly act in the
intestinal mucosa, protecting it from immune activation, and tolerizing approaches that
may provide longterm protection appear most attractive.
References
1. Singh P, Arora A, Strand TA, et al. Global Prevalence of Celiac Disease: Systematic
Review and Meta-analysis. Clin Gastroenterol Hepatol 2018; 16: 823-836 e2.
2. Schuppan D, Junker Y, Barisani D. Celiac disease: from pathogenesis to novel
therapies. Gastroenterology 2009; 137: 1912-1933.
3. Lundin KE, Sollid LM. Advances in coeliac disease. Curr Opin Gastroenterol 2014;
30: 154-162.
4. Lebwohl B, Sanders DS, Green PHR. Coeliac disease. Lancet 2018; 391: 70-81.
5. Kahaly GJ, Frommer L, Schuppan D. Celiac disease and endocrine autoimmunity -
the genetic link. Autoimmun Rev 2018; 17: 1169-1175.
6. Daveson AJM, Popp A, Taavela J, et al. Baseline quantitative histology in
therapeutics trials reveals villus atrophy in most patients with coeliac disease who
appear well controlled on gluten‐free diet. GastroHep 2020; 2: 22-30.
7. Syage JA, Kelly CP, Dickason MA, et al. Determination of gluten consumption in
celiac disease patients on a gluten-free diet. Am J Clin Nutr 2018; 107: 201-207.
8. Leffler D, Kupfer SS, Lebwohl B, et al. Development of Celiac Disease
Therapeutics: Report of the Third Gastroenterology Regulatory Endpoints and
Advancement of Therapeutics Workshop. Gastroenterology 2016; 151: 407-411.
9. Ludvigsson JF, Ciacci C, Green PH, et al. Outcome measures in coeliac disease
trials: the Tampere recommendations. Gut 2018; 67: 1410-1424.
10. Lähdeaho ML, Kaukinen K, Laurila K, et al. Glutenase ALV003 attenuates gluteninduced
mucosal injury in patients with celiac disease. Gastroenterology 2014; 146:
1649-1658.
11. Murray JA, Kelly CP, Green PHR, et al. No Difference Between Latiglutenase and
Placebo in Reducing Villous Atrophy or Improving Symptoms in Patients With
Symptomatic Celiac Disease. Gastroenterology 2017; 152: 787-798.
12. Wolf C Siegel JB Tinberg C, et al. Engineering of Kuma030: A Gliadin Peptidase
that rapidly degrades immunogenic gliadin peptides in gastric conditions. J Am
Chem Soc 2015; 137: 13106–13113.
13. Wei G, Helmerhorst EJ, Darwish G, et al. Gluten Degrading Enzymes for Treatment
of Celiac Disease. Nutrients 2020; 12(7): 2095.
14. Lähdeaho ML, Scheinin M, Vuotikka P, et al. Safety and efficacy of AMG 714 in
adults with coeliac disease exposed to gluten challenge: a phase 2a, randomised,
double-blind, placebo-controlled study. Lancet Gastroenterol Hepatol 2019; 4: 948-
959.
15. Truitt KE, Daveson AJM, Ee HC, et al. Randomised clinical trial: a placebocontrolled
study of subcutaneous or intradermal NEXVAX2, an investigational immunomodulatory peptide therapy for coeliac disease. Aliment Pharmacol Ther
2019; 50: 547-555.
16. Freitag TL, Podojil JR, Pearson RM, et al. Gliadin Nanoparticles Induce Immune
Tolerance to Gliadin in Mouse Models of Celiac Disease. Gastroenterology 2020;
158: 1667-1681.e12.
17. Carambia A, Freund B, Schwinge D, et al. Nanoparticle-based autoantigen delivery
to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity
in mice. J Hepatol 2015; 62: 1349-1356.
18. Dieterich W, Ehnis T, Bauer M, et al. Identification of tissue transglutaminase as the
autoantigen of celiac disease. Nat Med 1997; 3: 797-801.
19. Molberg O, McAdam SN, Korner R, et al. Tissue transglutaminase selectively
modifies gliadin peptides that are recognized by gut-derived T cells in celiac disease.
Nat Med 1998; 4: 713-717.
20. van de Wal Y, Kooy Y, van Veelen P, et al. Selective deamidation by tissue
transglutaminase strongly enhances gliadin-specific T cell reactivity. J Immunol
1998; 161: 1585-1588.
21. Abadie V, Kim SM, Lejeune T, et al. IL-15, gluten and HLA-DQ8 drive tissue
destruction in coeliac disease. Nature 2020; 578: 600-604.
Densities of IL4+ and TCRγδ+ T cell subsets as
biomarkers of intestinal mucosa damage in coeliac
disease
Serena Vitale1, Stefania Picascia1, Mariantonia Maglio2, Renata Auricchio2,
Riccardo Troncone1,2, Carmen Gianfrani1,2
1 Institute of Biochemistry and Cell Biology - CNR, Naples, Italy
2 Department of Translational Medicine & European Laboratory for the Investigation
of Food-Induced Diseases, University Federico II, Naples, Italy
Abstract
Coeliac disease (CD) comprises several clinical conditions, all characterised by the
presence of HLA-risk alleles and specific antibodies. Furthermore, a large spectrum of
intestinal lesions is reported that ranges from morphologically normal mucosa,
condition known as potential-CD, to villous atrophy, typical of acute disease. It is
envisaged that the disruption of the immunological balance is responsible of normal to
atrophic mucosa transition. As the immune mechanisms underlying the CD natural
history are not completely elucidated, we investigated the immunophenotypic and
functional changes occurring in the gut biopsies of pediatric patients with potential- or
acute-CD. Forty-seven young subjects underwent endoscopy for suspicion of CD
disease. Nineteen children had a diagnosis of acute CD (CD, mean age 5.9 yrs), 16 had
normal mucosa but were positive for anti-tissue transglutaminase (TG2) antibody,
(potential-CD, mean age 8.7 yrs), and 12 non-coeliac control subjects (HC, mean age
6.3 yrs). Cell phenotype (CD3, CD4, CD8, and TCRγδ+ T cells) and cytokine
production (INF-γ, IL4, IL21, IL17) were analysed by flow cytometry either in ex-vivo
mucosal cells or in short-term T-cell lines.
T cells bearing the TCRγδ (TCRγδ+ cells) were markedly increased in children with
overt-CD compared to potential-CD or HC (p<0.05). In contrast, T cell producing IL4
(IL4+ cells) were significantly increased in potential-CD and HC (p<0.05). An indirect
correlation between the frequency of TCRγδ+ and IL4+ cells was observed in all
children enrolled (r=-0.5141, p=0.0013). A direct correlation was found between the
number of TCRγδ+ cells and the serum levels of anti-TG2 in CD patients (both overtand
potential-CD), (r=0.4635, p=0.0086). Conversely, IL4+ cells indirectly correlated
with the anti-TG2 titers (r=-0.5863, p=0.0013). In conclusion, the transition to villous
atrophy in CD patients is characterised by the expansion of TCRγδ+ cells concomitantly
with the disappearance of IL4+ cells in gut biopsies. These findings, along with the
indirect correlation between the anti-TG2 titers and IL4+ cell frequency, suggest that a
shift from Th2 to Th1 phenotype of mucosa infiltrating T lymphocytes occurs in the
transition from potential- to acute-CD.
Further studies are required to validate the IL4+ and TCRγδ+ cells as biomarkers of the
different CD forms. If these pilot findings will be confirmed, the combined detection of
these two cell subsets by flow cytometry could support the diagnosis of CD, mainly in
case of histological pitfall or borderline value of CD-serology. Furthermore, the
combined assessment of these cellular biomarkers could also provide a laboratory tool
for the clinical management of potential-CD patients.
Contamination of gluten in the gluten-free diet: a
quantitative study in children with coeliac disease
Chiara Monachesi, Anil K Verma, Giulia Naspi Catassi, Tiziana Galeazzi, Elisa
Franceschini, Valentina Perticaroli, Elena Lionetti, Carlo Catassi
Department of Pediatrics, Università Politecnica delle Marche, Ancona, Italy
Abstract
Background: A strict gluten-free diet (GFD) is notoriously difficult to maintain.
Protracted ingestion of gluten traces (10-50 mg/day) is sufficient to cause significant
damage in the architecture of the small intestinal mucosa in patients on treatment for
coeliac disease (CD). Only few data are available on the daily intake of contaminating
gluten in treated CD patients.
Objective: The aim of this study was to directly measure the level of contaminating
gluten in the daily diet of CD children following a GFD.
Design: From April 2019 to December 2019, consecutive CD children (2-18 years old)
on GFD for ≥ 6 months were offered to participate in the study. Patients and their
caregivers were invited to provide a representative portion (about 10 g) of all meals
consumed during a 24-hour period. The participants were requested to weight all
ingested food and report items in a 24-hour food diary and to document brand, and
ingredients. Gluten content of all food samples was quantified by R5 sandwich enzymelinked
immunosorbent assay method.
Results: Overall, 12/448 (2.73%) food samples contained detectable gluten
contamination; of them, 11 (92%) contained 5-20 part per million (ppm) and 1 (8%) >20
ppm. The 12 contaminated food samples belonged to 5 of the 69 enrolled patients. In all
investigated children the daily gluten intake was always well below the safety threshold
of 10 mg/day.
Conclusions: The present findings suggest that in a country characterised by high CD
awareness, the daily unintended exposure to gluten of treated CD children is very low;
reassuringly, the presence of gluten traces did not lead to exceed the tolerable threshold
of 10 mg/day of gluten intake in the GFD. These favourable results may be explained
by several factors: (a) pediatric age of investigated subjects. The diet of children is more
easily and fully controlled by the caregivers; (b) inclusion of highly compliant patients
who are regularly seen at the Coeliac Clinic; (c) generalised conformity of GF products
marketed in Italy with the International regulations for labelled gluten-free food; (d)
high level of awareness of the requirement of the GFD by the general population in Italy,
particularly due to the national Coeliac Protection law (n.123/2005) and the pro-active
role of the Italian Coeliac Association.
Symposium: Triggers and drivers of coeliac
disease
Gliadin and its peptide 31-43 as proinflammatory molecules
Maria Vittoria Barone1,2, Fernando G. Chirdo3, Salvatore Auricchio1,2, Riccardo
Troncone1,2
1 European Laboratory for the Investigation of Food Induced Diseases (ELFID),
University Federico II, Naples, Italy
2 Department of Translational Medical Science, University Federico II, Naples, Italy
3 Instituto de Estudios Inmunológicos y Fisiopatológicos - IIFP (UNLP-CONICET).
Departamento de Ciencias Biológicas. Facultad de Ciencias Exactas, La Plata,
Argentina
Abstract
Coeliac disease (CD) is an immune-mediated enteropathy triggered in genetically
susceptible individuals by a group of wheat proteins (commonly called gluten) and
related prolamins from toxic cereals. The HLA-restricted gliadin-specific intestinal T
cell response plays a central role in the pathogenesis of CD. A central question remains
unanswered, why a pro-inflammatory T cell response is generated instead of a regulatory
response, which normally promotes oral tolerance to dietary protein antigens. In an
inflamed environment enriched in cytokines such as IL-15 or type I interferons, T cells
tend to acquire a pro-inflammatory phenotype. Mice-based studies as well as
epidemiological data have suggested viral infections to create such an environment.
However, a number of other factors may contribute to the generation of a “sterile” inflammation. Most of the evidence point to gliadin itself and in particular, a peptide
from the N-terminal portion of -gliadin, named peptide p31-43.
The p31-43 peptide is part of the p31-55 peptide from α-gliadins that remains undigested
for a long time, and can be present in the small intestine after ingestion of a glutencontaining
diet. Different biophysical methods and molecular dynamic simulations have
shown that p31-43 spontaneously forms oligomeric nanostructures. Experimental
approaches using in vitro assays, mouse models, and human duodenal tissues have
shown that p31-43 is able to induce different forms of cellular stress by driving multiple
inflammatory pathways. Increased proliferative activity of the epithelial cells in the
crypts, enterocyte stress, activation of TG2, induction of Ca2+, IL-15, and NFB
signalling, inhibition of CFTR and activation of the inflammasome platform are some
of the biological effects of p31-43.
One possible mechanism candidate as responsible for this condition of inflammatory
environment is the alteration of vesicular trafficking. Interestingly in fact, inducing a delay of the endocytic trafficking by silencing the HRS protein, produces at cellular
level the same alterations that p31-43 does. Another important question to be answered
is why p31-43 affects particularly CD patients? The answer may reside in the
constitutive alterations present in coeliac subjects. These involve several biological
pathways, such as signalling/proliferation, stress/innate immune response and
inflammation, ultimately due to the alterations of vesicular trafficking.
In conclusion, CD is the prototype of a chronic inflammatory disorder induced by
dietary components. p31-43 comes into the spotlight as an important player in CD
pathogenesis. Its particular conformation and its ability to induce different forms of
cellular stress drive multiple inflammatory pathways, which, in the presence of
appropriate susceptibility and environmental factors, may act together to drive the
disease.
The role of viruses as triggers of coeliac disease
Valentina Discepolo
Department of Translational Medical Science, University Federico II, Naples, Italy
No abstract provided.
The role of bacteria as triggers of coeliac disease
Frits Koning
Leiden University Medical Centre, Department of Immunohaematology and
Bloodbank, Leiden, The Netherlands
No abstract provided.
T cell immunology in coeliac disease (the Oslo experience)
Knut E. A. Lundin
The KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
Dept of Gastroenterology, Oslo University Hospital, Oslo, Norway
Abstract
The role of the adaptive immune system, and particularly the recognition of gluten by
CD4+ T cells from the gut, has been the focus of the Oslo group under the leadership of
professor Ludvig M. Sollid for decades (1). This summary gives a short overview on
some aspects. In 1989 Sollid reported that «all» coeliacs expressed certain HLA-DQ
molecules, directly pointing to the possibility that gluten-reactive T cells were involved
(2). The presence of such T cells was published some very few years later, a major
break-through in this research field (3, 4). The next milestone was reached when the
immunogenic peptides were defined and we and others in the late 1990’s showed that
they were modified by the enzyme Transglutaminase 2 (5). Similar observations were
soon reported by others (6, 7). The anti-gluten T cell response is «strong», the frequency
of gluten-specific T cells in the gut is in the range 0,5 – 1,8% and the same cell persist
for decades (8, 9). Direct demonstration of such CD4+, gluten specific T cells by HLADQ:
gluten tetramers in the blood may be used as a diagnostic test (10) and
immunobiological studies have shown how they are key drivers of the
immunopathology in this disease (11). Importantly, gluten challenge experiments show
rapid activation of the gluten-specific T cells in all patients (12), and secretion of the T
cell cytokine Interleukin-2 correlate very well with symptoms like nausea and vomiting
after challenge (13). Taken together, these findings all points to the gluten-specific,
HLA-DQ restricted CD4+ T cells as the critical checkpoint and effector cell in coeliac
disease, and that they control much of the innate gluten-induced responses in coeliac
disease as well.
References
1. Lundin KE, Qiao SW, Snir O, Sollid LM. Coeliac disease - from genetic and
immunological studies to clinical applications. Scandinavian journal of
gastroenterology. 2015;50(6):708-717.
2. Sollid LM, Markussen G, Ek J, Gjerde H, Vartdal F, Thorsby E. Evidence for a
primary association of celiac disease to a particular HLA-DQ alpha/beta
heterodimer. The Journal of experimental medicine. 1989;169(1):345-350.
3. Lundin KE, Scott H, Hansen T, Paulsen G, Halstensen TS, Fausa O, et al. Gliadinspecific,
HLA-DQ(alpha 1*0501,beta 1*0201) restricted T cells isolated from the small intestinal mucosa of celiac disease patients. The Journal of experimental
medicine. 1993;178(1):187-196.
4. Lundin KE, Scott H, Fausa O, Thorsby E, Sollid LM. T cells from the small intestinal
mucosa of a DR4, DQ7/DR4, DQ8 celiac disease patient preferentially recognize
gliadin when presented by DQ8. Human immunology. 1994;41(4):285-291.
5. Molberg O, McAdam SN, Korner R, Quarsten H, Kristiansen C, Madsen L, et al.
Tissue transglutaminase selectively modifies gliadin peptides that are recognized by
gut-derived T cells in celiac disease. Nature medicine. 1998;4(6):713-717.
6. Vader W, Kooy Y, Van Veelen P, De Ru A, Harris D, Benckhuijsen W, et al. The
gluten response in children with celiac disease is directed toward multiple gliadin
and glutenin peptides. Gastroenterology. 2002;122(7):1729-1737.
7. Anderson RP, Degano P, Godkin AJ, Jewell DP, Hill AV. In vivo antigen challenge
in celiac disease identifies a single transglutaminase-modified peptide as the
dominant A-gliadin T-cell epitope. Nature medicine. 2000;6(3):337-342.
8. Bodd M, Raki M, Bergseng E, Jahnsen J, Lundin KE, Sollid LM. Direct cloning and
tetramer staining to measure the frequency of intestinal gluten-reactive T cells in
celiac disease. European journal of immunology. 2013;43(10):2605-2612.
9. Risnes LF, Christophersen A, Dahal-Koirala S, Neumann RS, Sandve GK, Sarna
VK, et al. Disease-driving CD4+ T cell clonotypes persist for decades in celiac
disease. The Journal of clinical investigation. 2018;128(6):2642-2650.
10. Sarna VK, Lundin KEA, Morkrid L, Qiao SW, Sollid LM, Christophersen A. HLADQ-
Gluten Tetramer Blood Test Accurately Identifies Patients With and Without
Celiac Disease in Absence of Gluten Consumption. Gastroenterology.
2018;154(4):886-96 e6.
11. Christophersen A, Lund EG, Snir O, Sola E, Kanduri C, Dahal-Koirala S, et al.
Distinct phenotype of CD4(+) T cells driving celiac disease identified in multiple
autoimmune conditions. Nature medicine. 2019;25(5):734-737.
12. Zuhlke S, Risnes LF, Dahal-Koirala S, Christophersen A, Sollid LM, Lundin KE.
CD38 expression on gluten-specific T cells is a robust marker of gluten re-exposure
in coeliac disease. United European gastroenterology journal. 2019;7(10):1337-
1344.
13. Goel G, Tye-Din JA, Qiao SW, Russell AK, Mayassi T, Ciszewski C, et al. Cytokine
release and gastrointestinal symptoms after gluten challenge in celiac disease. Sci
Adv. 2019;5(8):eaaw7756.
Statements on current developments
concerning gluten analysis, clinical and legal
aspects
Update on regulatory issues of gluten
Hertha Deutsch
AOECS Codex Delegate, Austrian Coeliac Society, Vienna
Abstract
AOECS, the Association Of European Coeliac Societies, has Observer status in the
Codex Alimentarius Commission since 1992. In the past months, some items important
for coeliacs regarding the gluten-free diet were discussed in the Codex Committee on
Food Hygiene (CCFH) in November 2019 and in the Codex Committee on Nutrition
and Foods for Special Dietary Uses (CCNFSDU) in November 2019. Because of the
Covid-19 situation, the Codex Alimentarius Commission (CAC) was hold in virtual
sessions in several days in September, October and November 2020. The reports of the
sessions are published on the Codex website www.fao.org. A short summary follows:
CCFH: The Committee agreed to forward the Draft Code of Practice on Food Allergen
Management for Food Business Operators (COP) for adoption at Step 8 (Appendix II,
page 24 - 44 of the report); to inform the Codex Committee on Food Labelling (CCFL)
of the status of the work; and that the COP could be revised upon completion of the
work on precautionary allergen labelling in CCFL and advice from FAO/WHO.
CCNFSDU: At the CCMAS session in May 2019, CCMAS agreed to refer the AACCI
proposal to delete „Gluten-free Foods“ in the Commodity of the Codex Standard
234 - 199 and replace it with „Corn- and Rice-Based Gluten-Free Foods“ and „Oat-
Based Gluten-Free Foods“ to CCNFSDU for consideration. CCNFSDU noted that it
was premature to consider this proposal as research is still ongoing to determine the
most appropriate method for determination of gluten and agreed to wait for the
completion of ring trial tests and to consider this matter at a future date when more
information became available. Furtheron, the Committee agreed to align section 5.2 of
the Standard for Foods for Special Dietary Use for Persons Intolerant to Gluten (CXS
118 - 1979) with the wording from the Procedural Manual. That means that any method
of analysis will only be contained in the Codex Standard 234 - 1999 and not in individual
Standards. The Committee agreed to submit this editorial amendment to CXS 118-1979
to CAC for adoption.
CAC: The Draft Code of Practice on Food Allergen Management for Food Business
Operators and the editorial amendment to CXS 118-1979 were adopted.
In the USA, the FDA issued a final rule on the gluten-free labeling of fermented or
hydrolyzed foods on August 12, 2020. The PWG statement to this rule is available.
Perspectives and action plan of the PWG
Peter Koehler
Biotask AG, Esslingen, Germany
The Prolamin Working Group executive meeting and joint discussion held on 15
October 2020, led to the decisions and statements outlined below.
Action plan
I. Analytical
The PWG gliadin reference material is available from Arbeitsgemeinschaft
Getreideforschung e.V. (Association of Cereal Research), Mr. Tobias
Schumacher, Schuetzenberg 10, 32756 Detmold, Germany, E-mail: info@agfdetmold.
de. The PWG has set the price for one batch (100 mg) to 150 Euro.
The collaborative study Gluten in a broad range of food ingredients and food
products by Quantitative Enzyme Immunoassay R-Biopharm RIDASCREEN® Gliadin Test Kit supervised by Katharina Scherf was carried out in 2020.
The PWG will take the lead in preparing new gliadin reference material. An
isolated protein preparation is preferred over flour because of limited stability of
flour. Possible production facilities for the reproduction of PWG-gliadin
reference material will be identified.
II. Clinical
The PWG keeps considering becoming a working group under the umbrella of
the International Society For The Study Of Celiac Disease
III. Members, Policy
Stefania Masci, University of Tuscia, Viterbo, Italy is a new member of the
group.
Olivier Tranquet, INRAE, Nantes, France left the group and will be replaced by
Sandra Denery from the same institute.
Bob Anderson, Wesley Medical Research Ltd, Brisbane, Australia has been
suggested as a new member of the group.
A joint position paper Recent Progress and Recommendations on Celiac Disease
From the Working Group on Prolamin Analysis and Toxicity has been published
Open Access in March 2020 (doi: 10.3389/fnut.2020.00029).
A joint opinion letter Statement of the Prolamin Working Group on the
Determination of Gluten in Fermented Foods Containing Partially Hydrolyzed
Gluten has been published Open Access in December 2020 (doi:
10.3389/fnut.2020.626712).
An Open Access publication with the working title Update on gluten analysis
and considerations on the effect of low gluten doses on intestinal health has been
suggested by Carlo Catassi.
Proceedings of this meeting will be available free of charge in electronic form
from the PWG website (http://www.wgpat.com).
Next meeting: 2021
We are very pleased to announce the venue for our meeting in 2021:
Wageningen, The Netherlands
Host:
René Smulders, Twan America, Ingrid van der Meer, Peter Weegels
Wageningen University & Research
E-mail: rene.smulders@wur.nl
Time: 28 - 30 October 2021 (preliminary)
Focus of the meeting:
Gluten analysis and clinical effects of low gluten doses
The format of the meeting will depend on the global Corona pandemic
situation. At the moment, no decision has been taken.
The invitation and registration deadline will be sent in summer 2021.
Very special thanks to the host of this kind invitation!
|
|
List of Participants
GROUP MEMBERS
Prof. Dr. Carlo Catassi
Università Politecnica delle Marche
Department of Pediatrics
Via Corridoni 11
60123 ANCONA, ITALY
Phone: +39 071 5962364
E-mail: c.catassi@staff.univpm.it
Prof. Dr. Fernando G. Chirdo
Universidad Nacional de La Plata
Facultad de Ciencias Exactas
Instituto de Estudios Immunologicos y
Fisiopatologicos - IIFP
Calle 47 y 115
1900 LA PLATA, ARGENTINA
Phone: +54 221 423 5 333 (Int 45)
E-mail: fchirdo@biol.unlp.edu.ar
Prof. Dr. Paul J. Ciclitira
University of East Anglia
Medical School
Bob Champion Building
James Watson Road
BR4 7UJ NORWICH
UNITED KINGDOM
Phone: +44 203 751 1104
E-mail: pciclitira@btinternet.com
Prof. Dr. Conleth Feighery
University of Dublin, Department of
Immunology, St. James’s Hospital
James’s Street
DUBLIN 8, IRELAND
Phone: +353 879969041
E-mail: cfighery@tcd.ie
Dr. Carmen Gianfrani
Institute of Biochemistry and
Cell Biology - CNR
Via Pietro Castellino 111
80131 NAPLES, ITALY
Phone: +39 081 6132224
E-mail: c.gianfrani@ibp.cnr.it
Prof. Dr. Peter Koehler
Biotask AG
Schelztorstraße 54-56
73728 ESSLINGEN, GERMANY
Phone: +49 711 31059068
E-mail: peter.koehler@biotask.de
Prof. Dr. Frits Koning
Leiden University Medical Centre, E3-Q
Department of Immunohaematology
and Bloodbank
Albinusdreef 2
2333 ZA LEIDEN, THE NETHERLANDS
Phone: +31 715 266673
E-mail: fkoning@lumc.nl
Prof. Dr. Knut Lundin
University of Oslo
Institute of Clinical Medicine
Postboks 1171, Blindern
0881 OSLO, NORWAY
Phone: +47 90980325
E-mail: knut.lundin@medisin.uio.no
Prof. Dr. Stefania Masci
University of Tuscia
Department of Agricultural and Forest
Sciences (DAFNE)
Via San Camillo de Lellis s.n.c.
01100 VITERBO, ITALY
E-mail: masci@unitus.it
Prof. Dr. Katharina Scherf
Karlsruhe Institute of Technology (KIT)
Institute of Applied Biosciences
Department of Bioactive and
Functional Food Chemistry
Adenauerring 20 a
76131 KARLSRUHE, GERMANY
Phone: +49 721 608 42929
E-mail: katharina.scherf@kit.edu
Prof. Dr. Dr. Detlef Schuppan
I. Medizinische Klinik und Poliklinik
Universitätsmedizin der Johannes
Gutenberg-Universität Mainz
Institut für Translationale Medizin
Langenbeckstraße 1
55131 MAINZ, GERMANY
Phone: +49 6131 177355/177356/177104
E-mail:
detlef.schuppan@unimedizin-mainz.de
Dr. René Smulders
Wageningen University & Research,
Plant Research
Droevendaalsesteeg 1
6708 PB WAGENINGEN,
THE NETHETRLANDS
Phone: +31 620298266
E-mail: rene.smulders@wur.nl
Dr. Olivier Tranquet
INRA
Rue de la Géraudière BP 71627
44316 NANTES, FRANCE
Phone: +33 2406 75027
E-mail: olivier.tranquet@inra.fr
Prof. Dr. Riccardo Troncone
University Federico II
Department of Pediatrics
Via Pansini 5
80131 NAPLES, ITALY
Phone: +39 3483132274
E-mail: troncone@unina.it
HOSTS
Mrs. Jacqueline Pante
Dr. Schär AG / SPA
Director of Corporate Nutrition Service
Winkelau 9
39014 BURGSTALL/POSTAL, ITALY
E-mail: Jacqueline.Pante@drschaer.com
Mrs. Fabiana Saorin
Dr. Schär AG / SPA
Corporate Nutrition Service
- HCP Relations
Winkelau 9
39014 BURGSTALL/POSTAL, ITALY
E-mail: Fabiana.Saorin@drschaer.com
INVITED SPEAKERS
Prof. Dr. Valentina Discepolo
University of Naples Federico II
Department of Translational
Medical Sciences
Via Sergio Pansini 5
80131 NAPLES, ITALY
E-mail: valentina.discepolo@unina.it
Mrs. Hertha Deutsch
Österreichische Arbeitsgemeinschaft
Zöliakie
Anton Baumgartner Straße 44/C5/2302
1230 VIENNA, AUSTRIA
E-mail: hertha.deutsch@chello.at
GUESTS
Dr. Guenther Augustin
Dr. Schär AG / SPA
Winkelau 9
39014 BURGSTALL/POSTAL, ITALY
E-mail: guenther.augustin@drschaer.com
Mrs. Sophie Ballmann
R-Biopharm AG
An der neuen Bergstraße 17
64297 DARMSTADT, GERMANY
E-mail: s.ballmann@r-biopharm.de
Mrs. Sofia Beisel
Deutsche Zöliakiegesellschaft e.V.
Kupferstraße 36,
70565 STUTTGART, GERMANY
E-mail: sofia.beisel@dzg-online.de
Dr. Markus Brandt
Ernst Böcker GmbH & Co KG
Ringstrasse 55-57
32427 MINDEN, GERMANY
E-mail: markus.brandt@sauerteig.de
Mr. Martin Candia
Romer Labs Division Holding GmbH
Erber Campus 1
3131 GETZERSDORF, AUSTRIA
E-mail: martin.candia@romerlabs.com
Dr. Linda Cavaletti
Via Carso 28b
22069 ROVELLASCA (CO), ITALY
E-mail:
lindacavaletti@ricercaperlavita.it
Dr. Angel Cebolla
Biomedal, SL
Calzada Romana 40
41900 CAMAS, SPAIN
E-mail: acebolla@biomedal.com
Dr. Virna Cerne
Dr. Schär R&D Centre
c/o AREA Science Park
Padriciano, 99
34149 TRIESTE, ITALY
E-mail: Virna.Cerne@drschaer.com
Prof. Dr. Michelle Colgrave
CSIRO
306 Carmody Road
St. Lucia QLD 4067, AUSTRALIA
E-mail: michelle.colgrave@csiro.au
Dr. Maria Cristina Comelli
NEMYSIS LIMITED
7 D’Olier Street
D02HF60 DUBLIN, IRELAND
E-mail: comelli@nemysisltd.com
Mr. Silvano Ciani
Dr. Schär R&D Centre
c/o AREA Science Park
Padriciano, 99
34149 TRIESTE, ITALY
E-mail: Silvano.Ciani@drschaer.com
Dr. Johan De Meester
Cargill R&D Centre Europe
Havenstraat 84
B-1800 VILVOORDE, BELGIUM
E-mail: Johan_De_Meester@cargill.com
Dr. Sandra Denery
INRAE
Rue de la Géraudière BP 71627
44316 NANTES, FRANCE
E-mail: sandra.denery@inrae.fr
Mrs. Tina Dubois
R-Biopharm AG
An der neuen Bergstraße 17
64297 DARMSTADT, GERMANY
E-mail: t.dubois@r-biopharm.de
Dr. Margareta Elding-Pontén
Fria Bröd AB
Fältspatsgatan 12
421 30 VÄSTRA FRÖLUNDA,
SWEDEN
E-mail: Margareta.Elding-Ponten@fria.se;
margareta@fria.se
Mr. Luke Emerson-Mason
Bia Diagnostics
480 Hercules Dr.
5446 COLCHESTER, VT, USA
E-mail: luke@biadiagnostics.com
Dr. Sophia Escobar-Correas
CSIRO
306 Carmody Road
St. Lucia QLD 4067, AUSTRALIA
E-mail: sophia.escobarcorreas@csiro.au
Mr. Richard Fielder
Bio-Check (UK)
Spectrum House, Llys Edmund Prys
St. Asaph Business Park
LL170JA ST. ASAPH,
UNITED KINGDOM
E-mail: richard@biocheck.uk.com
Mrs. Maren Finke
Hermann Kröner GmbH
Lengericher Straße 158
49479 IBBENBÜREN, GERMANY
E-mail: finke@kroener-staerke.de
Dr. Carlos Galera
Hygiena Diagnostica España
Calle Cañada Real 31-35
41900 CAMAS, SEVILLA, SPAIN
E-mail: cgalera@hygiena.com
Dr. Thomas Grace
Bia Diagnostics Laboratories
480 Hercules Dr.
5446 COLCHESTER, VT, USA
E-mail: thomasgrace@biadiagnostics.com
Dr. Xin Huang
University of Helsinki
Department of Food and
Environmental Sciences
Agnes Sjöbergin katu 2, PL66
14 HELSINKI, FINLAND
E-mail: xin.huang@helsinki.fi
Dr. Katharina Kessler
NEMYSIS LIMITED
7 D’Olier Street
D02HF60 DUBLIN, IRELAND
E-mail: kessler@nemysisltd.com
Mrs. Tunde Koltai
Hungarian Coeliac Society
Palanta utca 11
1025 BUDAPEST, HUNGARY
E-mail: tunde.koltai@gmail.com;
coeliac@t-online.hu"
Mr. Lukas Kraft
R-Biopharm AG
An der neuen Bergstraße 17
64297 DARMSTADT, GERMANY
E-mail: l.kraft@r-biopharm.de
Dr. Götz Kröner
Hermann Kröner GmbH
Lengericher Str. 158
49479 IBBENBÜREN, GERMANY
E-mail: kroener@kroener-staerke.de
Dr. Colette Larré
INRAE
Rue de la Géraudière BP 71627
44316 NANTES, FRANCE
E-mail: Colette.larre@inrae.fr
Ms Marie-Christin Lay
Leibniz-Institute for Food Systems
Biology
Lise-Meitner-Strasse 34
85354 FREISING, GERMANY
E-mail: m.lay.leibniz-lsb@tum.de
Dr. Gabriela Lopez Velasco
3M Company
3M Center 0260-B-01
55144 ST. PAUL, MN, USA
E-mail: glopez3@mmm.com
Dr. Patrick Mach
3M Company
3M Center, 0260-B-01
55144 ST. PAUL, MN, USA
E-Mail: pamach1@mmm.com
Dr. Veronica Marin
Dr. Schär R&D Centre
c/o AREA Science Park
Padriciano, 99
34149 TRIESTE, ITALY
E-mail: veronica.marin@drschaer.com
Dr. Chiara Monachesi
Università Politecnica delle Marche
Department of Pediatrics
Via Corridoni 11
60123 ANCONA, ITALY
E-mail: chiara.monachesi28@gmail.com
Dr. Elisa Mora de Checa
SMAP Celíacs Catalunya
Independencia, 257
08026 BARCELONA, SPAIN
E-mail: elisamoracheca@gmail.com
Dr. Susanna Neuholt
Associazione Italiana Celiachia
Via Caffaro, 10
16124 GENOVA, ITALY
E-mail: alimenti@celiachia.it
Dr. Luisa Novellino
Associazione Italiana Celiachia
Via Caffaro, 10
16124 GENOVA, ITALY
E-mail: lnovellino@celiachia.it
Mrs. Ombretta Polenghi
Dr. Schär R&D Centre
c/o AREA Science Park
Padriciano, 99
34149 TRIESTE, ITALY
E-mail: ombretta.polenghi@drschaer.com
Dr. Lea Pollak
Croatian Institute of Public Health
Rockefellerova 7
10000 ZAGREB, CROATIA
E-mail: lea.pollak@hzjz.hr
Dr. Adrian Rogers
Romer Labs UK Ltd.
The Health Business and
Technical Park
WA74QX RUNCORN, CHESHIRE,
UNITED KINGDOM
E-mail: adrian.rogers@romerlabs.com
Mrs. Cristina Romero
Eurofins INGENASA
Avda. Institución Libre de Enseñanza, 39
28037 MADRID, SPAIN
E-mail: cromero@ingenasa.com
Mr. Stefan Schmidt
R-Biopharm AG
An der neuen Bergstraße 17
64297 DARMSTADT, GERMANY
E-mail: st.schmidt@r-biopharm.de
Dr. Juan Ignacio Serrano-Vela
Asociación de Celíacos y Sensibles Al
Gluten, Comunidad de Madrid
Calle Lanuza 19-bajo
28028 MADRID, SPAIN
E-mail: nachoserrano@celiacosmadrid.org
Prof. Dr. Edurne Simón
University of the Basque Country
Paseo de la Universidad, 7
1006 VITORIA-GASTEIZ, SPAIN
E-mail: edurne.simon@ehu.es
Dr. Tuula Sontag-Strohm
University of Helsinki
Department of Food and
Environmental Sciences
Agnes Sjöbergin katu 2, PL66
14 HELSINKI, FINLAND
E-mail: tuula.sontag-strohm@helsinki.fi
Dr. Karoline Terberger
Böcker Sauerteig GmbH & Co. KG
Ringstraße 55-57
32427 MINDEN, GERMANY
E-mail: karoline.terberger@sauerteig.de
Dr. Catherine Torgler
Hygiena Diagnostica España
Calle Cañada Real 31-35
41900 CAMAS, SEVILLA, SPAIN
E-mail: ctorgler@hygiena.com
Dr. Heidi Urwin
Coeliac UK
3rd Floor, Apollo Centre, Desborough
Road
HP112QW HIGH WYCOMBE,
BUCKS, UNITED KINGDOM
E-mail: Heidi.Urwin@coeliac.org.uk
Dr. Anil K. Verma
Università Politecnica delle Marche
Department of Pediatrics
Via Corridoni 11
60123 ANCONA, ITALY
E-mail: anilkrvermaa@gmail.com
Dr. Niklas Weber
R-Biopharm AG
An der neuen Bergstraße 17
64297 DARMSTADT, GERMANY
E-mail: n.weber@r-biopharm.de
Dr. Paul Wehling
General Mills, Inc.
9000 Plymouth Ave N
55427 GOLDEN VALLEY, USA
E-mail: paul.wehling@genmills.com
Dr. Thomas Weiss
R-Biopharm AG
An der neuen Bergstraße 17
64297 DARMSTADT, GERMANY
E-mail: t.weiss@r-biopharm.de
Impressum
Proceedings of the 34th Meeting
WORKING GROUP
on PROLAMIN ANALYSIS and TOXICITY
15 – 16 October 2020
Postal, Italy
This work including all parts is subject to copyright. All rights are reserved and any
utilisation is only permitted under the provisions of the German Copyright Law.
Permissions for use must always be obtained from the publisher. This is in particular
valid for reproduction, translation, conversion to microfilm and for storage or
processing in electronic systems.
Scientific Organisation
Prof. Dr. Peter Koehler
biotask AG
Schelztorstraße 54-56, 73728 ESSLINGEN, GERMANY
Phone: +49 711 31059068; Fax: +49 711 31059070
E-mail: peter.koehler@biotask.de
Host
Mr. Ulrich Ladurner & Ms. Jacqueline Pante
Dr. Schär AG / SPA
Winkelau 9, 39014 Burgstall / Postal, Italy
Phone: +39 0473 293 351
E-mail: Jacqueline.Pante@drschaer.com
Cover picture
Dr. Schaer AG / SPA
View of the Dr. Schaer company building, location of the 34th PWG-meeting, 2020
© Peter Koehler 2020
|