2006-Detecting Allergens in Food - ISBN-10 0-8493-2574-9
2006-Detecting Allergens in Food - ISBN-10 0-8493-2574-9
2006-Detecting Allergens in Food - ISBN-10 0-8493-2574-9
Edited by
Stef J. Koppelman and Sue L. Hefle
CRC Press
Boca Raton Boston New York Washington, DC
Published in North America by CRC Press LLC, 6000 Broken Sound Parkway, NW,
Suite 300, Boca Raton, FL 33487, USA
First published 2006, Woodhead Publishing Ltd and CRC Press LLC
© 2006, Woodhead Publishing Ltd
The authors have asserted their moral rights.
This book contains information obtained from authentic and highly regarded sources.
Reprinted material is quoted with permission, and sources are indicated. Reasonable
efforts have been made to publish reliable data and information, but the authors and
the publishers cannot assume responsibility for the validity of all materials. Neither
the authors nor the publishers, nor anyone else associated with this publication, shall
be liable for any loss, damage or liability directly or indirectly caused or alleged to be
caused by this book.
Neither this book nor any part may be reproduced or transmitted in any form or by
any means, electronic or mechanical, including photocopying, microfilming and
recording, or by any information storage or retrieval system, without permission in
writing from the publishers.
The consent of Woodhead Publishing Limited does not extend to copying for
general distribution, for promotion, for creating new works, or for resale. Specific
permission must be obtained in writing from Woodhead Publishing Limited for such
copying.
The publishers’ policy is to use permanent paper from mills that operate a sustainable
forestry policy, and which has been manufactured from pulp which is processed
using acid-free and elementary chlorine-free practices. Furthermore, the publishers
ensure that the text paper and cover board used have met acceptable environmental
accreditation standards.
Chapter 1 Chapter 3
Professor Steve L. Taylor Professor Sue L. Hefle
Food Allergy Research & Resource Food Allergy Research & Resource
Program Program
University of Nebraska University of Nebraska
143 Food Industry Bldg 255 Food Industry Bldg
Lincoln, NE 68583-0919 Lincoln, NE 68583-0919
USA USA
Chapter 2 Chapter 4
Professor Dr Heimo Breiteneder Dr Robert G. Hamilton
Department of Pathophysiology Johns Hopkins University School
Medical University of Vienna of Medicine
AKH-EBO-3Q Johns Hopkins DACI Reference
Waehringer Guertel 18-20 Laboratory
1090 Vienna Room 1A20-JHAAC
Austria 5501 Hopkins Bayview Circle
Baltimore, MD 21224
E-mail: USA
heimo.breiteneder@meduniwien.ac.at
E-mail: rhamilto@jhmi.edu
xii Contributor contact details
Chapter 5 Chapter 9
Dr Ronald van Ree Dr Ingrid Malmheden Yman
Department of Immunopathology Swedish National Food
Sanquin Research Administration
PO Box 9190 PO Box 622
NL-1006 AD SE-751 26 Uppsala
Amsterdam Sweden
The Netherlands
E-mail:
E-mail: r.vanree@sanquin.nl ingrid.malmheden.yman@slv.se
Chapter 6 Chapter 10
Dr Jupiter Yeung Dr Sabine Baumgartner*
Food Products Association BOKU – University of Natural
Suite 300 Resources and Applied Life
13501 Street NW Sciences, Vienna
Washington, DC 20005 C/O Department for
USA Agrobiotechnology, IFA – Tulln
Centre for Analytical Chemistry
E-mail: jyeung@fpa-food.org Konrad-Lorenz-Strasse 20
A-3430 Tulln
Austria
Chapter 7
Dr Stefan Vieths E-mail:
Paul-Ehrlich-Institut sabine.baumgartner@boku.ac.at
Paul-Ehrlich-Strasse 51-59
D-63225 Langen Dr Rob van Herwijnen
Germany
E-mail: info@evlonline.nl (att. Dr
E-mail: viest@pei.de Rob van Herwijnen)
Chapter 8 Chapter 11
Professor Michael G. Zeece Professor Sue L. Hefle
Department of Food Science & Food Allergy Research & Resource
Technology Program
University of Nebraska University of Nebraska
354 Food Industry Bldg 255 Food Industry Bldg
Lincoln, NE 68583-0919 Lincoln, NE 68583-0919
USA USA
Chapter 12 Chapter 15
Dr Stef Koppelman Dr Stef Koppelman*
Department of Dermatology/ Department of Dermatology/
Allergology Allergology
University Medical Centre Utrecht University Medical Centre Utrecht
NL-3508 GA Utrecht NL-3508 GA Utrecht
The Netherlands The Netherlands
E-mail: rene.crevel@unilever.com
xiv Contributor contact details
Chapter 18 Chapter 21
Dr Ulrike Immer Heereluurt Heeres LL.M., B.Sc.
R-Biopharm AG TNO Quality of Life
Landwehrstrasse 54 PO Box 360
D-64293 Darmstadt NL-3700 AJ Zeist
Germany The Netherlands
Chapter 19 Chapter 22
Dr Roland E. Poms* Dr Stef Koppelman*
International Association for Cereal Department of Dermatology/
Science and Technology (ICC) Allergology
Marxergasse 2 University Medical Centre Utrecht
A-1030 Vienna NL-3508 GA Utrecht
Austria The Netherlands
Chapter 20
Mr Martin J. Hahn
Hogan & Hartson LLP
555 13th Street NW
Washington, DC 20004
USA
E-mail: MJHahn@HHLaw.com
Preface
With food allergies on the increase in Western societies, and new legislation
on allergen labeling in both Europe and the USA, the food industry is confronted
with the issue of providing safe foods for the food-allergic consumer. While
most food-allergic reactions occur after ingestion of non-packaged food
products, the food industry has been subjected to increasing scrutiny of its
allergen controls; the resulting impact on the industry has been remarkable.
In the past 15 years, the food industry has made significant investment,
effort, and improvements in allergen control. In the past eight years, tests for
some allergenic foods have been commercialized and have proven useful to
the industry in controlling allergens, and also to regulatory agencies
investigating food-allergic consumer complaints. There are many strategies
food manufacturers can exercise in controlling allergens in their plants, from
changing raw materials to improving cleaning procedures and using
precautionary labeling indicating allergens that might be present. However,
measuring the content of particular allergenic residues on processing equipment,
in ingredients or in final products, provides information that can also be used
for risk assessment, enabling the food industry to provide the food-allergic
consumer with practical information. The inspiration for this book originated
in the early years of this millennium when increasing numbers of commercial
test kits for measuring allergenic food residues were launched. The purpose
of the book is to serve as a resource for experts from different backgrounds
such as biochemistry, food chemistry, food legislation, and allergology with
regard to the technical possibilities and limitations of food allergen detection
methods.
The first part of the book deals with the nature of food allergy and introduces
the reader to the range of allergenic foods. What causes an individual to be
at risk from food allergy? What makes a protein an allergen? How much
allergenic residue is too much for a food-allergic individual? What is the
nature of the immune response to food that causes allergic reactions? These
questions will be discussed as a basis for the remainder of the book.
Part II deals with methods designed for measuring food allergens, based
xvi Preface
Preface ................................................................................... xv
Stimulates
Allergen production
of
IgE Sensitized
Mast cell
basophil cell
Sensitized cell
Allergen
Degranulation
Release:
histamine and
other mediators
dose-response modeling for peanut, milk, and egg (where the largest number
of challenges has been obtained) indicate that only 1% or less of the affected
population would react to residual levels of the allergenic food of 0.26 mg
for egg, 0.76 mg for peanut, and 7.76 mg for milk.51 Additional controlled
challenge studies conducted with standard challenge protocols will likely
reduce the uncertainty of these estimates in coming years.
Existing test methods for residues of allergenic foods appear to be in the
ideal range to detect potentially hazardous residues of undeclared allergens
in foods. Current immunoassay methods have lower limits of about 1 ppm
(1 mg/kg). If minimal eliciting doses are 200 μg or higher and typical serving
sizes are 50–100 g or less, then potentially hazardous foods would be easily
detected with these immunoassay methods. If a highly sensitive individual
ingested a typical 50 g serving of a food containing 200 μg of undeclared
residues of an allergenic food, that food would have contained 4 ppm of the
offending food. Previously, the advice given was that foods containing less
than 10 ppm of undeclared residues of an allergic food would present little
risk to food-allergic consumers.53 That advice remains fairly sound now that
much more evidence exists on minimal eliciting doses. The development of
analytical procedures that are more sensitive than the current ones does not
seem to be justified.
Food-allergic consumers, especially those susceptible to comparatively
low threshold levels, can occasionally experience reactions to packaged food
products even when the ingredient list indicates that the particular item
should be safe. In manufacturing situations, foods may become contaminated
with trace amounts of other foods through various means including the use
of rework and the use of shared equipment.54,55 No avoidance diet provides
absolute safety, but careful adherence to an effective avoidance diet will
minimize the chances of a reaction.
1.3.3 Cross-reactions
Ubiquitous statements cannot be made about cross-reactions between closely-
related foods. Cross-reactivity to closely-related foods occurs in some situations,
but not others. For example, individuals with a shrimp allergy are often
sensitive to all species of shrimp and to other crustacean shellfish such as
crab and lobster.65 Tropomyosin is a pan-allergen existing in all crustacean
shellfish66 and possibly also in some species of molluscan shellfish.66 Fish
have a distinctly different pan-allergen, parvalbumin, that exists in all species
of fish,63 although fish may also contain certain other minor allergens and
cross-reactivity has not been universally observed.63 Some peanut-allergic
individuals are allergic to other legumes, such as soybeans,67 although this is
not a frequent occurrence. Clinical hypersensitivity to one legume, such as
peanuts or soybeans, does not warrant dietary elimination of the entire legume
food family unless allergy to each legume is individually confirmed by double-
blind, placebo-controlled food challenges (DBPCFC).68 In contrast, it is
known that cross-reactions commonly occur between different species of
avian eggs69 and between cows’ milk and goats’ milk.70
The nature of food allergy 15
these methods will be important quality assurance tools for the food industry
because they will allow the validation of sanitation practices on shared
equipment. Shared equipment is an economic necessity in the food industry
and is common in a wide variety of food industry sectors (e.g. ice cream,
confectionary, bakery, pasta). Proper sanitation of that shared equipment is
critical to mitigation of potential allergen hazards, and analytical tools will
facilitate the validation of sanitation practices. Clearly, analytical methods
must be specific, allowing the detection of an allergenic food in the presence
of others, and must be appropriately sensitive. The ideal range of sensitivity
of the methods should be driven by knowledge of minimal eliciting doses.
The goal should be to protect allergic consumers while allowing them the
greatest variety of foods in their diets rather than the detection of miniscule
levels of allergenic food residues that have no adverse health consequences
on even the most sensitive individuals.
1.6 References
1. Bush, R K and Hefle S L (1996) ‘Food allergens’, Crit Rev Food Sci Nutr, 36
(suppl.), S119–S163.
2. Kidd, J M, Cohen, S H, Sosman A J and Fink J N (1983) ‘Food-dependent exercise-
induced anaphylaxis’, J Allergy Clin Immunol, 71, 407–411.
3. Castells, M C, Horan, R F and Sheffer, A L (2003) ‘Exercise-induced anaphylaxis’,
Curr Allergy Asthma Rpts, 3, 15–21.
4. Knicker, W T (1999) ‘Delayed and non-IgE-mediated reactions’, in Frieri, M and
Kettelhut, B, Food Hypersensitivity and Adverse Reactions – A Practical Guide to
Diagnosis and Management, New York, Marcel Dekker, 165–217.
5. Taylor, S L and Hefle, S L (2001) ‘Food allergies and other food sensitivities’, Food
Technol, 55(9), 68–83.
6. Suarez, F L and Savaiano, D A (1997) ‘Diet, genetics, and lactose intolerance’, Food
Technol, 51, 74–76.
7. Bush, R K, Taylor S L and Hefle S L (2003) ‘Adverse reactions to food and drug
additives’, in Adkinson, N F, Yunginger, J W, Busse, W W, Bochner, B S, Holgate,
S T and Simons, F E R, Middleton’s Allergy Principles and Practice, Vol. 2, 6th edn,
St Louis, MO, Mosby, 1645–1663.
8. Taylor, S L, Stratton, J E and Nordlee, J A (1989) ‘Histamine poisoning (scombroid
fish poisoning): an allergy-like intoxication’, Clin Toxicol, 27, 225–240.
9. Sampson, H A (1990) ‘Food allergy’, Curr Opinion Immunol, 2, 542–547.
10. Sicherer, S H, Munoz-Furlong, A and Sampson, H A (2004) ‘Prevalence of seafood
allergy in the United States determined by a random telephone survey’, J Allergy
Clin Immunol, 114, 159–165.
11. Sicherer, S H, Munoz-Furlong, A, Burks, A W and Sampson, H A (1999) ‘Prevalence
of peanut and tree nut allergy in the US determined by a random digit dial telephone
survey’, J Allergy Clin Immunol, 103, 559–562.
12. Sampson, H A and McCaskill, C C (1985) ‘Food hypersensitivity and atopic dermatitis:
evaluation of 113 patients’, J Pediatr, 107, 669–675.
13. Taylor, S L and Hefle, S L (2002) ‘Allergic reactions and food intolerances’, in
Kotsonis, F N and Mackey, M A, Nutritional Toxicology, 2nd edn, London, Taylor
and Francis, 93–121.
14. Sampson, H A, Mendelson, L and Rosen, J P (1992) ‘Fatal and near-fatal anaphylactic
reactions to food in children and adolescents’, N Engl J Med, 327, 380–384.
15. Bock, S A, Munoz-Furlong, A and Sampson, H A (2001) ‘Fatalities due to anaphylactic
reactions to foods’, J Allergy Clin Immunol, 107, 191–193.
16. Pastorello, E A and Ortolani, C (2003) ‘Oral allergy syndrome,’ in Metcalfe, D D,
Sampson, H A and Simon, R A, Food Allergy – Adverse Reactions to Foods and Food
Additives, 3rd edn, Malden, MA, Blackwell Science, 169–182.
17. Taylor, S L and Lehrer, S B (1996) ‘Principles and characteristics of food allergens’,
Crit Rev Food Sci Nutr, 36 (Suppl), S91–S118.
18. Calkhoven, P G, Aalbers, M, Koshte, V L, Pos, O, Oei, H D and Aalberse, R C
(1987) ‘Cross-reactivity among birch pollen, vegetables and fruits as detected by
IgE antibodies is due to at least three distinct cross-reactive structures’, Allergy, 42,
382–390.
19. Maulitz, R M, Pratt, D S and Schocket, A L (1979) ‘Exercise-induced anaphylactic
reaction to shellfish’, J Allergy Clin Immunol, 63, 433–434.
20. Kushimoto, H and Aoki, T (1985) ‘Masked type I wheat allergy. Relation to exercise-
induced anaphylaxis’, Arch Dermatol, 121, 355–360.
21. Strober, W (1986) ‘Gluten-sensitive enteropathy: a nonallergic immune hypersensitivity
of the gastrointestinal tract’, J Allergy Clin Immunol, 78, 202–211.
22. Taylor, S L, Hefle, S L and Gauger, B J (2001) ‘Food allergies and sensitivities’, in
Helferich, W and Winter, C K, Food Toxicology, Boca Raton, FL, CRC Press, 1–36.
18 Detecting allergens in food
23. Prausnitz, C and Kustner, H (1921) ‘Studien uber die Ueberempfindlichkeit’, Centralbl
fur Bakt, 86, 160–169.
24. Ishizaka, K, Ishizaka, T and Hornbrook, M M (1966) ‘Physico-chemical properties
of reaginic antibody. V. Correlation of reaginic activity with gamma-E-globulin
antibody’, J Immunol, 97, 840–853.
25. Ishizaka, K, Ishizaka, T and Hornbrook, M M (1966) ‘Physico-chemical properties
of human reaginic antibody. IV. Presence of a unique immunoglobin as a carrier of
reaginic activity’, J Immunol, 97, 75–85.
26. Ishizaka, T (1982) ‘IgE and mechanisms of IgE-mediated hypersensitivity’, Ann
Allergy, 48, 313–319.
27. Taylor, S L (2002) ‘Protein allergenicity assessment of foods produced through
agricultural biotechnology’, Ann Rev Pharmacol Toxicol, 42, 99–112.
28. Esch, R E and Bush, R K (2003) ‘Aerobiology of outdoor allergens’, in Adkinson,
N F, Yunginger, J W, Busse, W W, Bochner, B S, Holgate, S T and Simons, F E R,
Middleton’s Allergy Principles and Practice, Vol. 1, 6th edn, St Louis, MO, Mosby,
529–572.
29. Sicherer, S H (2002) ‘Food allergy’, Lancet, 360, 701–710.
30. Church, M K, Shute, J K and Sampson, A P (2003) ‘Mast cell-derived mediators’,
in Adkinson, N F, Yunginger J W, Busse, W W, Bochner, B S, Holgate, S T and
Simons, F E R, Middleton’s Allergy Principles and Practice, Vol 1, 6th edn, St Louis,
MO, Mosby, 189–212.
31. Bush, R K and Taylor, S L (2003) ‘Histamine’, in Caballero, B, Trugo, L and
Finglas, P, Encyclopedia of Food Sciences and Nutrition, 2nd edn, Oxford, Elsevier
Science, 3108–3111.
32. Holt, P G and Björksten, B (2003) ‘Development of immunological tolerance to
food antigens’, in Metcalfe, D D, Sampson, H A and Simon, R A, Food Allergy –
Adverse Reactions to Foods and Food Additives, 3rd edn, Malden, MA, Blackwell
Science, 81–90.
33. Chandra, R K (1987) ‘Food allergy: setting the theme’, in Chandra, R K, Food
Allergy, Nutrition Research Education Foundation, St. John’s, Newfoundland, 3–5.
34. O’Connor, M E and Schocket, A L (2003) ‘Exercise- and pressure-induced syndromes’,
in Metcalfe, D D, Sampson, H A and Simon, R A, Food Allergy – Adverse Reactions
to Foods and Food Additives, 3nd edn, Malden, MA, Blackwell Science, 262–269.
35. Sampson, H A (1991) ‘Immunologic mechanisms in adverse reactions to foods’,
Immunol Allergy Clin North Am, 11, 701–716.
36. Stern, M and Walker, W A (1985) ‘Food allergy and intolerance’, Pediatr Clin North
Am, 32, 471–492.
37. Ford, R P K, Hill, D J and Hosking, C S (1983) ‘Cows’ milk hypersensitivity:
immediate and delayed onset clinical patterns’, Arch Dis Child, 58, 856–862.
38. Ferguson, A (1997) ‘Gluten-sensitive enteropathy (celiac disease)’, in Metcalfe, D
D, Sampson, H A and Simon, R A, Food Allergy – Adverse Reactions to Foods and
Food Additives, 2nd edn, Cambridge, Blackwell Science, 287–301.
39. Janatuinen, E K, Pikkarainen, P H, Kemppainen, T A, Kosma, V M, Jarvinen, R M
K, Uusitupa, M I J and Julkunen, R J K (1995) ‘A comparison of diets with and
without oats in adults with celiac disease’, N Engl J Med, 333, 1033–1037.
40. Skerritt, J H, Devery, J M and Hill, A S (1990) ‘Gluten intolerance: chemistry,
celiac-toxicity, and detection of prolamins in foods’, Cereal Foods World, 35, 638–
644.
41. Taylor, S L (1990) ‘Food allergies and related adverse reactions to foods: a food
science perspective’, in Perkin, J E, Food Allergies and Adverse Reactions, Rockville,
MD, Aspen Publishers, 189–206.
42. Furukawa, C T (1988) ‘Nondietary management of food allergy’, in Chiaramonte, L
T, Schneider, A T and Lifshitz, F, Food Allergy – A Practical Approach to Diagnosis
and Management, New York, Marcel Dekker, 365–375.
The nature of food allergy 19
63. Taylor, S L, Kabourek, J L and Hefle, S L (2004) ‘Fish allergy: fish and products
thereof’ J Food Sci, 69, R175–180.
64. Hansen, T K, Poulsen, L K, Skov, P S, Hefle, S L, Hlywka, J J, Taylor, S L,
Bindslev-Jensen, U and Bindslev-Jensen, C (2004) ‘A randomized, double-blinded,
placebo-controlled oral challenge study to evaluate the allergenicity of commercial,
food-grade fish gelatin’ Food Chem Toxicol, 42, 2037–2044.
65. Lehrer, S B (1986) ‘The complex nature of food antigens: studies on cross-reacting
crustacea allergens’, Ann Allergy, 57, 267–272.
66. Reese, G, Ayuso, R and Lehrer, S B (1999) ‘Tropomyosin: an invertebrate pan-
allergen’, Int Arch Allergy Immunol, 119, 247–258.
67. Herian, A M, Taylor, S L and Bush, R K (1990) ‘Identification of soybean allergens
by immunoblotting with sera from soy-allergic adults’, Int Arch Allergy Appl Immunol,
92, 193–198.
68. Bernhisel-Broadbent, J and Sampson, H A (1989) ‘Cross-allergenicity in the legume
botanical family in children with food hypersensitivity’, J Allergy Clin Immunol, 83,
435–440.
69. Langeland, T (1983) ‘A clinical and immunological study of allergy to hen’s egg
white. VI. Occurrence of proteins cross-reacting with allergens in hen’s egg white as
studied in egg white from turkey, duck, goose, seagull, and in hen egg yolk, and hen
and chicken sera and flesh’, Allergy 38, 399–412.
70. Bellioni-Businco, B, Paganelli, R, Lucenti, P, Giampietro, P G, Perborn, H and
Businco, L (1999) ‘Allergenicity of goat’s milk in children with cow’s milk allergy’,
J Allergy Clin Immunol, 103, 1191–1194.
71. Van Ree, R and Aalberse, R C (1993) ‘Pollen-vegetable food crossreactivity: serological
and clinical relevance of crossreactive IgE’, J Clin Immunoassay, 16, 124–130.
72. Ballmer-Weber, B K, Vieths, S, Luttkopf, D, Heuschmann, P and Wuthrich, B (2000)
‘Celery allergy confirmed by double-blind, placebo-controlled food challenge: a
clinical study in 32 subjects with a history of adverse reactions to celery root’, J
Allergy Clin Immunol, 106, 373–378.
73. Enberg, R N, Leickly, F E, McCullough, J, Bailey, J and Ownby, D R (1987)
‘Watermelon and ragweed share allergens’, J Allergy Clin Immunol, 79, 867–875.
74. Blanco, C, Carrillo, T, Castillo, R, Quiralte, J and Cuevas, M (1994) ‘Latex allergy:
clinical features and cross-reactivity with fruits’, Ann Allergy, 73, 309–314.
75. Aalberse, R C (1992) ‘Clinically significant cross-reactivities among allergens’, Int
Arch Allergy Immunol, 99, 261–264.
2
2.1 Introduction
Plant tissues including those used for human consumption contain thousands
of different proteins. Proteins are clustered together into families if they have
residue identities of 30% and greater, or if they have lower sequence identities
but their functions and structures are very similar.1 Families whose proteins
have low sequence identities but whose structures and functional features
suggest a probable common evolutionary origin are placed together in
superfamilies.1 The Structural Classification of Proteins (SCOP) database
(http://scop.mrc-lmb.cam.ac.uk/scop/count.html) to date includes 2164 protein
families and 1232 protein superfamilies.
However, most plant food allergens are included in only a few protein
families and superfamilies. They belong to the cupin superfamily (7S and
11S seed storage proteins), the prolamin superfamily (2S albumins, non-
specific lipid transfer proteins (nsLTPs), alpha-amylase/trypsin inhibitors,
prolamin storage proteins of cereals), or the papain superfamily of cysteine
proteinases (Table 2.1). The pathogenesis-related (PR) proteins represent a
heterogeneous collection of 14 plant protein families that are all involved in
plant resistance to pathogens or adverse environmental conditions.2 Many
plant food allergens are homologous to such PR proteins (Table 2.2).3,4
Storage proteins are the cause of well-known allergic reactions to peanuts
and cereals.3 PR proteins are responsible for pollen–fruit or latex–fruit cross-
reactive syndromes.3,4 In addition, there are some structural and metabolic
plant protein families which harbour allergenic proteins such as the profilins
or isoflavone reductase homologues (Table 2.3).3 In animal-derived food
products one finds a much smaller variety of allergens. Seafood contains
22 Detecting allergens in food
Cupin superfamily
Vicilins (7S seed storage proteins) Ara h 1 (peanut), alpha-subunit of beta
conglycinin (soybean), Jug r 2 (English
walnut), Len c 1 (lentil), Ana o 1
(cashew), Ses i 3 (sesame)
Legumins (11S seed storage proteins) Ara h 3 and Ara h 4 (peanut), glycinin
subunits (soybean), Cor a 9 (hazelnut),
AMP (almond)
Prolamin superfamily
2S albumins Sin a 1 (yellow mustard), Ber e 1 (Brazil
nut), Jug r 1 (English walnut), Ses i 2
(sesame), Ara h 2, Ara h 6, Ara h 7
(peanut)
Non-specific lipid transfer proteins Pru p 3 (peach), Mal d 3 (apple), Pru ar 3
(apricot), Cor a 8 (hazelnut), Aspa o 1
(asparagus), Lac s 1 (lettuce)
Alpha-amylase/protease inhibitors Hor v 15 (barley), Sec c 1 (rye), RAPs
(rice allergenic proteins)
Cereal prolamins Tri a 19 (wheat), Sec c 20 (rye), Hor v
21 (barley)
Papain superfamily of cysteine proteases
Papain-like cysteine proteases Act c 1 (kiwi), papain (papaya), bromelain
(pineapple), P34/Gly m Bd 30K (soybean)
Vicilins
Molecular characteristics
The mature 7S globulins are homotrimeric proteins of about 150–190 kDa
but may undergo reversible aggregation into hexamers, depending on the
ionic strength of the ambient medium.8 The molecular weights of the subunits
range from about 40–80 kDa. Vicilins lack cysteines and, therefore, contain
no disulfide bonds. Their detailed subunit compositions vary considerably
due to differences in proteolytical processing and glycosylation.9 Among the
vicilins are two major variants in size, the regular vicilins with subunits of
about 50 kDa and the large vicilins of the convicilin and beta-conglycinin
type which have additional N-terminal insertions of about 80–190 residues
in length.10 The three-dimensional structures of three 7S globulins have been
determined: canavalin from jack bean,11,12 phaseolin from French bean,13
and the beta subunit of beta-conglycinin from soybean.14 These structures
show that the trimeric vicilins are disk shaped.
Classifying food allergens 25
Allergenic vicilins
Probably the best characterized allergenic vicilin is the major peanut allergen
Ara h 1, one of the main storage proteins of the peanut, and recognized by
serum IgE from over 90% of peanut allergic patients.15 Its linear IgE-epitopes
have been mapped and shown to consist of 23 independent binding sites.16
Mutational analysis of the immunodominant epitopes revealed that single
amino acid changes within these peptides had dramatic effects on IgE-binding
characteristics. The 63.5 kDa Ara h 1 forms highly stable homotrimers, a
characteristic that may be important for its allergenic properties.17 The majority
of the IgE-binding epitopes are located in the area of the subunit–subunit
contacts. These sites are protected from protease degradation, indicating that
the protein structure may play a significant role in overall allergenicity.18
Ara h 1 purified from peanuts and subjected to dry heat treatments at different
temperatures exhibited IgE-binding properties similar to those found for
native Ara h 1, indicating that the allergenicity of Ara h 1 is heat-stable
although the conformation of native Ara h 1 undergoes significant heat-
induced change.19 However, compared with dry roasted peanuts, the relative
amount of Ara h 1 was reduced in fried and boiled preparations as practised
in China which resulted in a significant reduction of IgE-binding intensity.20
Clinically relevant cross-reactivity between pea and peanut does occur. Vicilin
homologues in pea and peanut (Ara h 1) are the molecular basis for this
cross-reactivity.21
Beta-conglycinin, a glycoprotein of 180 kDa, is a major soybean globulin
that comprises about 50% of the 7S fraction. It forms trimers that are composed
of three subunits, alpha, alpha′, and beta. These have molecular weights of
76, 72, and 53 kDa, respectively.22 Beta-conglycinin is a heterogenous mixture
of different molecular species resulting from the various combinations of the
three subunits. One of the major allergenic proteins in the soybean 7S-
globulin fraction was identified as the alpha subunit of beta-conglycinin,
also known as Gly m Bd 60K.23
To date, several other 7S globulin family members have been identified as
food allergens. These include Jug r 2, a 47 kDa allergenic vicilin from
English walnuts;24 Len c 1, a gamma-vicilin subunit from lentils;25 Ana o 1
from cashew;26 and Ses i 3 from sesame seeds.27
Legumins
Molecular characteristics
The mature 11S globulins that are initially assembled as intermediate trimers
are hexameric.28 They comprise six subunit pairs that interact non-covalently
and are arranged in an open ring structure of approximately 360 kDa. Each
subunit pair consists in turn of an acidic 30–40 kDa polypeptide linked by a
disulfide bond to a basic polypeptide of ~ 20 kDa. Each subunit pair is
synthesized as a single precursor that is post-translationally cleaved after
disulfide bridge formation into an acidic and a basic polypeptide chain. The
basic or C-terminal chain of the 11S legumins is related to the C-terminal
26 Detecting allergens in food
Allergenic legumins
Two important peanut allergens belong to the 11S legumin-like seed storage
proteins. The 14 kDa Ara h 3 represents the N-terminal part of a 57 kDa
peanut glycinin subunit. A cDNA clone encoding the full-length protein was
isolated and the linear IgE-binding epitopes of Ara h 3 were mapped.30 The
recombinant Ara h 3 was expressed in E. coli and was recognized by serum
IgE from about 50% of a peanut allergic patient population.31 The cDNA
clone of Ara h 4 encodes a protein of 35.9 kDa with a sequence similarity of
70% to the glycinin family of proteins.32 The structure of Ara h 3 is similar
to that of soybean glycinin, and both the basic and the acidic subunit can
bind IgE from peanut allergic individuals.33
The 11S fraction of soybean proteins consists almost entirely of glycinin,
the principal soy globulin. Soybean glycinin is a major seed storage protein
that makes up more than 20% of the soybean seed by dry weight and 35–
40% of total soy protein. Native glycinin is a 350 kDa hexamer composed of
six non-identical subunits. The five known subunits (G1–G5) of soybean
glycinins are encoded by a small gene family. IgE epitopes for the soybean
glycinin G1 acidic chain have been determined and found to be similar to
IgE epitopes of the peanut glycinin Ara h 3.34 Moreover, each basic chain
from the five glycinin subunits reacted with IgE from soybean allergic
individuals to a similar extent.35 The linear IgE epitopes of the basic chain of
the glycinin G2 subunit were mapped and a three-dimensional model was
developed using the known structure of the 7S phaseolin.36 Additional 11S
allergenic plant food globulins were identified as Cor a 9 in hazelnut,37 in
coconut and walnut,38 and as AMP (almond major protein) in almond.39
2S albumins
Molecular characteristics
The 2S albumins constitute a family of structurally related homologous proteins
and form a major group of storage proteins in many dicotyledonous plant
species.46 Many 2S albumins possess high levels of sulfur-containing amino
acid residues. Typical 2S albumins, such as the napins from the Brassicaceae
or the Brazil nut 2S albumin, are heterodimeric proteins consisting of two
polypeptide chains with molecular weights of about 4 and 9 kDa. They are
synthesized as single precursor proteins that are proteolytically cleaved with
the loss of a linker peptide and short peptides from both the N- and the C-
terminus.47 These proteins are rich in alpha-helices and are held together by
four disulfide bonds involving eight conserved cysteine residues.46 The major
role of 2S albumins is as seed storage proteins for the developing seed. They
appear to be stable to proteolysis and can bind lipids. In addition, antifungal
activity against a number of plant-pathogenic fungi has been shown for the
napins from radish.48
Allergenic 2S albumins
Water-soluble allergenic 2S albumin storage proteins occur in many cultivated
species of dicotyledonous plants and include Sin a 1 from yellow mustard
seeds (Sinapis alba),49 Bra j 1 from oriental mustard seeds (Brassica juncea),50
the napin BnIII/Bra n 1 from oilseed rape (Brassica napus),51 Ber e 1 from
Brazil nut (Bertholletia excelsior),52 Jug r 1 from the English walnut (Juglans
28 Detecting allergens in food
Allergenic nsLTPs
The nsLTPs have an even wider distribution than the 2S albumins with
sequences being available from seeds, fruits, and vegetables. In the
Mediterranean area, Rosaceae fruits, particularly peach, are among the most
frequent causes of food-induced allergic reactions. LTPs represent major
plant food allergens in populations in these regions that are virtually free of
Fagales trees, in particular birch. LTPs have been identified as major peach
(Pru p 3),63,64 apple (Mal d 3),65 and apricot (Pru ar 3)66 allergens in
Mediterranean populations. In general, the LTP allergens of the Prunoideae
subfamily of the botanical family of the Rosaceae whose sequence identities
are between 88 and 98% are highly cross-reactive. In addition to peach and
apricot, this subfamily includes sweet cherry and the European plum, whose
LTPs have been described as the allergens Pru av 367 and Pru d 3,68 respectively.
LTPs seem to be able to sensitize subjects who have not been previously
sensitized by pollen allergens. The full spectrum of cross-reactivity with
different foods containing LTPs has yet to be investigated. Some of the
Classifying food allergens 29
patients sensitized to peach LTP are also clinically allergic to tomato and
corn, whereas the hazelnut LTP Cor a 8 was described as highly cross-
reactive with the peach LTP.69 The chestnut LTP, Cas s 8, was found to share
only some IgE epitopes with the corresponding peach allergen.70 The list of
allergenic nsLTPs is continuously expanding, encompassing more seeds (Zea
m 14 from corn),71 vegetables (Aspa o 1 from asparagus),72 and fruits (Vit v
1 from grape).73 Although allergic reactions to lettuce are not frequently
reported, its nsLTP was shown to cause anaphylaxis in susceptible individuals
and, therefore, received the nomenclature designation Lac s 1.74
Cereal prolamins
The ethanol-soluble cereal prolamins, gliadins in wheat, secalins in rye, and
hordeins in barley, are the major storage proteins found in the endosperm of
cereal grains. They are unusually rich in proline and glutamine. Several
sulfur-rich members of the prolamin superfamily were identified as allergens
from wheat. The highest IgE reactivity was found for low molecular mass
(LMM) glutenin followed by alpha-gliadin and gamma-gliadin.86 The allergenic
LMM glutenin was shown to contain a number of pentapeptide repeat motifs,
Gln-Gln-Gln-Pro-Pro.87 Alpha-gliadin, gamma-gliadin, and LMM glutenin
30 Detecting allergens in food
PR-2: beta-1,3-glucanases
Molecular characteristics
O-Glycosyl hydrolases are a widespread group of enzymes that hydrolyze
the glycosidic bond between two or more carbohydrates, or between a
carbohydrate and a non-carbohydrate moiety.104 A classification system for
glycosyl hydrolases, based on sequence similarity, has led to the definition
of 85 different families.105 Plant beta-1,3-glucanases are monomers with a
molecular weight in the 25–35 kDa range. Most plant beta-1,3-glucanases
are endoglucanases with the potential to partially degrade fungal cell walls
by hydrolyzing the beta-1,3-glucan fibers of the growing hyphae of filamentous
fungi.
32 Detecting allergens in food
PR-2-like allergens
The latex–fruit syndrome describes allergy to Hevea brasiliensis latex products
and an associated allergy to certain plant foods, especially avocado, banana,
chestnut, fig, and kiwi.106 It is speculated that the inhalation of PR-2 proteins
is one of the causes for the cross-reactivity of latex with plant foods.
Homologous proteins present in these fruits and vegetables are regarded as
responsible for the observed cross-reactivity. The basic beta-1,3-glucanases
from Hevea latex were recognized by specific IgE from food-allergic patients
suffering from hypersensitivity to banana, potato, and tomato – rather than to
latex products.107 Certain isoforms of the allergenic beta-1,3-glucanases from
H. brasiliensis are glycosylated. The carbohydrate moieties seem to harbour
the major IgE epitopes and may be responsible for the observed cross-
reactivities to beta-1,3-glucanases from fruits and vegetables.108 Banana is a
commonly reported cross-reactive food included in the latex–fruit syndrome.
An abundant beta-1,3-endoglucanase was isolated from ripe bananas and its
cDNA was cloned.109 Its allergenic potential has not yet been determined.
PR-3-like allergens
Relevant allergens of chestnut and avocado have been identified as class I
chitinases.112 Pers a 1, an endochitinase and major allergen from avocado,
was cloned and expressed in the yeast Pichia pastoris.113 Two major IgE-
binding proteins from banana were identified as class I chitinases.114 Class
I chitinases have an N-terminal domain that is homologous to hevein, one of
the major allergens of latex. Hevein (Hev b 6.02) occurs in Hevea latex
cleaved from prohevein and shares high sequence similarities with the hevein
domain of class I chitinases. Hevein contributes to the increased prevalence
of fruit allergies in individuals allergic to latex. Sensitization to Pers a 1
seems to be caused by hevein in the majority of patients allergic to latex.115
Class I chitinases are now regarded as the major pan-allergens in fruits
associated with the latex–fruit syndrome. These enzymes are inactivated by
heat treatment which may explain why only freshly consumed fruits are
associated with the latex–fruit syndrome. The hevein-like domains of allergenic
class I chitinases seem to include all the main IgE epitopes.116
Classifying food allergens 33
PR-4-like allergens
Wounding and chemical treatment of turnip (Brassica rapa) plants induced
the expression of an 18.7 kDa allergen that was recognized by IgE of 80%
of 60 sera of natural rubber latex allergic individuals.121 It seems likely that
prohevein and hevein were the primary sensitizers and the IgE binding to the
turnip allergen was a result of allergen cross-reactivity. Peptide sequencing
of the turnip allergen revealed 70% identity to prohevein and high similarities
to wound-induced proteins from tomato (74%) and potato (71%). Cloning of
a hevein-like fruit protein from mature elderberry (Sambucus nigra) fruits
demonstrated the occurrence of a hybrid gene encoding a protein consisting
of the N-terminal hevein-like domain of PR-4 proteins and the C-terminal
domain of class V chitinases.122 The allergenic properties of this elderberry
protein remain to be investigated.
PR-5-like allergens
Mal d 2 is an important allergen of apple that is associated with IgE-mediated
symptoms in apple-allergic individuals. The cDNA sequence of Mal d 2 was
34 Detecting allergens in food
PR-9: peroxidases
Molecular characteristics
Peroxidases are heme-containing enzymes that utilize H2O2 for a series of
oxidative reactions. Heme peroxidases include two superfamilies, one found
in bacteria, fungi, and plants, and the other found in animals. The first one
consists of three major classes.133 Class III includes the secretory plant
peroxidases that are monomeric glycoproteins and contain four conserved
disulfide bridges and two calcium ions. They share the same architecture,
two all alpha domains between which the heme group is located. Peroxidases
have been thoroughly researched in higher plants where their activities are
correlated with a large number of growth, developmental, and defense processes.
Specific lignin-forming peroxidases, induced by pathogens and involved in
plant defense against pathogens, have been designated PR-9.134
PR-9-like allergens
A prominent 36 kDa allergen was isolated from wheat flour and identified by
N-terminal and internal amino acid sequences as a peroxidase.135 This wheat
allergen, also referred to as Tri a Bd 36K, was characterized as a glycoprotein.
The carbohydrate moiety seemed to be at least partially involved in the IgE
binding.136 IgE of food-allergic patients bound to a glycosylated peroxidase
from tomato whose cDNA coded for seven potential N-linked glycosylation
sites.137 In general, the role of glycosylation in the allergenicity of plant
peroxidases has not yet been elucidated.
PR-10-like allergens
The oral allergy syndrome (OAS), an association of food allergies to fruits,
nuts, and vegetables in patients with pollen allergy, is the most frequent
clinical syndrome caused by cross-reactive IgE antibodies. In the majority of
the cases, OAS in individuals allergic to tree pollen is caused by IgE cross-
reactivity between the major birch pollen allergen Bet v 1 and its homologous
proteins. Bet v 1-homologous food allergens have been identified in fruits of
Rosaceae species (Mal d 1 in apple146, Pru av 1 in sweet cherry147, Pru ar 1
in apricot148, Pyr c 1 in pear149), or somatic tissues, vegetables of Apiaceae
species (Api g 1 in celery150, Dau c 1 in carrot151, pcPR1 and pcPR2 in
parsley152). Additional Bet v 1-homologous proteins capable of binding anti-
Bet v 1 IgE were described as pSTH-2 and pSTH-21 from potato.152 Recently,
the Bet v 1-related major allergen of hazelnuts Cor a 1.04 has been characterized
in detail.153 The Bet v 1 homologous allergen SAM22/Gly m 4 of soybean
was found to be responsible for inducing an OAS of extraordinary severity
and severe systemic reactions in some birch pollen allergic individuals.154
Allergenic profilins
In most cases, patients who are sensitized to pollen profilins characteristically
react with a wide range of profilins from nutritive allergen sources. Profilins
are involved in the celery–mugwort–spice syndrome.157 A hazelnut profilin,
Cor a 2, was identified as a relevant IgE-binding protein for a minority of
pollen–nut allergic individuals.158 Grass pollen profilin-allergic patients also
reacted to homologues in celery and carrots, Api g 4 and Dau c 4, respectively.159
36 Detecting allergens in food
Lectins
Molecular characteristics
Lectins, also known as plant agglutinins, are a class of proteins that bind to
specific sequences of sugar determinants on glycoproteins. Lectins are found
in seeds, especially those of legumes. Some lectins react unspecifically with
the carbohydrate moieties of IgE, induce histamine release and can thus
induce allergy-like symptoms.172
Allergenic lectin
The 31 kDa peanut agglutinin was identified as a lectin that is specifically
recognized by IgE from a minority of peanut allergic patients.173
Classifying food allergens 37
Parvalbumins
Parvalbumins constitute a class of calcium-binding proteins characterized by
the presence of several helix-loop-helix (EF-hand) motifs. They are present
in the white muscle of many fish species in relatively high amounts of up to
5 mg/g fresh weight. Parvalbumin is assumed to be important for the relaxation
of muscle fibers by binding free calcium in the cells.197 Parvalbumins are
heat stable and resistant to denaturation and proteolytic digestion. The major
allergenic protein of cod (Gadus callarias) is a 12.3 kDa parvalbumin. It was
named allergen M or Gad c 1 and has been intensively studied both structurally
and immunologically.198 Recently, two distinct parvalbumin transcripts were
identified in Atlantic cod suggesting that isotypic variants are generally present
in fish.199 Allergenic parvalbumins have also been described from a second
cod species Gadus morhua as Gad m 1,200 from the salmon Salmo salar as
Classifying food allergens 39
Sal s 1,201 from the carp Cyprinus carpio as Cyp c 1,202 from tuna,203 from
the edible frog Rana esculenta,204 and from mackerel.205
2.3.2 Tropomyosins
Tropomyosins are a family of closely related proteins present in muscle and
non-muscle cells.206 Tropomyosin plays a key regulatory role in muscle
contraction together with actin and myosin. In non-muscle cells, tropomyosin
is believed to play a role in the regulation of cell morphology and motility.
In muscle cells, two alpha-helical tropomyosin molecules are wound around
each other forming a parallel dimeric alpha-helical coiled-coil structure.
Tropomyosins have a sequence (284 amino acid residues long in most isoforms)
with a seven-residue (‘heptad’) repeat of the form a-b-c-d-e-f-g, where a and
d are generally apolar residues. Most isoforms have been shown to have an
unbroken series of 40 continuous heptads. Tropomyosin molecules bond
head-to-tail with a short overlap to form an unbroken coiled-coil cable that
winds around the actin helix.
Allergenic tropomyosins
Two invertebrate groups, Crustacea and Mollusca, are generally referred to
as shellfish and are common constituents in the diet of many populations.
Tropomyosins are heat stable cross-reactive food allergens present in
crustaceans and molluscs. Tropomyosins were identified as the major allergens
of shrimp by several laboratories. They include the allergenic tropomyosins
Pen i 1 from the Indian shrimp Penaeus indicus,207 Par f 1 from the most
common Taiwanese shrimp Parapenaeus fissurus ,208 Pen a 1 from the brown
shrimp Penaeus aztecus,209 and Met e 1 from Metapenaeus ensis.210 Additional
allergenic crustacean tropomyosins were described as Cha f 1 from the common
crab Charybdis feriatus,211 as Pan s 1 from the spiny lobster Panulirus
stimpsonii, 212 and as Hom a 1 from the American lobster Homarus
americanus.213 Allergenic tropomyosins are also common in molluscs including
Cra g 1214,215 and Cra g 2216 of the Pacific oyster Crassostrea gigas, Tur c 1
of the gastropod Turbo cornutus,217 Tod p 1 from the squid Todarodes
pacificus,218 Per v 1 from the tropical green mussel Perna viridis,219 Hel as
1 from the brown garden snail,220 and the allergenic tropomyosins of the
abalone Haliotis diversicolor and the scallop Chlamys nobilis.221
Arginine kinases
Arginine kinases catalyze the transfer of phosphate from ATP to arginine. An
arginine kinase of the shrimp species Parapenaeus fissurus was discovered
as a major and novel allergen of crustaceans and named Par f 1.223 The
homologous allergen was also isolated from the crab Portunus
trituberculotus.223 Another allergenic arginine kinase was identified from
the shrimp species Penaeus monodon by two-dimensional immunoblotting,
designated Pen m 2, and its cDNA was cloned.224 Additional allergenic
crustacean arginine kinases from the sand shrimp Metapenaeus ensis, the
lobster species Homarus gammarus, the crawfish species Metanephrops
thomsoni, and the crab species Scylla serrata were described.224
Alpha-lactalbumin, Bos d 4
Lactalbumin, which attaches to beta-galactosyltransferase to create the lactose
synthetase complex, is essential for milk production.227 Alpha-lactalbumin is
a monomeric globular calcium-binding metalloprotein of 14.4 kDa, with
four disulfide bridges.228 The calcium is bound in a loop that is superficially
similar to the classic EF-hand motif. Bovine alpha-lactalbumin is a major
cows’ milk allergen. Alpha-lactalbumin and hen’s egg white lysozyme are
closely related, having evolved from a common ancestral gene.229
Lysozyme, Gal d 4
Lysozyme is a muramidase that catalyzes the hydrolysis of beta-1,4-links
between N-acetyl-muramic acid and N-acetyl-D-glucosamine in the
peptidoglycan of bacterial cell walls. Egg white lysozyme, Gal d 4, is one of
the main allergens of hen’s egg.230
2.3.5 Lipocalins
The lipocalins are a diverse family of proteins comprising extracellular ligand-
binding proteins with high specificity for small hydrophobic molecules.231
These proteins transport pheromones or nutrients, control cell regulation, or
play a role in cryptic coloration or the enzymatic synthesis of prostaglandins.
The crystal structures of several lipocalins have been elucidated and show a
novel eight-stranded anti-parallel beta-barrel fold well conserved within the
family.
Classifying food allergens 41
Beta-lactoglobulin, Bos d 5
Beta-lactoglobulin (Bos d 5) is the major whey protein in the milk of ruminants
and many other mammals. It binds a wide variety of hydrophobic ligands,
but its function remains unknown. Bos d 5, a major cows’ milk allergen, is
absent from human breast milk. Bos d 5 occurs as a 36 kDa dimer. Each
subunit consists of 162 amino acid residues and possesses two disulfide
bonds and one free cysteine. The relative resistance of Bos d 5 to acid
hydrolysis as well as to proteases allows some of the protein to remain intact
after digestion. There are two main isoforms which differ only in two amino
acid positions.232 The crystal structure of Bos d 5 has been determined,
confirming its membership of the lipocalin protein family.233
2.3.7 Immunoglobulins
IgE directed against bovine IgG, Bos d 7, was detected in raw beef in 83%
of beef-allergic subjects but in only 24% of beef-tolerant subjects. Complete
inhibition of the IgE reactivity to the bovine IgG was obtained with lamb,
venison, and milk. Bovine IgG appears to be a major cross-reacting meat
allergen that could predict beef allergy.238 The role of Bos d 7 in cows’ milk
remains to be studied.
2.3.8 Alpha/beta-caseins
Caseins are the major protein constituent of milk. The biological function of
the caseins is considered as modulating the precipitation of calcium phosphate
from solution.239 The major allergens of the casein fraction are the calcium-
sensitive alpha-s1-, alpha-s2-, and beta-caseins (whole casein fraction = Bos
d 8).240,241 Alpha-s1 and alpha-s2 caseins from cows, goats, and sheep share
87–98% identical amino acids. The alpha-caseins from these animal species
are highly cross-reactive and consequently milk of sheep and goat is not a
suitable alternative for individuals suffering from cows’ milk allergy
42 Detecting allergens in food
(CMA).235 Oral challenge studies clearly showed that goats’ milk is not an
appropriate cows’ milk substitute for children with IgE-mediated CMA.236
In addition, IgE from children allergic to cows’ milk were capable of recognizing
most of the milk proteins from ewe, goat, and buffalo while no serum contained
IgE that reacted with camels’ milk proteins.242 Interestingly, mares’ milk can
be regarded as a good substitute for cows’ milk, having produced a positive
oral challenge in only 1/24 children with CMA.243
2.3.9 Transferrins
Transferrins are eukaryotic iron-binding glycoproteins that control the level
of free iron in biological fluids. The proteins have arisen by duplication of a
domain, each duplicated domain binding one iron atom. Members of the
family include milk lactotransferrin (lactoferrin) and egg white ovotransferrin
(conalbumin). The structural comparison of allergenic sites in alpha-lactalbumin
and beta-lactoglobulin with the structure of lactoferrin has shown that lactoferrin
also possesses allergenic sites.244 Lactoferrin reacted with IgE from 45% of
patients allergic to cows’ milk.241 Ovotransferrin from hen’s egg, also known
as Gal d 3 or conalbumin, has been identified as another major egg white
allergen.245
Ovomucoids
Avian ovomucoids contain three Kazal-like inhibitory domains.248 Chicken
ovomucoid has been shown to be the dominant hen’s egg white allergen Gal
d 1.190 Gal d 1 comprises 186 amino acid residues that are arranged in three
tandem domains (Gal d 1.1, Gal d 1.2, Gal d 1.3). Each domain contains
three intradomain disulfide bonds. Gal d 1.1 and Gal d 1.2 contain two
carbohydrate chains each, and about 50% of the Gal d 1.3 domains contain
one carbohydrate chain.189 Significantly more IgE from hen’s egg-allergic
patients reacts with the second ovomucoid domain. It has been further suggested
that conformational B-cell epitopes play a significant role in ovomucoid
allergenicity and that the carbohydrate moieties have a minor affect on
allergenicity.189
Serpins
The serpins (serine proteinase inhibitors) and related proteins constitute one
Classifying food allergens 43
Ovalbumin, Gal d 2
It has become clear that ovomucoid is the immunodominant protein fraction
in egg white and that the use of commercially purified ovalbumin has led to
an over-estimation of the dominance of ovalbumin as a major egg allergen in
human beings.190
2.6 Acknowledgement
This work was supported by the Austrian Science Fund grant SFB F018-02.
2.7 References
1. Murzin, A G, Brenner, S E, Hubbard, T and Chothia, C (1995) ‘SCOP: a structural
classification of proteins database for the investigation of sequences and structures’,
J Mol Biol, 247(4), 536–540.
2. Van Loon, L C and Van Strien, E A (1999) ‘The families of pathogenesis-related
proteins, their activities, and comparative analysis of PR-1 types proteins’, Physiol
Mol Plant Pathol, 55(1), 85–97.
3. Breiteneder, H and Ebner, C (2000) ‘Molecular and biochemical classification of
plant-derived food allergens’, Allergy Clin Immunol 106(1 Pt 1), 27–36.
4. Hoffmann-Sommergruber, K (2000) ‘Plant allergens and pathogenesis-related
proteins. What do they have in common ?’, Int Arch Allergy Immunol 122(3), 155–
166.
5. Dunwell, J M, Khuri, S and Gane, P J (2000) ‘Microbial relatives of the seed
storage proteins of higher plants: conservation of structure and diversification of
function during evolution of the cupin superfamily’, Microbiol Mol Biol Rev, 64(1),
153–179.
46 Detecting allergens in food
and immunoblotting in children with atopic dermatitis and positive soy challenges’,
J Allergy Clin Immunol, 81(6), 1135–1142.
23. Ogawa, T, Bando, N, Tsuji, H, Nishikawa, K and Kitamura, K (1995) ‘Alpha-
subunit of beta-conglycinin, an allergenic protein recognized by IgE antibodies of
soybean-sensitive patients with atopic dermatitis’, Biosci Biotechnol Biochem, 5(5),
831–833.
24. Teuber, S S, Jarvis, K C, Dandekar, A M, Peterson, W R and Ansari, A A (1999)
‘Identification and cloning of a complementary DNA encoding a vicilin-like
proprotein, Jug r 2, from English walnut kernel (Juglans regia), a major food
allergen’, J Allergy Clin Immunol, 104(6), 1311–1320.
25. Sanchez-Monge, R, Pascual, C Y, Diaz-Perales, A, Fernandez-Crespo, J, Martin-
Esteban, M and Salcedo, G (2000) ‘Isolation and characterization of relevant allergens
from boiled lentils’, J Allergy Clin Immunol, 106(5), 955–961.
26. Wang, F, Robotham, J M, Teuber, S S, Tawde, P, Sathe, S K, Roux, K H (2002)
‘Ana o 1, a cashew (Anacardium occidental) allergen of the vicilin seed storage
protein family’, J Allergy Clin Immunol, 110(1), 160–166.
27 Beyer, K, Bardina, L, Grishina, G and Sampson, H A, ‘Identification of sesame
seed allergens by 2-dimensional proteomics and Edman sequencing: seed storage
proteins as common food allergens’, J Allergy Clin Immunol, 110(1), 154–159.
28. Müntz, K, Jung, R and Saalbach, G (1993) ‘Synthesis, processing and targeting of
legume seed proteins’, in Shewry, P, Prand, W and Stobart, A K (eds), Seed Storage
Compounds: Biosynthesis, Interactions, and Manipulation, Oxford University Press,
128–146.
29. Adachi, M, Takenaka, Y, Gidamis, AB, Mikami, B and Utsumi, S (2001) ‘Crystal
structure of soybean proglycinin A1aB1b homotrimer’, J Mol Biol, 305(2), 291–
305.
30. Rabjohn, P, Helm, E M, Stanley, J S, West, C M, Sampson, H A, Burks, A W and
Bannon, G A (1999) ‘Molecular cloning and epitope analysis of the peanut allergen
Ara h 3’, J Clin Invest, 103(4), 535–542.
31. Rabjohn, P, West, C M, Connaughton, C, Sampson, H A, Helm, R M, Burks, A W
and Bannon, G A (2002) ‘Modification of peanut allergen Ara h 3: effects on IgE
binding and T cell stimulation’, Int Arch Allergy Immunol, 128(1), 15–23.
32. Kleber-Janke, T, Crameri, R, Appenzeller, U, Schlaak, M and Becker, W M (1999)
‘Selective cloning of peanut allergens, including profilin and 2S albumins, by
phage display technology’, Int Arch Allergy Immunol, 19(4), 265–274.
33. Koppelman, S J, Knol, E F, Vlooswijk, R A, Wensing, M, Knulst, A C, Hefle, S L,
Gruppen, H and Piersma, S (2003) ‘Peanut allergen Ara h 3: isolation from peanuts
and biochemical characterization’, Allergy, 58(11), 1144–1151.
34. Beardslee, T A, Zeece, M G, Sarath, G and Markwell, J P (2000) ‘Soybean glycinin
G1 acidic chain shares IgE epitopes with peanut allergen Ara h 3’, Int Arch Allergy
Immunol, 123(4), 299–307.
35. Helm, R M, Cockrell, G, Connaughton, C, Sampson, H A, Bannon, G A, Beilinson,
V, Livingstone, D, Nielsen, N C and Burks, A W (2000) ‘A soybean G2 glycinin
allergen. 1. Identification and characterization’, Int Arch Allergy Immunol, 123(3),
205–212.
36. Helm, R M, Cockrell, G, Connaughton, C, Sampson, H A, Bannon, G A, Beilinson,
V, Nielsen, N C and Burks, A W (2000) ‘A soybean G2 glycinin allergen. 2.
Epitope mapping and three-dimensional modeling’, Int Arch Allergy Immunol,
123(3), 213–219.
37. Beyer, K, Grishina, G, Bardina, L, Grishin, A and Sampson, H A (2002) ‘Identification
of an 11S globulin as a major hazelnut food allergen in hazelnut-induced systemic
reactions’, J Allergy Clin Immunol, 110(3), 517–523.
38. Teuber, S S and Peterson, W R (1999) ‘Systemic allergic reaction to coconut
(Cocos nucifera) in 2 subjects with hypersensitivity to tree nut and demonstration
48 Detecting allergens in food
and Bol, J F (1991) ‘Tobacco and tomato PR proteins homologous to win and pro-
hevein lack the ‘hevein’ domain’, Mol Plant Microbe Inter, 4(6), 586–592.
121. Hanninen, A R, Mikkola, J H, Kalkkinen, N, Turjanmaa, K, Ylitalo, L, Reunala, T
and Palosuo, T (1999) ‘Increased allergen production in turnip (Brassica rapa) by
treatments activating defense mechanisms’, J Allergy Clin Immunol, 104(1), 194–
201.
122. Van Damme, E J, Charels, D, Roy, S, Tierens, K, Barre, A, Martins, J C, Rouge, P,
Van Leuven, F, Does, M and Peumans, W J (1999) ‘A gene encoding a hevein-like
protein from elderberry fruits is homologous to PR-4 and class V chitinase genes’,
Plant Physiol, 119(4), 1547–1556.
123. Dudler, R, Mauch, F and Reimmann, C (1994) Thaumatin-like proteins, in Witty,
M, Higginbotham, J D (eds), Thaumatin, Boca Raton, FL, CRC Press Inc, 93–199.
124. Koiwa, H, Kato, H, Nakatsu, T, Oda, J, Yamada, Y and Sato, F (1999) ‘Crystal
structure of tobacco PR-5d protein at 1.8 A resolution reveals a conserved acidic
cleft structure in antifungal thaumatin-like proteins’, J Mol Biol, 286(4), 1137–
1145.
125. Batalia, M A, Monzingo, A F, Ernst, S, Roberts, W and Robertus, J D (1996) ‘The
crystal structure of the antifungal protein zeamatin, a member of the thaumatin-
like, PR-5 protein family’, Nat Struct Biol, 3(1), 19–23.
126. Ogata, C M, Gordon, P F, de Vos, A M and Kim, S H (1992) ‘Crystal structure of
a sweet tasting protein thaumatin I, at 1.65 A resolution’, J Mol Biol, 228(3), 893–
908.
127. Krebitz, M, Wagner, B, Ferreira, F, Peterbauer, C, Campillo, N, Witty, M, Kolarich,
D, Steinkellner, H, Scheiner, O and Breiteneder, H (2003) ‘Plant-based heterologous
expression of Mal d 2, a thaumatin-like protein and allergen of apple (Malus
domestica), and its characterization as an antifungal protein, J Mol Biol, 329(4),
721–730.
128. Inschlag, C, Hoffmann-Sommergruber, K, O’Riordain, G, Ahorn, H, Ebner, C,
Scheiner, O and Breiteneder, H (1998) ‘Biochemical characterization of Pru a 2, a
23-kD thaumatin-like protein representing a potential major allergen in cherry
(Prunus avium)’, Int Arch Allergy Immunol, 116(1) 22–28.
129. Jensen-Jarolim, E, Santner, B, Leitner, A, Grimm, R, Scheiner, O, Ebner, C and
Breiteneder, H (1998) ‘Bell peppers (Capsicum annuum) express allergens (profilin,
pathogenesis-related protein P23 and Bet v 1) depending on the horticultural strain’,
Int Arch Allergy Immunol, 116(2), 103–109.
130. Fuchs, H C, Hoffmann-Sommergruber, K, Wagner, B, Krebitz, M, Scheiner, O and
Breiteneder, H (2002) ‘Heterologous expression in Nicotiana benthamiana of Cap
a 1, a thaumatin-like protein and major allergen from bell pepper (Capsicum annuum)’,
J Allergy Clin Immunol, 109(S), 134–135.
131. Pastorello, E A, Farioli, L, Pravettoni, V, Ortolani, C, Fortunato, D, Giuffrida, M G,
Perono Garoffo, L, Calamari, A M, Brenna, O and Conti, A (2003) ‘Identification
of grape and wine allergens as an endochitinase 4, a lipid-transfer protein, and a
thaumatin’, J Allergy Clin Immunol, 111(2), 350–359.
132. Gavrovic-Jankulovic, M, Cirkovic, T, Vuckovic, O, Atanaskovic-Markovic, M,
Petersen, A, Gojgic, G, Burazer, L and Jankov, R M (2002) ‘Isolation and biochemical
characterization of a thaumatin-like kiwi allergen’, J Allergy Clin Immunol 110(5),
805–810.
133. Welinder, K G (1992) ‘Superfamily of plant, fungal and bacterial peroxidases’,
Curr Opin Struct Biol, 2(3), 388–393.
134. Van Loon, L C, Pierpoint, W S, Boller, T and Conejero, V (1994) ‘Recommendations
for naming plant pathogenesis-related proteins’, Plant Mol Biol Rep, 12(2), 245–
264.
135. Sánchez-Monge, R, García-Casado, G, López-Otín, C, Armentia, A and Salcedo,
G (1997) ‘Wheat flour peroxidase is a prominent allergen associated with baker’s
asthma’, Clin Exp Allergy, 27(10), 1130–1137.
54 Detecting allergens in food
221. Chu, K H, Wong, S H and Leung, P S (2000) ‘Tropomyosin is the major mollusk
allergen: reverse transcriptase polymerase chain reaction, expression and IgE
reactivity’, Mar Biotechnol (NY), 2(5), 499–509.
222. Strong, S J and Ellington, W R (1995) ‘Isolation and sequence analysis of the gene
for arginine kinase from the chelicerate arthropod, Limulus polyphemus: insights
into catalytically important residues’, Biochim Biophys Acta 1246(2), 197–200.
223. Lin, R Y, Shen, H and Han, S H (1993 ‘Identification and characterization of a 30
kd major allergen from Parapenaeus fissurus’, J Allergy Clin Immunol 92(6), 837–
845.
224. Yu, C J, Lin, Y F, Chiang, B L and Chow, L P (2003) ‘Proteomics and immunological
analysis of a novel shrimp allergen, Pen m 2’, J Immunol 170(1), 445–453.
225. Henrissat, B and Bairoch, A (1993) ‘New families in the classification of glycosyl
hydrolases based on amino acid sequence similarities’, Biochem J 293 (Pt 3), 781–
788.
226. Henrissat, B and Bairoch, A (1996) ‘Updating the sequence-based classification of
glycosyl hydrolases’, Biochem J 316 (Pt 2), 695–696.
227. Shewale, J G, Sinha, S K and Brew, K (1984) ‘Evolution of alpha-lactalbumins.
The complete amino acid sequence of the alpha-lactalbumin from a marsupial
(Macropus rufogriseus) and corrections to regions of sequence in bovine and goat
alpha-lactalbumins’, J Biol Chem 259(8), 4947–4956.
228. Brew, K, Castellino, F J, Vanaman, T C and Hill, R L (1970) ‘The complete amino
acid sequence of bovine alpha-lactalbumin’, J Biol Chem 245(17), 4570–4582.
229. Nitta, K and Sugai, S (1989) ‘The evolution of lysozyme and alpha-lactalbumin’,
Eur J Biochem 182(1), 111–118.
230. Anet, J, Back, J F, Baker, R S, Barnett, D, Burley, R W and Howden, M E (1985)
‘Allergens in the white and yolk of hen’s egg. A study of IgE binding by egg
proteins’, Int Arch Allergy Appl Immunol 77(3), 364–371.
231. Pervaiz, S and Brew, K (1985) ‘Homology of beta-lactoglobulin, serum retinol-
binding protein, and protein HC’, Science 228(4697), 335–337.
232. Jamieson, A C, Vandeyar, M A, Kang, Y C, Kinsella, J E and Batt, C A (1987)
‘Cloning and nucleotide sequence of the bovine beta-lactoglobulin gene’, Gene
61(1), 85–90.
233. Brownlow, S, Morais Cabral, J H, Cooper, R, Flower, D R, Yewdall, S J, Polikarpov,
I, North, A C and Sawyer, L (1997) ‘Bovine beta-lactoglobulin at 1.8 A resolution
– still an enigmatic lipocalin’, Structure 5(4), 481–495.
234. He, X M and Carter, D C (1992) ‘Atomic structure and chemistry of human serum
albumin’, Nature 358(6383), 209–215.
235. Spuergin, P, Walter, M, Schiltz, E, Deichmann, K, Forster, J and Mueller, H (1997)
‘Allergenicity of alpha-caseins from cow, sheep, and goat’, Allergy, 52(3), 293–
298.
236. Bellioni-Businco, B, Paganelli, R, Lucenti, P, Giampietro, P G, Perborn, H and
Businco, L (1999) ‘Allergenicity of goat’s milk in children with cow’s milk allergy’,
J Allergy Clin Immunol, 103(6), 1191–1194.
237. Quirce, S, Maranon, F, Umpierrez, A, de las Heras, M, Fernandez-Caldas, E and
Sastre, J (2001) ‘Chicken serum albumin (Gal d 5) is a partially heat-labile
inhalant and food allergen implicated in the bird-egg syndrome’, Allergy 56(8),
754–762.
238. Ayuso, R, Lehrer, S B, Lopez, M, Reese, G, Ibanez, M D, Esteban, M M, Ownby,
D R and Schwartz, H (2000) ‘Identification of bovine IgG as a major cross-reactive
vertebrate meat allergen’, Allergy 55(4), 348–354.
239. Holt, C and Sawyer, L (1988) ‘Primary and predicted secondary structures of the
caseins in relation to their biological functions’, Protein Eng 2(4), 251–259.
240. Wal, J M (2001) ‘Structure and function of milk allergens’, Allergy, 56(Suppl67),
35–38.
60 Detecting allergens in food
characterization and in vitro efficacy of the major peanut allergens for use in
immunotherapy’, Int Arch Allergy Immunol, 124(1–3), 70–72.
259. Rabjohn, P, West, C M, Connaughton, C, Sampson, H A, Helm, R M, Burks, A W
and Bannon, G A (2002) ‘Modification of peanut allergen Ara h 3: effects on IgE
binding and T cell stimulation’, Int Arch Allergy Immunol, 128(1), 15–23.
260. Cocco, R R, Jarvinen, K M, Sampson, H A and Beyer, K (2003) ‘Mutational
analysis of major, sequential IgE-binding epitopes in alpha-s1-casein, a major
cow’s milk allergen’, J Allergy Clin Immunol, 112(2), 433–437.
261. Valenta, R and Kraft, D (2002) ‘From allergen structure to new forms of allergen-
specific immunotherapy’, Curr Opin Immunol, 14(6) 718–727.
262. Brusic, V, Millot, M, Petrovsky, N, Gendel, S M, Gigonzac, O and Stelman, S J
(2003) ‘Allergen databases’, Allergy 58(11), 1093–1100.
Part II
Antibodies
S. Hefle, University of Nebraska, USA, J. Yeung, Food
Products Association, USA and R. Helm, University of Arkansas,
USA
F(ab′)2
Fab
Papain
Pepsin
Fc
Fc′
Fig. 3.1 Antibody (immunoglobulin) structure showing heavy and light chains.
Papain and pepsin cleavage sites are also indicated. Digestion by papain gives Fc and
Fab fragments. Digestion by pepsin separates the molecule into two parts, a bivalent
fraction called F(ab′)2 that contains both antigen binding sites, but also the Fc into
small parts called Fc′.
3.3.2 Adjuvants
Often the use of adjuvants is necessary for antibody production. The function
of an adjuvant is to enhance the immunogenicity of the antigen by increasing
the immune response in several ways; it increases the efficiency of antigen
presentation and the number of antibody-secreting B cells, acts as an
immunogen depot (for extended antigen stimulation), protects the immunogen
from rapid removal and catabolism, and increases the affinity and avidity of
70 Detecting allergens in food
the antibody response (Spier and Griffiths, 1985; Catty, 1988; Roitt et al.,
1989). Adjuvants are available in many forms (Harlow and Lane, 1988). The
best known adjuvant is complete Freund’s adjuvant (CFA), a suspension of
killed mycobacteria in mineral oil which was first described over 50 years
ago. Incomplete Freund’s adjuvant (IFA) is the mineral oil minus the bacteria;
sometimes IFA can be used successfully by itself (Campbell, 1996). There
are many other types of adjuvants that can be used besides CFA and IFA
(Hefle, 1995; Campbell, 1996).
3.3.4 Dose
The dose of immunogen depends on the its nature and also on the animal
being used. Large doses may induce tolerance, but in some cases
hyperimmunization schedules utilizing a relatively high amount of immunogen
have been successful (Cordle et al., 1991). The desired result is an antibody
with high avidity and affinity; therefore, use of the lowest amount of immunogen
necessary to achieve antibody production is usually the best approach. A
typical dose for rabbits is 100 μg for the primary injection, with 50–100 μg
used for booster injections (Harlow and Lane, 1988), although amounts well
above that have been reported and can be used. For larger animals, 500–
1000 μg is usual to start with. For booster injections, the traditional amount
used is 10–50% of the primary dose. Smaller booster doses promote clonal
selection for high-affinity Ab. For antigens in precious supply, one or two
Antibodies 71
injections with a low dose to prime the animals followed by a larger booster
injection can be successful (Harlow and Lane, 1988).
A specific antibody in serum can usually be detected about a week after
the initial immunization. The first type of antibody produced is IgM with
high avidity but low affinity (Deshpande, 1996c), but then a switch to IgG of
lower avidity, but higher affinity, occurs (Fig. 3.2). Responses to booster
injections are an increase in titer; as the time of immunization extends,
antibodies of the IgG class dominate. In general, a three to four week interval
between the primary injection and a booster injection is recommended
(Deshpande, 1996c). Usually, high titer is attained in about two to four
weeks after booster injections. The immune response matures, producing
higher-affinity and avidity antibodies as time goes on. For this reason, it is
possible to pool polyclonal antibodies after a certain amount of time, rather
than including early bleeds. Although intervals between booster injections
can be varied, there should be adequate time allowed for the circulating
antibody level to drop low enough to prevent prompt clearance of the injected
immunogen. For IgG, the half life is 20–25 days in the blood circulation, and
the clearance is 10–15% per day (Cruse and Lewis, 1999). As a result, to
avoid this problem, booster injections should be done every four to six weeks.
The quality of the antiserum is more important in immunoassays than in
other techniques. During immunization, antibody production and quality
(titer) needs to be monitored. Serum samples are collected 7–14 days after
booster injection, which corresponds to peak IgG production rate. For polyclonal
antibody production, test bleeds (small volume) are taken until sufficient
titer is reached, at which time production bleeds (larger volume) are done.
Serum samples from individual animals should not be pooled in the initial
stages; once a good titer and affinity have been developed, individual bleeds
can then be pooled to form a larger quantity of homogeneous antiserum.
Titers are usually monitored using indirect ELISA, in which the antigen is
72 Detecting allergens in food
coated onto microtiter plates and the antiserum is diluted serially and added
to plates. Titer can be defined in many ways, but one common method is the
mid-linear point of the titration curve. Antigen-specific IgG of approximately
8–14 mg/mL serum can be obtained.
Antibody affinity can also be ascertained using indirect ELISA, but using
the analyte of interest as an inhibitor. The dilution of antibody used in the
ELISA is the dilution identified by the titer. The horizontal displacement of
the inhibitor curve is an indirect measure of the affinity of the antibody
(Morris, 1985), as the greater the displacement, the greater the affinity of the
antibody. Cross-reactivity of the antibody can also be determined using a
similar technique; the curve generated from other substances should be able
to be superimposed on the antibody dilution curve.
3.3.6 Cross-reactivity
Antigen–antibody reactions show a high degree of specificity when the binding
sites of the antibodies are not complementary and thus do not recognize or
Antibodies 73
Antigen
Polyclonal serum
antibodies have
variable affinity
for epitope
Myeloma PEG B cells from
cell line spleen
Propagate
3.3.8 Purification
Antiserum either from polyclonal antiserum or monoclonal culture supernatant
or ascites can be further purified. Often, crude serum or semi-purified serum
can be used, but in some cases further purification is recommended or necessary.
Antibodies 75
and vimentin (Blose et al., 1982). Lane and Koprowski (1982) have suggested
two possible causes for monoclonal antibody cross-reaction; that the antibody
detects structural similarities in the two antigens, or that the binding site of
the antibody could combine with unrelated antigens in a multispecific way,
and recognize two or more entirely different epitopes (Richards et al., 1975).
An antigen–antibody complex occurs whenever there are enough and the
right strength of inter- (and intra-) molecular forces. Goding (1996) states
that it is not surprising that monoclonal antibodies can have multispecificity
and, indeed, Hefle et al. (1994) noted cross-reactivity among their developed
monoclonal antibodies, to different peanut proteins. Unusual cross-reactions
are almost never seen with polyclonal antibodies because the odd cross-
reaction is ‘diluted out’ in the enormity of the rest of the response and the
‘consensus’ of different clones (Goding, 1996). Specificity also depends on
this consensus and polyclonal antibodies bind to determinants that cover
almost the entire external surface of the antigen (Benjamin et al., 1984).
Therefore, minute changes in the structure of the antigen due to genetic
polymorphism, heterogeneity of glycosylation, or slight denaturation will
usually have little or no effect on polyclonal antibody binding. A subset of
polyclonal antibodies will usually bind to antigen that has been modified or
denatured, even if this was not the form of the immunogen used for the
immunization (Burnette, 1981). In contrast, monoclonal antibodies most
often bind to a single unique epitope – if for any reason this site is altered
(due to denaturation, unfolding, aggregation, formation of Maillard reaction
products, etc.), the monoclonal antibody may not bind (Goding, 1996). Another
potential drawback to monoclonal antibodies is that they may only be able to
cross-link antigen molecules with two or more binding sites (Goding, 1996),
although this requirement is usually not an issue for allergenic proteins. If
cross-reactivity is present with monoclonal antibodies, it is hard to remove,
unlike for polyclonal antibodies. Monoclonal antibodies have sensitivity to
only part of the total antigenicity of the immunogen, resulting in poorer
performance compared to that of polyclonal antibodies in many applications.
Polyclonal antibodies may be generated in a great variety of species, including
rabbit, goat, sheep, donkey and chicken, giving the immunoassay developer
many choices in design of assay; this is in contrast to monoclonal antibodies,
which are usually only produced in rodents due to practical considerations.
It can be difficult to obtain polyclonal antisera with identical properties
from different animals. Even from the same animal, polyclonal antisera
collected at different times can have different properties. These limitations
of polyclonal antibodies can justify producing monoclonal antibodies, as the
latter have identical physical, biochemical and immunological properties.
Due to their specificity, monoclonal antibodies are good for use as a primary
antibody in immunoassays. In addition, the specificity of monoclonal antibodies
spurs efficient binding of antigen in a mixture of related molecules, and this
property can be exploited in such techniques as affinity purification. A
disadvantage of monoclonal antibodies is that their preparation can be time-
Antibodies 77
3.5 References
Benjamin, D C, Berzofsky, J A, Esat, I J, Gurd, F R N, Hannum, C, Leach, S J, Margoliash,
E, Michael, J G, Miller, A, Prager, E M, Reichlin, M, Sercarz, E E, Smith-Gill, S J,
Todd, P E and Wilson, A C (1984) ‘The antigenic structure of proteins: a reappraisal’,
Ann Rev Immunol, 2, 67–101.
Blose, S H, Matsumara, F and Lin, J J C (1982) ‘Structure of vimentin 10-nm filaments
probed with a monoclonal antibody that recognizes a common antigenic determinant
on vimentin and tropomyosin’, Cold Spring Harbor Symp Quant Biol, 46, 455–463.
Burnette, W N (1981) ‘“ Western blotting”; electrophoretic transfer of proteins from
sodium dodecyl sulfate-polyacrylamide gels to unmodified nitrocellulose and
radiographic detection with antibody and radioiodinated protein A’, Anal Biochem,
112, 195–203.
Campbell, A M (1996) Production and purification of antibodies, in Diamands, E P and
Christopoulos, T K (eds), Immunoassay, San Diego, CA, Academic Press, 95–115.
Catty, D (1988) Antibodies, a Practical Approach, Vol 1, Oxford, UK, IRL Press.
Cordle, C T, Mahmoud, M I and Moore, V (1991) ‘Immunogenicity evaluation of protein
hydrolysates for hypoallergenic infant formulae’, J Pediatr Gastroenterol Nutr, 13,
270–276.
Cruse, J M and Lewis, R E (1999) ‘Atlas of Immunology’, Washington, DC, CRC Press,
127–142.
Davies, D R and Padlan, E A (1990) ‘Antibody–antigen complexes’, Ann Rev Biochem,
59, 439–473.
Deshpande, S S (1996a) ‘Antibodies: biochemistry, structure, and function’, in Deshpande,
S S, Enzyme Immunoasssays: From Concept to Product Development, New York, NY,
Chapman and Hall, 24–51.
Deshpande, S S (1996b) ‘Antigen-antibody reactions’, in Deshpande S S, Enzyme
Immunoasssays: From Concept to Product Development New York, NY, Chapman and
Hall, 52–71.
Deshpande, S S (1996c) ‘Antibody production’, in Deshpande S S, Enzyme Immunoasssays:
From Concept to Product Development, New York, NY, Chapman and Hall, 117–154.
Feldkamp, C S and Carey, J S (1996) ‘Immune function and antibody structure’, in
Diamands, E P and Christopoulos, T K (eds), Immunoassay, San Diego, CA, Academic
Press, 5–24.
Goding, J W (1986) Monoclonal Antibodies: Principles and Practice, 2nd edn, London,
UK, Academic Press, 3–5, 45–103.
Harlow, E and Lane, D (1988) Antibodies: A Laboratory Manual, Cold Spring Harbor,
NY, Cold Spring Harbor Laboratory Press.
Harlow, E and Lane, D (1999) Antigen-antibody interactions, in Harlow, E and Lane, D,
Using Antibodies, A Laboratory Manual, Cold Spring Harbor, NY, Cold Spring Harbor
Laboratory Press, 23–37.
Hawcroft, D, Hector, T and Rowell, F (1987) Quantitative Bioassay, New York, NY, John
Wiley & Sons, Inc.
Hefle, S L (1995) ‘Immunoassay fundamentals’, Food Technol, 49, 102–107.
Hefle, S L, Folgert, J P, Chu, F S and Bush, R K (1994) ‘Monoclonal antibodies against
selected peanut allergens: production and use as affinity agents, Food Agric Immunol,
6, 197–208.
Hermann, S H and Mescher, M F (1979) ‘Purification of the H-2K k molecule of the
murine major histocompatibility complex’, J Biol Chem, 254, 8713–8716.
78 Detecting allergens in food
4.1 Introduction
Allergens are foreign proteins or immunogens that, when introduced in an
immunocompetent and predisposed host, elicit the formation of IgE antibodies.
Once induced, IgE circulates in the blood and binds on to high-affinity IgE
Fcε receptors on mast cells and basophils. This process leads to a state of
sensitization that is evidenced by the detection of allergen-specific IgE antibody
on skin mast cells and blood basophils. Upon further allergen exposure by
ingestion, injection or inhalation, allergen can cross-link receptor bound IgE
antibody, causing degranulation of vasoactive mediators from mast cells and
leading to a spectrum of allergic reactions. Allergen-specific IgE antibody is
thus the key analyte in both in vivo – skin test, basophil meditator release –
and in vitro radioallergosorbent (RAST)/fluorescent enzyme immunoassay
(FEIA)/enzyme-linked immunosorbent assay (ELISA) – immunoblot –
inhibition assays that permits qualitative identification and quantitative
measurement of allergens in complex biological substances such as foods.1
In vivo, allergen that is injected into the skin of a sensitized individual
induces a wheal and flare reaction that can be measured and used to identify
the presence and quantify the allergen’s potency. Alternatively, controlled
ingestion of foods in a double-blind, placebo-controlled manner allows in
vivo food challenges which many allergists consider a definitive diagnostic
test for an individual’s sensitization to a particular food allergen. Due to its
relative hazard, however, this method is rarely used to identify or quantify
the levels of allergenic protein in particular foods. In vitro, allergen can
competitively inhibit the binding of allergen-specific IgE antibody to solid
phase allergen in the RAST2 and its newer non-isotopic counterparts such as
80 Detecting allergens in food
4.2.1 Reagents
Food allergen extracts
While virtually any food can cause allergic symptoms, relatively few foods
reportedly induce allergic reactions with any frequency in humans. The
commonly implicated allergenic foods include cows’ milk, egg, wheat, soy,
peanut, tree nuts, fish and crustacean shellfish.6 Diagnostic skin test extracts
are commonly prepared from fresh or cooked foods in a defined weight
versus physiological extraction buffer ratio (e.g. 1:20 w/v). They are sometimes
defatted, concentrated and put into 50% glycerin to minimize degradation
during storage. Except in the case of recombinant and some purified research
food allergens, diagnostic food allergen extracts tend to be crude allergenic
mixtures that display multiple protein and glycoprotein bands on
polyacrylamide gel electrophoresis and isoelectric focusing. Food allergens
contain linear allergenic epitopes that bind to IgE antibody and appear highly
resistant to denaturation by heat and proteases and conformational epitopes
that require three-dimensional folding of the allergenic protein to permit
binding to IgE antibodies. A comprehensive listing of food allergen specificities
together with their genus and species is presented elsewhere.7
Allergen-specific human IgE antibody-based analysis of food 81
Reference materials
Since there are few established diagnostic food allergen reference preparations,
most materials that are used as reference or calibration reagents in IgE-based
quantitative assays have been through a previous allergenic potency assessment
by the in vivo or in vitro methods described in this chapter. They are often
first characterized by separation and identification of their component proteins
using sodium dodecyl sulphate (SDS) polyacrylamide gel electrophoresis
and isoelectric focusing. Their total protein concentration is also determined
by one of several assays. They are then analyzed in an in vitro inhibition
assay for the purpose of determining the relative allergenic potency of test
allergen extracts, presumably with the same allergenic protein profile.
1:10
1:100
1:1000
1:10 000
Fig. 4.1 Intradermal skin test titration in which the photograph was taken 20 minutes
(right side) and five hours (left side) after the administration of four ten-fold
concentrations of allergen to the forearm of a sensitized individual. The swelling
(wheal) and redness (erythema) on the right side of the arm after 20 minutes identify
the titer or the most dilute concentration of allergen that produces a positive skin
reaction in comparison to a saline control. Reproduced with permission from
Roitt et al.25
extract of defined allergenic potency. Often the allergen content in the reference
material is reported in arbitrary units such as AUs (allergen units), BAUs
(biological allergen units) or RP units. Occasionally, the reference extract’s
total protein has been quantified using the modified ninhydrin assay12 or the
primary allergen content of the extract. Allergen levels in the test extracts are
interpolated and reported in relationship to these arbitrary units in the reference
extract.
Analytical specificity
The allergenicity of a substance depends on the extent of its foreigness (size,
stability, 1°, 2°, and 3° protein structure), the extent of the individual’s
exposure (concentration) and the genetic (atopic) predisposition of the exposed
individual. Plant-derived food allergens have been classified into discrete
allergen groups that explain the molecular basis of the cross-reactivity seen
among different food groups13 (see Chapter 2). Some individuals, for instance,
who develop an IgE antibody response to proteins in natural rubber latex
from Hevea brasiliensis (Hev b) trees also exhibit an oral allergy syndrome
when they eat certain foods such as avocados, kiwi, bananas and chestnuts.
This has been called the latex–fruit syndrome and it stems from cross-reactivity
of IgE antibodies to proteins in these foods and natural rubber latex14. For
example, the class I chitinases from the pathogenesis-related (PR) group 4
84 Detecting allergens in food
Variability
Both repeatability (intra-assay) and reproducibility (inter-assay) should be
maximized by applying each skin test dilution in duplicate. The reproducibility
reported in the parallel-line skin test assay using the intradermal method of
application was 14.2% coefficient of variation (mean +/– 2SD = 102 +/–
14.8 RP).8 Since the slope of the puncture skin test dose–response line is
significantly flatter than the intradermally applied skin test results, there is
increased variability of an assay using the puncture skin testing application
method.
4.3.1 Reagents
There are a number of required reagents that are needed to perform the
competitive RAST inhibition analysis. These include the allergosorbent that
is composed of a solid-phase matrix and an allergen component, allergen-
specific IgE antibody containing human serum, a radioisotope-, enzyme- or
fluorophor-labelled anti-human IgE detection reagent and characterized allergen
calibrators.
Allergen
There are several hundred food allergen specificities that are known to induce
IgE antibody responses in humans. Of these, there are a handful of allergen
specificities that produce much of the food allergy symptoms involving the
GI tract (vomiting, diarrhoea), respiratory tract (pneumonitis, asthma) and
skin (chronic urticaria, atopic dermatitis). These include cows’ milk, chicken
egg, wheat, soybean, fish, crustacean shellfish, peanuts, tree nuts, cereal
grains and citrus fruits. In vitro assay manufacturers have classified them as
a group with the letter F (for foods) and a sequential number that has no
significance other than when it was identified as an allergen source. For
instance F13 is the peanut specificity that is prepared from raw and shelled
Arachis hypogaea. Within the food group, the allergens can be segregated
into subgroups, such as dairy, fish, grains/grass, seed/nut, legume, crustacean,
vegetable, meat, fowl, mollusk, fungi, fruit, spice and stimulant (chocolate,
coffee). A number of complex food allergen extracts have been further
characterized with immunochemical methods so that individual allergenic
proteins have been identified. For instance, five allergens have been identified
in chicken egg white: ovomucoid (Gal d 1), ovoalbumin or conalbumin (Gal
d 2), ovotransferrin (Gal d 3), lysozyme (Gal d 4) and chicken serum albumin
(Gal d 5).17,18
For the purposes of this discussion, food extracts should be considered
complex protein mixtures. They are prepared by selecting fresh or cooked
food, grinding it up and preparing a physiological extract that is sometimes
defatted and then centrifuged and sterile filtered. Initial quality control of the
crude extract begins with analysis in one of several immunochemical methods
(isoelectric focusing immunoblot, SDS-polyacrylamide gel electrophoresis
separation followed by Western blot analysis, competitive binding
immunoassay) to ensure that it contains allergenic proteins. Allergen extracts
are subject to contamination, inherent biological variation and laboratory
misclassification. The use of fresh or cooked food, the precise method and
buffer used in extraction and the subsequent processing (column
chromatography, precipitation steps) can lead to highly variable extracts.
Stability during storage and following chemical manipulations such as coupling
onto a solid support or labelling with biotin can add to the heterogeneity.
Allergen-specific human IgE antibody-based analysis of food 87
Allergosorbent
The solid phase allergen or allergosorbent is one of the principal components
of the competitive RAST inhibition assay. The earliest RAST assay used
paper discs that were activated with cyanogen bromide to covalently couple
proteins. Because the paper disc was a defined size that fit into a 12 × 75 mm
test tube, it was difficult to vary the allergen amount bound to the disc. This
led researchers to covalently couple allergenic proteins to a variety of mobile
cyanogens/bromide-activated carbohydrate-based particles such as cellulose
(Avicel®/FMC BioPolymer, Philadelphia, PA) and cross-linked agarose
(SepharoseTM CL-4B; GE Healthcare AB, Uppsala, Sweden).19,20 Allergenic
proteins have also been coupled to cellulose threads, immobilon or nitrocellulose
membranes or more recently to a high binding capacity immobile sponge
that is used in the Pharmacia CAP SystemTM.3 Diagnostic Products Corporation
has chosen to biotinylate allergen mixtures and then insolubilize them on an
avidin-coated solid phase.5 In each of these cases, the covalent binding
process has permitted the immobilization of proteins of widely varying
molecular weights and isoelectric points to a single solid phase. Ideally, the
allergen concentration on the solid phase should be in molar excess to the
amount of allergen-specific IgE antibody present in the human serum.
Some commercially-available allergosorbents have also been prepared
with food allergen mixes. For instance, the F × 5E from Pharmacia contains
major allergens from the five foods (chicken egg white, cows’ milk, fish
[Gadus morhua], wheat [Triticum aestivum], peanut [Arachis hypogaea] and
soybean [Glycine max]) that are known to produce most food allergies in
young children. While these food allergen mixes containing allergosorbents
are useful in screening children for confirmation of sensitization to foods,
they are not generally useful in inhibition assays for assessing allergenic
potency of food extracts and thus they are not discussed further in this
chapter.
A different allergosorbent preparation approach has been used in microtiter
plate-based non-isotopic assays. While the inhibition format of ELISAs is
discussed elsewhere in this volume, it is important to note here that adsorption
of proteins onto a plastic microtiter plate surface has severe limitations for
complex protein mixtures such as those present in food extracts. Adsorption
is a random process in which proteins of select isoelectric points and molecular
weights adhere to the plastic surface by weak ionic and hydrophobic
interactions. This adsorption process in allergosorbent preparation functions
best for recombinant or native allergens that have been purified from other
‘irrelevant’ proteineous material in the extract. Unfortunately, it has not been
possible to ensure the binding of all the relevant allergens from crude allergen
extracts, especially those prepared from foods. Therefore, it is recommended
that a covalent coupling method be employed if mixtures of proteins from
foods are being used to prepare the allergosorbent.
88 Detecting allergens in food
4.4 Applications
A number of studies provide unique insights into the practical use of the
RAST inhibition assay in the assessment of foods for the presence and
relative quantity of allergens. As an illustration, the RAST inhibition assay
has been used to investigate the allergenicity of commercially-available peanut-
containing foods and to identify peanut contamination of non-peanut derived
foods.19,21,22 In one such protocol, 100 gm of each test product is defatted
92 Detecting allergens in food
once with 250 mL of acetone and then five times with 250 ml of ethyl ether.
The residual, defatted product is then dried and mixed for 20 hours at 25 °C
with 300 mL of 0.1 M ammonium bicarbonate adjusted to pH 8.0. The
extract is clarified by centrifugation of the supernatant for 30 minutes at
24 000 × g at 2 °C and sterile filtration. A total protein is performed using the
Folin phenol reagent described by Lowry et al.23 Only the peanut oil was
tested directly without defatting or extraction.
In these studies, the competitive RAST inhibition analysis was performed
using a human IgE antibody pool comprising sera from multiple peanut
allergic individuals with highly positive peanut specific IgE antibody levels.
The peanut allergosorbent was prepared by coupling crude raw peanut extract
to cyanogen bromide-activated microcrystalline cellulose particles. RAST
inhibitory activity of the test extracts was compared to the competitive inhibition
reference curve produced by a reference peanut extract.
Qualitatively, 17 of the 20 peanut products tested were shown to contain
peanut allergen by the competitive RAST inhibition assay. Two were raw
peanuts and the others were processed foods containing peanut that had
involved shelling, blanching, dry roasting, oil roasting, toasting, grinding,
defatting, extracting and/or combining them with other ingredients. Three
figures have been reproduced from the Nordlee et al. paper19 to illustrate
several points. Figure 4.2 displays similar RAST inhibition curves produced
J I L
STD
K
70 K
J LK
60 J LK
I
J LI L
% Inhibition
50 J L K
I I
J I L
40
J L
30 I K
20 I
J
10
0
0.1 0.6 1.1 1.6 2.1 2.6 3.1 3.6
Log μg
Fig. 4.2 RAST inhibition curves produced by extracts of four peanut butter products.
The percent inhibition of the binding of peanut-specific IgE-containing serum to raw
peanut extract allergosorbent that is induced by the various extracts is plotted as a
function of the log of the protein concentration in the extract in micrograms. Extract I
= peanut butter powder; extract J = peanut butter; extract K = peanut butter syrup;
extract L = peanut butter flavoured chips. The inhibition curves are essentially
parallel, with slopes that are analogous to the RAST inhibition curve slope produced
by the standard raw peanut extract. Only the slope of the peanut butter extract (J)
differs significantly from the standard curve (p < 0.025) and extract J was more potent
than extracts from the other peanut butter products that contained a mixture of peanut
and whey proteins. Reproduced with permission from Nordlee et al.19
Allergen-specific human IgE antibody-based analysis of food 93
O Q M STD
P
M PP N
70 P P
Q P P
60 N
M P N
O QQQ
% Inhibition
M M N N
50 Q
M P
O N
40 Q
Q N
N M
30 O
Q
MO
20
O
10 O
0
0.1 0.6 1.1 1.6 2.1 2.6 3.1 3.6
Log μg
Fig. 4.3 RAST inhibition curves produced with raw and roasted peanut extracts.
Extract M = deflavored peanuts; extract N = raw Virginia peanuts; extract O = oil
roasted Virginia peanuts; extract P = raw Florunner peanuts; extract Q = dry roasted
Florunner peanuts. These curves suggest differences in the allergenic potency among
the different varieties of peanuts. Moreover, other factors are most likely involved
because the inhibition curves produced by extracts of the deflavored peanuts (M) and
dry roasted Florunner peanuts (Q) had approximately the same degree of allergenicity
in comparison to the standard peanut extract. Reproduced with permission from
Nordlee et al.19
with the peanut butter product extracts that appear to have a similar quantity
and specificity distribution of peanut allergens. In contrast, Fig. 4.3 displays
inhibition curves with significantly different slopes suggesting that the extracts
from Florunner peanuts are less allergenic than the deflavored and dry roasted
peanuts. This also may suggest varietal differences in the nature of the allergenic
determinants among the different sources of peanuts. Figure 4.4 illustrates
RAST inhibition curves for peanut oil and the acid hydrolyzed peanuts that
contained no detectable peanut allergen. The negative slope of the inhibition
curve with the peanut oil suggests non-specific interference in the RAST
inhibition assay. In Fig. 4.4, the peanut hull flour contained low levels of
peanut allergen in comparison to the reference peanut extract as evidenced
by the large shift of the inhibition curve (albeit parallel) from the reference
curve. The authors conclude that peanut hypersensitive individuals should
avoid all peanut-containing products with the exception of peanut oil and
possibly acid hydrolyzed peanut where the allergens appear to have been
destroyed. The authors also caution that some non-peanut foodstuffs can
become cross-contaminated with peanut allergens as detectable by competitive
RAST inhibition due to inadequate cleaning of common processing
equipment.21
94 Detecting allergens in food
STD
70
T
60 T
T
% Inhibition
50
T
T
40
T
30
T
R R
20
T S S S
R S R S S
10 R R S
S
R
0 R R
0.1 0.6 1.1 1.6 2.1 2.6 3.1 3.6
Log μg (R is Log μl)
Fig. 4.4 RAST inhibition curves produced by a variety of peanut containing foods
and peanut oil. Extract R = peanut oil (analyzed per microliter volume of oil); S =
hydrolysed peanut protein; extract T = peanut hull flour. The flat or inverse slope of
extracts S and R, respectively indicate that these materials do not contain any
detectable peanut allergen. In contrast, the peanut hull flour extract contains only
small amounts of peanut allergen as evidenced by the parallel slope that is displaced
extensively to the right. Reproduced with permission from Nordlee et al.19.
4.5.1 Strengths
The competitive RAST inhibition’s strength is its ability to detect and quantify
cross-reactivity among allergenic proteins from structurally similar allergens
in different foods and between pollens and food.24 Because allergen-specific
human IgE antibody is used, clinically relevant allergens are detected that
may induce human IgE antibody responses in some clinically sensitive
individuals. The slope and displacement of the test extract inhibition curve
from the reference curve provides useful information about the relative
allergenic potency and specificity distribution of complex protein allergen
mixtures, especially when specific allergens have not yet been identified.
Allergen-specific human IgE antibody-based analysis of food 95
4.5.2 Weaknesses
All human IgE-based methods suffer from a dependence on the unique
allergenic specificity of the IgE antibody that is naturally present in individual
sera or serum pools. Second, inhibition assays that use IgE antibody containing
human sera consume large amounts of serum because the IgE antibody is
normally present in small (ng/mL) quantities. For this reason, IgE antibody-
based inhibition assays are rarely used in food manufacturing facilities where
IgE containing sera are not readily available. Third, the heterogeneity of the
IgE antibody specificity among different sensitized individuals can be a
problem for reproducibility of the RAST inhibition assay. Cross-validation
of serum pools is important to verify consistency of the IgE antibody specificity
and quantity in new serum pools which are being prepared for use. Fourth,
radioisotopes such as I125 in the RAST assay need special care and skill and
specialized laboratory licences ensuring appropriate surveying programs to
permit their handling. Some laboratories prefer to use non-isotopic methods
such as enzymes or fluorophors that are safer and easier to handle and
dispose of after the assay is completed. This has been largely addressed by
the development of commercial RAST-like assays that employ an enzyme
label and colorimetric or fluorescent substrates to detect IgE bound to the
allergosorbent following the serum incubation step.
4.7 References
1. Hamilton, R G and Adkinson, N F Jr (2004) ‘In vitro assays for the diagnosis of IgE
mediated disorders’ J Allergy Clin Immunol, 114, 213–225.
2. Schellenberg, R R and Adkinson, N F Jr (1975) ‘Measurement of absolute amounts
of antigen-specific human IgE by a radioallergosorbent test (RAST) elution technique’,
J Immunol, 115, 1577–1583.
3. Ewan, P W, and Coote, D (1990) ‘Evaluation of a capsulated hydrophilic carrier
polymer (the ImmunoCAP) for measurement of specific IgE antibodies’, Allergy,
45, 22–29.
4. Paganelli, R, Ansotegui, I J, Sastre, J, Lange, C E, Roovers, M H W M, de Groot, H,
Lindholm, N B and Ewan, P W (1998) ‘Specific IgE antibodies in the diagnosis of
atopic disease. Clinical evaluation of a new in vitro test system, UniCAP, in six
European allergy clinics’, Allergy, 53, 763–768.
5. Li, T M, Chuang, T, Tse, S, Hovanec–Burns, D, Said El and Shami, A (2004)
‘Development and validation of a third generation allergen-specific IgE assay on the
continuous random access IMMULITE® 2000 analyzer’, Ann Clin Lab Sci, 34, 67–
74.
6. Food and Agriculture Organization (1995) ‘Report of the FAO technical consultation
on food allergies’, Rome, FAO.
7. Matsson, P, Hamilton, R G, Adkinson, N F, Jr, Esch, R, Homburger, H, Maxim, P
and Williams, P (1998) Evaluation Methods and Analytical Performance Characteristics
of Immunological Assays for Human Immunoglobulin E (Ige) Antibodies of Defined
Allergen Specificities, Vol 16-LA20-A, 19, Wayne, PA, National Committee on Clinical
Laboratory Standards.
8. Turkeltaub, P C, Rostogi, S C, Baer, H, Anderson, M C and Norman, P S (1982) ‘A
standardized quantitative skin-test assay of allergen potency and stability: studies on
the allergen dose-response curve and effect of wheal, erythema, and patient selection
on assay results’, J Allergy Clin Immunol, 70, 343–352.
9. Taylor, S L, Hefle, S L, Bindslev-Jensen, C, Atkins, F M, Andre, C, Bruijnzeel-
Koomen, C, Burks, A W, Bush, R K, Ebisawa, M, Eigenmann, P A, Host, A, Hourihane,
J O, Isolauri, E, Hill, D J, Knulst, A, Lack, G, Sampson, H A, Moneret-Vautrin, D
A, Rance, F, Vadas, P A, Yunginger, J W, Zeiger, R S, Salminen, J W, Madsen, C and
Abbott, P (2004) ‘A consensus protocol for the determination of the threshold dose
for allergenic foods: how much is too much?’ Clin Exp Allergy, 34, 689–695.
10. Sampson, H A and Scanlon, S M (1989) ‘Natural history of food hypersensitivity in
children with atopic dermatitis’, J Pediatr, 115, 23–27.
11. Jansen, J N, Kardinaal, A F M, Huijbers, G, Vlieg-Boerstra, B J, Martens, B P M and
Ockhuizen, T (1994) ‘Prevalence of food allergy and intolerance in the adult Dutch
population’, J Allergy Clin Immunol, 93, 446–456.
12. Richman, P G and Cissel, D S (1988) ‘A procedure for total protein determination
with special application to allergen extract standardization’, J Biol Stand, 16, 225–
238.
13. Breiteneder, H. and Radauer, C (2004) ‘A classification of plant food allergens’, J
Allergy Clin Immunol, 113, 821–830.
14. Blanco, C, Carrillo, T, Castillo, R, Quiralte, J and Cuevas, M (1994) ‘Latex allergy:
clinical features and cross-reactivity with fruits’, Ann Allergy, 73, 309–314.
15. Diaz-Perales, A, Collada, C, Blanco, C, Sanchez-Monge, R, Carrillo, T, Aragoncillo,
C and Salcedo, G (1998) ‘Class I chitinases with hevein-like domains, but not class
II enzymes are relevant chestnut and avocado allergens’, J Allergy Clin Immunol,
102, 127–33.
16. Wide, L, Bennich, H and Johansson, S G O (1967) ‘Diagnosis of allergy by an in
vitro test for allergen-antibodies’, Lancet, 2, 1105–1108.
17. Langeland, T A (1982) ‘A clinical and immunological study of allergy to hen’s egg
Allergen-specific human IgE antibody-based analysis of food 97
5.1 Introduction
Since its introduction in health sciences (Burnette, 1981; Glass, Briggs and
Hnilica, 1981; Karcher et al., 1981; McMichael, Greisiger and Millman,
1981; Reiser and Wardale, 1981; Symington, Green and Brackmann, 1981),
immunoblotting or Western blotting has proven to be a very powerful research
tool and one that has spread rapidly. A search in PubMed using the keywords
‘immunoblotting’, ‘food’ and ‘allergens’ resulted in almost 500 hits, of which
the first go back to the second half of the 1980s (Theobald et al., 1986;
Enberg et al., 1987; Burks, Jr, Brooks and Sampson, 1988; Bush et al., 1988;
Enberg, McCullough and Ownby, 1988; Vallier et al., 1988 Naqpal et al.,
1989;). In most cases, immunoblotting (Kurien and Scofield, 2003) is used
in conjunction with sodium dodecyl sulphate-polyacrylamide gel
electrophoresis (SDS-PAGE) under reducing conditions (Fig. 5.1).
Separation of proteins prior to electrophoretic transfer to nitrocellulose or
polyvinylidene difluoride (PVDF) membranes can also be two-dimensional,
in which case SDS-PAGE is preceded by isoelectric focusing (Celis and
Gromov, 2000; Lilley, Razzaq and Dupree, 2002). Two-dimensional
electrophoresis allows separation of proteins with similar molecular mass
but different pI. In food allergen research, immunoblotting has been used for
many different purposes. Most frequently, the technique has been applied for
qualitative purposes, i.e. the identification of allergenic (IgE-binding) molecules
and the establishment of IgE cross-reactivity between foods or between
foods and inhalant allergens (Bauer et al., 1996; Teuber and Peterson, 1999;
Kazemi-Shirazi et al., 2000; Asturias et al., 2002; Lüttkopf et al., 2002;
Reindl et al., 2002; Miguel-Moncin et al., 2003; Wensing et al., 2003; Bolhaar
Immunoblotting in allergen detection 99
W
M
h
ig
H
W
M
L ow
MW
markers
(a)
Nitrocellulose
(b)
Secondary labeled or
Allergen-specific conjugated antibody
antibody
Contaminating
allergen
(c)
et al., 2004; Comstock et al., 2004). In the former case, serum samples of
food-allergic patients are used to identify proteins that are recognized by IgE
antibodies. A critical issue in such analyses is of course patient selection. In
the latter case, IgE binding to immunoblots is inhibited by allergen extracts
or purified natural or recombinant allergens with suspected cross-reactivity
to the allergens on immunoblot. Immunoblot inhibition with purified allergens
as an inhibitor is a powerful tool to identify or confirm the nature of an
allergen recognized on immunoblot.
Detection of allergenic molecules in food products is another area where
immunoblotting can be applied. Specific monoclonal antibodies (mAb) or
monospecific polyclonal antibodies against the major peanut allergen Ara h
1 could, for example, be used to trace contamination of chocolate products
with peanut. Also human sera from allergic patients have been used for the
detection in chocolate of trace amounts of allergens from, for example,
hazelnut and almond (Scheibe et al., 2001). Prerequisite for such an application
is selective recognition of the food contamination under investigation. Although
antibodies do not necessarily have to be directed to allergens (any protein
will do), we will limit ourselves to allergen-reactive antibody reagents.
1 2 3 4 5 6 7
1 2
38
28
17
14
Fig. 5.3 Detection of apple LTP (Mal d 3): monoclonals versus polyclonals. Mouse
monoclonal antibodies (lane 1) and rabbit polyclonal antibodies (lane 2) raised against
purified Mal d 3 both exclusively detect the 9 kDa LTP band, demonstrating their
mono-specificity.
1 2 3 4
1 2 3 4 5 6 7 1 2 3 4 5 6 7
(a) (b)
Fig. 5.5 Lack of specificity of polyclonal rabbit antisera. Rabbit antisera against two
grass pollen allergens, Lol p 1 (a) and Lol p 12 or profilin (b) detect multiple bands in
plant food extracts. Although anti-Lol p 12 indeed detects profilin (indicated by an
arrow) this antiserum is far from monospecific. The fact that both antisera recognize
very similar (glyco)proteins suggests that these antibodies were already present prior
to immunisation and are directed to highly cross-reactive (carbohydrate?) structures.
104 Detecting allergens in food
5.6 References
Akkerdaas, J H, van Ree, R, Aalbers, M, Stapel, S O and Aalberse, R C (1995) ‘Multiplicity
of cross-reactive epitopes on Bet v I as detected with monoclonal antibodies and
human IgE’, Allergy, 50(3), 215–220.
Asero, R, Mistrello, G, Roncarolo, D, de Vries, S C, Gautier, M F, Ciurana, C L, Verbeek,
E, Mohammadi, T, Knul-Brettlova, V, Akkerdaas, J H, Bulder, I, Aalberse, R C and
van Ree, R (2000) ‘Lipid transfer protein: a pan-allergen in plant-derived foods that
is highly resistant to pepsin digestion’, Int Arch Allergy Immunol, 122(1), 20–32.
Asero, R, Mistrello, G, Roncarolo, D, Amato, S, Caldironi, G, Barocci, F and van Ree, R
(2002) ‘Immunological cross-reactivity between lipid transfer proteins from botanically
unrelated plant-derived foods: a clinical study’, Allergy, 57(10), 900–906.
Asturias, J A, Eraso, E, Arilla, M C, Gomez-Bayon, N, Inacio, F and Martinez, A (2002)
‘Cloning, isolation, and IgE-binding properties of Helix aspersa (brown garden snail)
tropomyosin’, Int Arch Allergy Immunol, 128(2), 90–96.
Bando, N, Tsuji, H, Hiemori, M, Yoshizumi, K, Yamanishi, R, Kimoto, M and Ogawa, T
(1998) ‘Quantitative analysis of Gly m Bd 28K in soybean products by a sandwich
enzyme-linked immunosorbent assay’, J Nutr Sci Vitaminol. (Tokyo), 44(5) 655–664.
Bardor, M, Faveeuw, C, Fitchette, A C, Gilbert, D, Galas, L, Trottein, F, Faye, L and
Lerouge, P (2003) ‘Immunoreactivity in mammals of two typical plant glyco-epitopes,
core alpha(1,3)-fucose and core xylose’, Glycobiology, 13(6), 427–434.
Bauer, L, Ebner, C, Hirschwehr, R, Wüthrich, B, Pichler, C, Fritsch, R, Scheiner, O, and
Kraft, D (1996) ‘IgE cross-reactivity between birch pollen, mugwort pollen and celery
is due to at least three distinct cross-reacting allergens: immunoblot investigation of
the birch-mugwort-celery syndrome’, Clin Exp Allergy, 26(10), 1161–1170.
Becker, W M (1997) ‘Characterization of Ara h 1 by two-dimensional electrophoresis
immunoblot and recombinant techniques: new digestion experiments with peanuts
imitating the gastrointestinal tract’, Int Arch Allergy Immunol, 113(1–3), 118–121.
Björksten, F, Halmepuro, L, Hannuksela, M and Lahti, A (1980) ‘Extraction and properties
of apple allergens’, Allergy, 35(8), 671–677.
Bolhaar, S T, Ree, R, Bruijnzeel-Koomen, C A, Knulst, A C and Zuidmeer, L (2004)
‘Allergy to jackfruit: a novel example of Bet v 1-related food allergy’, Allergy, 59(11),
1187–1192.
Bouwstra, J B, Spoelstra, E C, De Waard, P, Leeflang, B R, Kamerling, J P and Vliegenthart,
J F (1990) ‘Conformational studies on the N-linked carbohydrate chain of bromelain’,
Eur J Biochem, 190(1), 113–122.
Breiteneder, H and Ebner, C (2000) ‘Molecular and biochemical classification of plant-
derived food allergens’, J Allergy Clin Immunol, 106(1 Pt 1), 27–36.
Burks, A W, Jr, Brooks, J R and Sampson, H A (1988) ‘Allergenicity of major component
proteins of soybean determined by enzyme-linked immunosorbent assay (ELISA) and
immunoblotting in children with atopic dermatitis and positive soy challenges’, J
Allergy Clin Immunol, 81(6), 1135–1142.
106 Detecting allergens in food
6.1 Introduction
Immunoassays such as Enzyme-Linked Immunosorbent Assays (ELISAs)
have been found to have considerable application in clinical diagnostics.
They are in fact the method of choice, but have had thus far little impact on
food analysis. However, since 1990 there have been an increasing number of
reports on the use of ELISA techniques for environmental contaminants in
foods. Unfortunately, only a few have been reported for food allergens.
Until 1988, when Yunginger et al.1 reported a series of eight accidental
deaths due to food-induced anaphylaxis, allergic reactions to food were not
considered a significant public health risk. Subsequent reports of fatalities and
severe allergic reactions2–4 due to food allergies heightened the awareness of
both regulators and the food industry that a small segment of the population
could experience serious reactions to food allergens. With the increased attention
to allergic reactions to food and, especially, allergen-related recalls, there was
a need for quick and accurate methods to detect allergenic residues in food.
Prior to the current emphasis on immunochemical methods for the detection
of allergens, most of the detection used organoleptic techniques, such as
subjective sensory (smell and taste) tests, or chromatographic fatty acid
profile analyses. Unfortunately, these sensory and chromatographic methods
were neither specific to the allergenic proteins, nor sensitive enough to test
for allergen residues. Methods currently used to test for allergens, including
commercially available test kits, are primarily based upon immunochemical
procedures, although some polymerase chain reaction (PCR) kits have also
been developed. These allergen detection methods have recently been reviewed
by Besler5 and Poms et al6; an update is provided in the second part of this
book.
110 Detecting allergens in food
6.2.1 Antibodies
Immunoglobulins (as discussed in Chapter 3) are a group of closely related
glycoproteins composed of 82–96% protein and 4–18% carbohydrate. An
antibody is an immunoglobulin, a protein produced by the immune system in
response to the presence of an antigen. Antibodies exist as one or more
copies of a Y-shaped unit, composed of four polypeptide chains. Each Y
contains two identical copies of a ‘heavy chain’ (H), and two identical copies
of a ‘light chain’ (L), named as such by their relative molecular weights,
linked together by inter-chain disulfide bonds (Fig. 6.1a). Intra-chain disulfide
en te bi An
nd tig
ig si
nt ng Variable
in en
g
A di si
n constant te
bi
Light chain
Heavy chain
(a)
Fig. 6.1 (a) Antibody structures. (b) VH and VL regions on an antibody. (c) Pepsin
and papain digestion of an IgG antibody.
Enzyme-linked immunosorbent assays (ELISAs) 111
+H 3N
+H 3N
+H 3N
+H 3N
VH
S
S
S
S
S
S
Lc
ch in
VL
S
H
L
ain
ch ain
ha
C H1
ch
ai
S
n
S
S
Fab
Fa
S
S
b
C
S
L
S
SS
S
S
S
S
Hin
C H2
ge
S
S
reg
H chain
H chain
ion
Fc
S
S
C H3
S
S
COO–
COO–
(b)
Fab
F(ab′)2
Pa
in pa
ps dig in
Pe tio
n es
es tio Fab
d i g IgG n
Peptides
Fc
pFc′
(c)
bonds are responsible for the formation of loops, leading to the compact,
domain-like structure of the molecule. The amino terminal portions of the
heavy and light chains, characterized by a highly variable amino-acid
composition among different antibodies, are referred to as VH and VL,
respectively. The constant parts of the light chain are designated as CL, while
the constant parts of the heavy chains are further divided into three distinct
subunits: CH1, CH2 and CH3 (Fig. 6.1b). Functionally, the V regions are
involved in antigen binding. The C regions interact to hold the molecule
together and are involved in several biological activities, such as complement
binding and binding to cell membranes.
Antibodies are divided into five major classes: IgM, IgG, IgA, IgD and
IgE, based on their constant region structure and immune function. The most
commonly used antibody is IgG, which can be cleaved into three parts, two
F(ab) regions and one Fc, by the proteolytic enzyme papain, or into two
parts, one F(ab′)2 and one Fc by the proteolytic enzyme pepsin. While both
F(ab) and F(ab′)2 fragments can be used in immunoassays to enhance sensitivity,
F(ab′)2 has higher avidity (Fig. 6.1c). When designing procedures, it is important
to differentiate between monoclonal and polyclonal antibodies, as these
differences are the foundation of both advantages and limitations for their
use.
Legend
A Capture antibody
A Antigen or allergen
E S Substrate
S
E
Competitive ELISA
The competitive inhibition ELISA is a technique that uses a one-epitope
approach for the Ab to recognize allergenic residue in a sample. In a competitive
assay, the Ag is coated on the wells, and a solution containing a limited
amount of first Ab along with the Ag or analyte is added. The assay is based
on the principle that an Ag in the sample will bind to an Ab and then compete
for the binding of the Ag coating on the wells. After the unbound Ab is
washed off, a second Ab-enzyme conjugate is used to detect the bound
Ag:Ab complex in the wells. Then a substrate of the enzyme is added (Fig.
6.3). In this format, the color produced is inversely proportional to the
concentration of the analyte, i.e. the higher the color the lower the concentration
of the protein.
114 Detecting allergens in food
A A
Legend
First antibody
A
A
A Antigen or allergen
A A
S Substrate
A A
S
E
A A
Fig. 6.3 A typical competitive inhibition ELISA. First antibody reacts with bound
antigen, and a labelled second antibody reacts with the primary antibody.
6.3 Applications
6.3.1 Methodologies
Numerous immunochemical methods for the detection of allergens have
been described in the literature9–46 and are discussed in detail elsewhere in
the book. These include ELISAs, dipsticks (see Chapter 10), biosensors (see
Chapter 9) and immunoblot (see Chapter 5) and immunoaffinity columns.
116 Detecting allergens in food
Availability
In the mid-1990s, commercial kits came to the market offering laboratories
the opportunity to analyze peanut proteins in processed food products or in
samples taken from various places in a processing facility, and others followed.
Currently, most of the available allergen test kits are ELISA-based methods,
but PCR-type kits are emerging. At the present time, there are five allergen
kit manufacturers (Table 6.1); it is expected that in the near future more
manufacturers will be producing a wider range of allergenic residue kits for
use by the food industry. Since detection limits declared by manufacturers
change in time, especially with the change of regulatory climate, readers are
referred to their respective kit manufacturers for details.
Other than the newly introduced PCR kits, all five commercial kit
manufacturers use a similar ELISA technology for the detection of allergenic
residues; hence, they all share the same pitfalls. Using peanut as an example,
one manufacturer claims a detection limit of 0.5 ppm peanut proteins while
the other four claim detection limits ranging from 1–5 ppm peanuts. Please
note, the reporting units are not equivalent. Since the LOD can vary greatly
depending on food matrix interferences, extraction efficiency, specificity of
antibodies and variation of peanut protein standards, should be using proper
validation suitable for its intended use is essential. However, none of the
commercial methods have been published in a peer-reviewed journal and
only test kits for peanuts are validated by inter-laboratory collaborative studies
to date, although work on this is ongoing at the time of writing.
In the real world, analytical sensitivity has limited practical value if
uncertainty is not defined, since precision decreases very rapidly as
concentration decreases. A question that remains to be answered is how
sensitive the detection methods should be. It is commonly recognized that
the required analytical sensitivity will vary, depending on the purpose of use
of the assay result. Allergen test results are ultimately used to protect allergic
consumers. In the absence of de minimis threshold levels for food allergens,
sensitivity of allergen methods should aim at detecting any amount of allergenic
food sufficient to elicit objective allergic reactions in allergic individuals
established by double-blind placebo-control food challenge (DBPCFC) studies.
Although allergen threshold levels are finite, and not zero,47 unfortunately,
regulatory guidance on action levels on undeclared allergens is still lacking.
Any positive ELISA result can only be treated as a presumptive positive,
and confirmation or further testing has to be performed. There is an urgent
need for confirmatory tests for the presence of allergenic residues in food.
The only unambiguous confirmatory test for peanut ELISA has demonstrated
the presence of a major peanut allergen, Ara h 1, in ice cream using liquid
chromatography/tandem mass spectrometry (LC/MS/MS). To identify potential
Ara h 1 biomarkers, Shefcheck and Musser48 digested the peanut proteins
with trypsin into their component peptides and then identified the four abundant
peptides that are unique to Ara h 1 as specific peptide biomarkers.
Finally, there is no consensus agreement on acceptable reference materials
by the analytical community, even though some standard reference materials
are commercially available, e.g. National Institute of Standards and Technology
(NIST), https://srmors.nist.gov/pricerpt.cfm; or The Institute for Reference
Materials and Measurements (IRMM), http://www.irmm.jrc.be/. Because
there is no uniform peanut protein standard known, kit manufacturers use
their own peanut protein extracts as standards and as immunogens. As a
result, quantitative results of identical samples will differ when they are
analyzed by different manufacturers’ kits. For example, our data showed that
when 10 ppm of commercial medium roasted peanut flour was added to
melted milk chocolate and mixed to homogeneity, results obtained from four
different commercial kits ranged from 2 to 40 ppm of peanuts or peanut
Enzyme-linked immunosorbent assays (ELISAs) 119
Table 6.2 Quality control samples analyzed by different commercial peanut kits
Chocolate 0 nd nd nd nd
10 positive1 26 2.3 9.2
50 positive 207 18.8 27.8
Cookies 0 nd nd nd nd
10 positive 10.9 0.9 4.8
50 positive 160 7.0 13.1
Cereal 0 nd nd nd nd
10 positive 43.5 2.3 6.7
50 positive 171 24.9 44.8
Ice cream 0 nd nd nd nd
10 38.5 40.2 2.0 8.7
50 154 222 7.5 56.0
Detection 1.0 2.5 0.5 2.5
limit (ppm)
proteins according to the reporting unit of the test kit used. Similar variations
were also observed in different matrices in our quality control samples that
were prepared similarly (Table 6.2). In order to resolve questions regarding
commercial test kit methods, reference standards must be available globally
from a recognized body such as the NIST or IRMM, reporting units need to
be harmonized and collaborative validation studies should be done. Without
reference standards, other issues such as false positives, false negatives,
sensitivity, matrix interference and recovery cannot be adequately assessed.
Sampling
Even when the best analytical methodology and quality assurance schemes
are in place, large errors can be introduced into allergenic residue analysis by
inadequate sampling. This is due, in part, to the inadvertent presence of
allergens being unevenly distributed in solid samples, such as finished food
products. Therefore, obtaining a representative sample is a way of minimizing
false results and increases the chances of accurate determination of allergens
in a batch or lot.
Different organizations/analysts develop different sampling protocols to
ensure that ‘representative’ samples are taken. Some ensure that ‘false negative’
results are minimized; others are more effective at reducing ‘false positive’
results. The ideal sampling plan minimizes the risks associated with both
these errors to protect consumer safety and reduce unnecessary product waste.
Nevertheless, it is important to have effective sampling plans to satisfy both
government and industry acceptable limits.
120 Detecting allergens in food
Dipstick technology
A promising approach in rapid food allergen detection is the use of a dipstick
format (discussed at length in Chapter 10). Such tests are inexpensive, rapid
and can be done anywhere. They can be used on-site rather than in the
laboratory, hence permitting early detection of potential problems, such as
the ineffective cleaning of equipment. Antibody-based dipsticks (lateral flow
devices) for peanut and gluten are now commercially available. Similar
developments for other allergens are expected to emerge shortly.
Biosensors
Biosensors offer the possibility of in-line detection of allergens. These devices
provide real-time data and require only minimal technical training to operate.
Biosensors are based on the coupling of two components: a sensor chip
containing a bioactive receptor such as an Ab that captures the analyte of
interest, and a transducer that converts the biochemical recognition step into
a quantifiable optical signal.49 For example, surface plasmon resonance (SPR)
biosensors use a direct sensing technique that can detect refractive index
changes that occur in the vicinity of a thin metal film surface where Ab
complexes with Ag. The change in refractive index is proportional to the
concentration of the analyte under investigation. For this type of assay, the
whole process takes less than five minutes. The SPR-based biosensor has
been successfully used for the determination of proteins, mycotoxins, drug
residues, pesticides and peanut proteins.50,51 It should be possible to use SPR
technology for the detection of all food allergens since they are all proteins.
Enzyme-linked immunosorbent assays (ELISAs) 121
6.5 Conclusions
Immunoassays, such as ELISA, have clearly had a major impact on food
allergen detection, but their potential in other areas of allergen research is
only now just beginning to be realized. ELISA-based methods can help
manufacturers to validate their allergen prevention programs, including
sanitation Standard Operation Procedures (sSOP), to prevent cross contact in
food processing operations and help government agencies to support regulatory
actions.
While allergenic residue testing techniques are continuing to improve
with respect to accuracy, reliability and speed, it should be stressed that their
performance is strongly affected by sampling strategies. It is important to
harmonize internationally accepted sampling protocols, validated
methodologies and approved reference standards in this global economic
climate. Validation and harmonization of quantitative ELISA is needed to
address regulatory compliance for food processing and food services industries
to ensure the effectiveness of their food safety programs.
There is an urgent need for the development of ELISA-based rapid methods,
such as dipsticks or biosensors and multi-residue procedures, which may
provide attractive tools for field monitoring of the integrity of good
manufacturing practices (GMP), whereby non-specialized personnel can
employ them in a cost-effective manner. Such tests can be used on-site rather
than in the laboratory, hence permitting early detection of potential problems,
such as the ineffective cleaning of line equipment before shifting to another
product. This early availability of critical data translates into early management
decisions to avoid the inadvertent introduction of allergens into the production
lines due to cross-contact.
Clearly, ELISA is a very powerful tool in food allergen detection. In fact,
it is the method of choice. The field of ELISA technology shows dynamic
growth driven by growing interest in allergenic residue analyses, and increasing
legislative and regulatory attention to the health risk of food allergens. As a
technical platform, current ELISA technology offers a significant improvement
in versatility and performance advantages over other detection technologies.
6.6 Acknowledgements
The author wishes to thank Regina McDonald, Dr Cecilia Fernandez and Dr
Mara Nogueira for providing some of the peanut kit evaluation data.
6.7 References
1. Yunginger, J W, Sweeney, K G, Sturner, W Q, Giannandrea, L A, Teigland, J D,
Bray, M, Benson, P A, York, J A, Biedzycki, L, Squillance, D L and Helm, R M
(1988) ‘Fatal food-induced anaphylaxis’, JAMA, 260, 1450–1452.
122 Detecting allergens in food
7.1 Introduction
For verification of allergen labeling of foods and in order to identify hidden
allergens in processed food it is important to provide analytical methods that
are able to detect very low amounts of allergenic residue in processed foods.
In general, these methods are based on the detection of species-specific
proteins by enzyme-linked immunoassay (ELISA).1–8 In addition, the detection
of species-specific DNA molecules by the polymerase chain reaction
(PCR) for the identification of allergenic foods at trace levels has been
demonstrated.9–18
PCR technology has already been established as the DNA-based method
for the identification of genetically modified organisms, pathogens and food-
related plant and animal species. Furthermore, PCR has been widely used for
many approaches in the field of molecular biology and routinely applied in
clinical chemistry. Since the mid 1990s this methodology has attracted
increasing attention for the detection of allergen traces in food products; this
is reflected by an increasing number of published and commercially available
PCR assays (summarized by Poms et al.19). This chapter will review published
data on the applicability of PCR as a possible alternative to, and as a tool for
the verification of, ELISA methods and discuss the potential and restrictions
of PCR used in the detection of allergenic foods.
Fig. 7.1 DNA extraction and purification based on DNA-binding silica membranes:
characteristic steps in DNA-isolation and necessary reagents and consumables.
95°C 45–65°C
Double-stranded
DNA Single-stranded
DNA
= Primer
2n
= Taq-polymerase = dNTPs
Fig. 7.2 Principle of the PCR. After heat denaturation of the genomic DNA (a),
sequence-specific primers bind to the single-stranded DNA during the annealing phase
(b). Subsequently, the Taq DNA polymerase extends the primers by filling up the
missing strand with desoxynucleotidetriphosphates (c). The exponential amplification
of the PCR products is displayed in (d).
128 Detecting allergens in food
Biotinylated
PCR amplicon
Single-stranded
amplicon bound
at the solid
Streptavidin phase
Sequence-specific Enzyme-
labeled probe conjugated
antibody
Enzyme
substrate
Fig. 7.3 Principle of the PCR-ELISA. Biotinylated PCR amplicons are bound to streptavidin-coated wells in a microtiterplate (a). After
denaturation of the double-stranded amplicons (b) a sequence-specific labelled probe can hybridize (c). The detection of the probe is done by an
enzyme-conjugated antibody specific for the probe label (d). The color development is measured as absorbance in a microplate reader.
Polymerase chain reaction (PCR) methods 131
3.0
2.5
Fluorescence
2.0
1.5
1.0
0.5
Threshold
0.0
–0.5
10 20 30 40 50 60
Cycle number
(a)
45
40 y = –3.9647x + 42.13
R2 = 0.985
35
Ct-value
30
25
20
100 101 102 103 104 105
Copy number
(b)
Fig. 7.4 Real-time PCR. (a) Amplification plots of serially diluted DNA. (b) Cycle
threshold (Ct) values of the amplification plots from (a) plotted against the logarithm
of the copy numbers of amplifiable DNA prior to PCR analysis. These result in linear
correlation.
132 Detecting allergens in food
14
Total
12
ELISA
10 PCR-ELISA
Sample number
0
00 10
0 10 >
1
<
1
tiv
e
10 > > a
>
N eg
mg/kg hazelnut protein
Fig. 7.5 Investigation of commercial food samples for hazelnut residue with hazelnut
specific protein sandwich-ELISA and DNA PCR-ELISA.13 Of the 41 investigated
chocolate, creme desert, breakfast flakes, cereal bars, and cookie samples, only three
discrepancies were observed for samples having less than 1 mg/kg hazelnut protein
(two samples were negative in ELISA but positive in PCR; one sample was positive in
ELISA but negative in PCR).
Polymerase chain reaction (PCR) methods 135
were not compared with those of a validated immunological assay, nor was
an unspiked biscuit analyzed as negative control. As reported by the authors,
the peanut PCR may be used for analysis if reference standards for peanut
become available.
The performance of a new peanut-specific real-time PCR was compared
with an in-house validated peanut ELISA.18 Thirty-three different food samples
were analyzed for the presence of peanut traces with both methods allowing
a detection of peanut at a level of ≤ 10 mg/kg. The results of ELISA and PCR
were in good correlation. There was only one discrepancy found in a chocolate
that was negative in ELISA (< 0.5 mg/kg of peanut protein), but positive in
PCR. The major characteristics of the published PCR methods are summarized
in Table 7.1.
soy and wheat were not optimized for sensitivity as the aim of the studies
was either the identification of meat adulterations at the percent level or the
detection of gluten-containing wheat at a level of 200–400 mg/kg (10–20
mg/kg gliadin). Hence, even better sensitivities may have been achieved with
optimized PCR protocols. Most of the published and commercial
immunological methods for soya have been developed for the detection of
meat adulteration with soybean products having high limits of detection of
approximately 1000 mg/kg of soy protein (summarized by Poms et al.19). In
contrast to the numerous publications about the detection of peanut, which is
a legume like soybean, only two scientific references about the detection of
soybean with ELISA at a suitable level of 1–2 mg/kg have been reported so
far.33,34
One reason why soya has been difficult to target at the protein level using
ELISA methods is the great number of different soya products, such as
defatted soybean flakes, texturized soybean protein, protein isolates and
concentrates, and protein hydrolysates. The differences in processing can
result in a variety of altered peptides and protein breakdown products with
differing structural properties and therefore differing detectabilities. Therefore,
soya detection by PCR methods may be a possible alternative to immunological
methods if the detection of soybean DNA correlates with the presence of
soybean protein and peptides, respectively. So far, no celery-specific and
sensitive ELISA has been published. One reason for this may be the difficulty
in generating a celery-specific antiserum that allows sensitive detection of
celery in foodstuffs without cross-reacting to other species of the Apiaceae
family. By contrast, the PCR has great potential for the differentiation between
phylogenetically closely related species. In this context, celery, fish, crustaceans,
and various tree nuts may be interesting candidates to detect with PCR
methods in addition to the application of PCR for verification of ELISA
results or even as a substitute for ELISA.
For hazelnut, peanut, wheat, soya, and celery, PCR has already been
shown to be an alternative to ELISA assays. However, more investigations
have to be done to support these findings. Additionally, the applicability of
both methodologies has to be thoroughly investigated for each allergenic
food and in the most important food matrices. Thereafter, a final conclusion
can be drawn about the potential use of both techniques depending on the
allergenic food and the food matrix.
7.6 References
1. Hefle, S L, Bush, R K, Yunginger, J W and Chu, F S (1994) ‘A sandwich enzyme-
linked immunosorbent assay (ELISA) for the quantitation of selected peanut proteins
in foods’, J Food Prot, 57, 419–423.
2. Yeung, J M and Collins, P G (1996) ‘Enzyme immunoassay for determination of
peanut proteins in food products’, J AOAC Int, 79, 1411–1416.
3. Holzhauser, T and Vieths, S (1999) ‘Indirect competitive ELISA for determination
of traces of peanut (Arachis hypogaea L.) protein in complex food matrices’, J Agric
Food Chem, 47, 603–611.
4. Holzhauser, T and Vieths, S (1999) ‘Quantitative sandwich ELISA for determination
of traces of hazelnut (Corylus avellana) protein in complex food matrixes’, J Agric
Food Chem, 47, 4209–4218.
5. Koppelman, S J, Knulst, A C, Koers, W J, Penninks, A H, Peppelman, H, Vlooswijk,
R, Pigmans, I, van Duijn, G and Hessing, M (1999) ‘Comparison of different
immunochemical methods for the detection and quantification of hazelnut proteins
in food products’, J Immunol Methods, 229, 107–120.
6. Hlywka, J J, Hefle, S L and Taylor, S L (2000) ‘A sandwich enzyme-linked
immunosorbent assay for the detection of almonds in foods’, J Food Prot, 63, 252–
257.
7. Hefle, S L, Jeanniton, E and Taylor, S L (2001) ‘Development of a sandwich enzyme-
linked immunosorbent assay for the detection of egg residues in processed foods’, J
Food Prot, 64, 1812–1816.
8. Pomes, A, Helm, R M, Bannon, G A, Burks, A W, Tsay, A and Chapman (2003)
‘Monitoring peanut allergen in food products by measuring Ara h 1’, J Allergy Clin
Immunol, 111, 640–645.
9. Allmann, M, Candrian, U, Höfelein, C and Luthy J (1993) ‘Polymerase chain reaction
(PCR): a possible alternative to immunochemical methods assuring safety and quality
of food’, Z Lebensm Unters Forsch, 196, 248–251.
10. Meyer, R, Chardonnens, F, Hübner, P and Lüthy, J (1996) ‘Polymerase chain reaction
(PCR) in the quality and safety assurance of food: detection of soya in processed
meat products’, Z Lebensm Unters Forsch, 203, 339–344.
11. Jankiewicz, A, Hübner, P, Bögl, K W, Dehne, L I, Vieths, S, Baltes, W and Lüthy, J
(1997) ‘Celery Allergy: PCR as a tool for the detection of trace amounts of celery in
processed foods’, Proceedings of Euro Food Chem IX, Interlaken, Switzerland, Sept
24–26.
12. Holzhauser, T, Wangorsch, A and Vieths, S (2000) ‘Polymerase chain reaction (PCR)
for detection of potentially allergenic hazelnut residues in complex food matrixes’,
Eur Food Res Technol, 211, 360–365.
13. Holzhauser, T, Stephan, O and Vieths, S (2002) ‘Detection of potentially allergenic
hazelnut (Corylus avellana) residues in food: a comparative study with DNA PCR-
ELISA and protein sandwich-ELISA’, J Agric Food Chem, 50, 5808–5815.
142 Detecting allergens in food
14. Dahinden, I, von Büren, M and Lüthy, J (2001) ‘A quantitative competitive PCR
system to detect contamination of wheat, barley or rye in gluten-free food for coeliac
patients’, Eur Food Res Technol, 212, 228–233.
15. Herman, L, Block, J D and Viane, R (2003) ‘Detection of hazelnut DNA traces in
chocolate by PCR’, Int J Food Sci Tech, 38, 633–640.
16. Hird, H, Lloyd, J, Goodier, R, Brown, J and Reece, P (2003) ‘Detection of peanut
using real-time polymerase chain reaction’, Eur Food Res Technol, 217, 265–268.
17. Sandberg, M, Lundberg, L, Ferm, M and Malmeheden Yman, I (2003) ‘Real time
PCR for the detection and discrimination of cereal contamination in gluten free
foods’, Eur Food Res Technol, 217, 344–349.
18. Stephan, O and Vieths, S (2004) ‘Development of a real-time PCR and a sandwich
ELISA for detection of potentially allergenic trace amounts of peanut (Arachis
hypogaea) in processed foods’, J Agric Food Chem, 52, 3754–3760.
19. Poms, R E, Klein, C L and Anklam, E (2004) ‘Methods for allergen analysis in food:
a review’, Food Addit Contam, 21, 1–31.
20. Taylor, S and Nordlee, J (1996) ‘Detection of food allergens’, Food Technol, 50,
231–234.
21. Hourihane, J O B, Kilburn, S A, Nordlee, J A, Hefle, S L, Taylor, S L and Warner,
J O (1997) ‘An evaluation of the sensitivity of subjects with peanut allergy to very
low doses of peanut protein: a randomized, double-blind, placebo-controlled food
challenge study’, J Allergy Clin Immunol, 100, 596–600.
22. Wensing, M, Penninks, A H, Hefle, S L, Koppelman, S J, Bruijnzeel-Koomen, C A
and Knulst, A C (2002) ‘The distribution of individual threshold doses eliciting
allergic reactions in a population with peanut allergy’, J Allergy Clin Immunol, 110,
915–920.
23. CEN, ‘Foodstuffs – Methods of analysis for the detection of genetically modified
organisms and derived products – Nucleic acid extraction’ (ISO/DIS 21571) prEN
ISO 21571, Brussels, CEN.
24. SLMB, Schweizerisches Lebensmittelbuch, Kapitel 52B, Molekularbiologische
Methoden, Bern, Bundesamt für Gesundheit Facheinheit Lebensmittelsicherheit (2003).
25. Newton, C R and Graham, A (1997) PCR (Introduction to biotechniques), Oxford,
Bios Scientific Pub Ltd.
26. Sambrook, J and Russels, D W (2001) Molecular Cloning: A Laboratory Manual,
Cold Spring Harbor Press, NY.
27. DIN, ‘Part 60: Polymerase chain reaction (PCR): Terminology, general method-
specific requirement’, Diagnostics of infectious diseases and diseases of the immune
system in serology and molecular biology, DIN 589667-60, Berlin, Beuth, 2001.
28. CEN, ‘Foodstuffs – Methods of analysis for the detection of genetically modified
organisms and derived products – Qualitative nucleic acid based methods’, (ISO/
DIS 21569) prEN ISO 21569 Brussels, CEN.
29. Meuer, S, Wittwer, C and Nakagawara, K (eds.), (2003) Rapid Cycle Real-time PCR
– Methods and Applications, Berlin, Springer.
30. CEN, ‘Foodstuffs – Nucleic acid based methods of analysis for the detection of
allergens – General requirements and definitions’, (ISO/DIS 24276) prEN ISO 24276,
Brussels, CEN.
31. Longo, M C, Berninger, M S and Hartley, J L (1990) ‘Use of uracil DNA glycosylase
to control carry-over contamination in polymerase chain reactions’, Gene, 93, 125–
128.
32. Stephan, O, Weisz, N, Weiser, T, Rabe, B, Vatterott, W and Vieths, S (2004) ‘Protein
quantification, sandwich ELISA, and real-time PCR used to monitor industrial cleaning
procedures for contamination with peanut and celery allergens’, J AOAC Int, 87,
1448–1457.
33. Yeung, J M and Collins, P G (1997) ‘Determination of soy proteins in food products
by enzyme immunoassay’, Food Technol Biotechnol, 35, 209–214.
Polymerase chain reaction (PCR) methods 143
8.1 Introduction
Proteomics has recently received much attention. The word ‘proteomics’
encompasses a family of technologies with which to determine the complement
of proteins in a cell, tissue or organism. Currently, these technologies include
multi-dimensional separations via electrophoresis or liquid chromatography;
various methods for protein identification including mass spectrometry; arrays
to map protein–protein interactions; and bioinformatics to analyze massive
data sets.
Proteomics is evolving from its roots in two-dimensional electrophoresis.
However, at present, the electrophoretic approach still represents a valuable
technique for separating proteins. Proteomics is especially useful for the
identification of IgE-binding proteins for several reasons. First, conditions
for solubilization of proteins are very aggressive, involving combinations of
urea, thiourea, non-ionic detergents and strong reducing agents. These reagents
increase the likelihood of detecting allergens. Second, spots in two-dimensional
separations are more likely to be homogenous compared to protein bands in
one-dimensional sodium dodecyl sulphate polyacrylamide gel electrophoresis
(SDS-PAGE) separations. In addition, two-dimensional separation provides
substantial information in the form of isoelectric point (pI), molecular weight
and heterogeneity (charge and/or size variants) of the IgE-binding component.
This information makes it possible to substantially narrow the search for the
protein’s identity. Finally, the value of a proteomics approach can be seen in
the number of newly discovered allergens (discussed below). While the number
of reports using proteomics to investigate IgE-binding proteins is low, most
have uncovered ‘novel’ allergens.
Proteomic assessment of allergens in food 145
IEF separation
Complex mixtures of proteins can be separated by IEF. The unique balance
of positive and negatively charged amino acids determines a protein’s isoelectric
point (pI). A protein’s pI corresponds to that pH at which the positive and
negative charges sum to zero. When the pH is equal to a protein’s pI, it is
electrically neutral and its migration in an electric field is halted. Typically,
IEF is conducted in the presence of high concentrations of urea (6–8 M),
non-ionic detergents (e.g. (3-[3-cholamidopropylammonio]-1-propanesulfonate
– CHAPS) and reducing agent (e.g. dithiothreitol – DTT) to promote solubility.
A pH gradient is established typically from 3.0–10.0 using immobilized
ampholines in a polymer-based IPG strip.
The second dimension separation is usually performed via SDS-PAGE
following appropriate equilibration. The combination of separations based
on different mechanisms of selectivity (pI vs relative mass) results in enhanced
resolution of component proteins. Thus 2-DE has distinct advantages for
characterizing the IgE-binding components in complex mixtures of proteins.
8.2.2 Quantitation
Significant improvements in staining methods and computer-driven image
analysis routines have greatly improved the ability to quantitatively assess
differences between samples. However, while the image analysis packages
may provide numerical evaluations, careful use is needed to ensure accuracy
(Miller et al., 2001; Miura, 2001).
Quantitation of stained proteins in gels requires several steps to establish
the validity of the measurement. Determination of linear range of detection
146 Detecting allergens in food
with that reported by Pastorello et al. (2001) shows differences sufficient for
them to be regarded as distinct allergens. The sesame 2S albumin reported
by Beyer et al. (2002b) is higher in sulfur-containing amino acids and is
deemed to be a novel allergen. It has been designated in the allergen
nomenclature as Ses i 2.
8.5 Conclusions
Proteomics is an increasingly valuable tool in allergy research. Presently, it
offers enhanced potential for the detection and identification of IgE-binding
proteins in complex mixtures. All of the examples included in this review
showed that proteomic analysis resulted in new and more detailed information
regarding the profile of allergens in the materials examined. Proteomics was
also shown to be of value in the quantitative assessment of allergen content
in genetically modified soybeans. In the future, the development of new
156 Detecting allergens in food
8.6 References
Asturias, J A, Arilla, M C, Gomez-Bayon, N, Martinez, A, Martinez, J and Palacios, R
(1998) ‘Sequencing and high level expression in Escherichia coli of the Tropomyosin
allergen (Der p 10) from Dermatophagoides pteronyssinus’, Biochim Biophysic Acta,
1397, 27–30.
Awazuhara, H, Kawai, H, Baba, M, Matsui, T and Komiyama, A (1998) ‘Antigenicity of
the proteins in soy lecithin and soy oil in soybean allergy’, Clin Exp Allergy, 28, 1559–
1564.
Berggren, K, Chernokalskaya, E, Steinberg, T H, Kemper, C, Lopez, M F, Diwu, Z,
Haugland, R P and Patton, W F (2000) ‘Background-free high sensitivity staining of
proteins in one- and two-dimensional sodium dodecyl sulfate polyacrylamide gels
using a luminescent ruthenium complex’, Electrophoresis, 21, 2509–2521.
Beyer, K, Grishina, G, Bardina, L, Grishin, A and Sampson, H A (2002a) ‘Identification
of an 11S globulin as a major hazelnut food allergen in hazelnut-induced systemic
reactions’, J Allergy Clin Immunol, 110, 517–523.
Beyer, K, Bardina, L, Grishina, G and Sampson, H A (2002b) ‘Identification of sesame
seed allergens by 2-dimensional proteomics and Edman sequencing: seed storage
proteins as common food allergens’, J Allergy Clin Immunol, 110, 154–159.
Chen, J, Lee, C L, Shen, Y, Smith, R D and Baehrecke, E H (2002) ‘Integration of
capillary isoelectric focusing with capillary reversed-phase liquid chromatography for
two-dimensional proteomics separation’, Electrophoresis, 23, 3143–3148.
Figeys, D and Pinto, D (2001) ‘Proteomics on a chip: promising developments’,
Electrophoresis, 22, 208–216.
Gu, X, Beardslee, T, Zeece, M G, Sarath, G and Markwell, J (2001) ‘Identification of
IgE-binding proteins in lecithin’, Int Arch Allergy Immunol, 126, 218–225.
Helm, R M, Cockrell, G, Herman, E, Burks, A W, Sampson, H A and Bannon, G A (1998)
‘Cellular and molecular characterization of a major soybean allergen’, Int Arch Allergy
Immunol, 117, 29–37.
Helm, R M, Cockrell, G, West, C M, Herman, E, Burks, A W, Sampson, H A, Bannon, G
A and Burks, A W (2000) ‘Mutational analysis of the IgE-binding epitopes of P34/Gly
m 1’, J Allergy Clin Immunol, 105, 378–384.
Herman, E M, Helm, R M, Jung, R and Kinney, A J (2003) ‘Genetic modification removes
an immunodominant allergen from soybean’, Plant Physiol, 132, 36–43.
Hirschwehr, R, Valenta, R, Ebner, C, Sperr, F, Valent, W R, Rohac, M, Rumpold, H,
Scheiner, O and Kraft, D (1992) ‘Identification of common allergenic structures in
hazel pollen and hazelnuts: a possible explanation for sensitivity to hazelnut in patients
allergic to tree pollen’, J Allergy Clin Immunol, 909, 927–936.
Lopez, M F, Berggren, K, Chernokalskaya, E, Lazarev, A, M, Robinson, T H and Patton,
W F (2000) ‘A comparison of silver stain and SYPRO ruby protein gel stain with
respect to protein detection in two-dimensional gels and identification by peptide
mass profiling’, Electrophoresis, 21, 3673–3683.
Mann, M, Hendrickson, R C and Pandey, A (2001) ‘Analysis of proteins and proteomes
by mass spectrometry’, Annl Rev Biochem, 70, 437–474.
Miller, M D, Acey, R A, Yu-Tzu-Lee, L and Edwards, A J (2001) ‘Digital imaging
considerations for gel electrophoresis analysis systems’, Electrophoresis, 22, 791–
800.
Miura, K (2001) ‘Imaging and detection technologies for image analysis in electrophoresis’,
Electrophoresis, 22, 801–813.
Proteomic assessment of allergens in food 157
9.1 Introduction
During recent years, awareness of food allergens has increased, resulting in
a growing demand for reliable, rapid and sensitive analytical methods to
detect them (Poms et al., 2004). Tracing and quantifying allergens in food is
often a challenging task. Allergens can be present in a wide variety of food
types, with different compositions and chemical properties. Some food
components may interfere with the assays. Food processing of different
kinds makes the extraction difficult. The sensitivity requirement is also high
since even very low doses may elicit allergic reactions. Tracing allergens in
food could very much be compared to finding a needle in a haystack. To be
able to reliably analyze as many allergens as possible in as many sample
matrixes as possible, a whole battery of methods is needed. Different analytical
techniques may complement each other.
As a research project, the Swedish National Food Administration has
started to investigate a novel analytical technique based on a surface plasmon
resonance (SPR) biosensor for food allergen detection. The objective is to
develop rapid and easy-to-use assays for common food allergens that would
complement existing techniques already employed, such as rocket
immunoelectrophoresis (RIE), enzyme-linked immunoassays (ELISAs) and
real-time PCR. Besides enhanced sensitivity, increased sample throughput is
a major driving force behind the development of the automated platform.
The high speed, ease-of-use and high degree of automation should also be
attractive features for food manufacturers who want to analyze ingredients
or final food products.
This chapter summarizes the literature on SPR technology related to food
Detecting with a surface plasmon resonance immunoassay 159
Optical
detection Intensity
Light- unit
source
I
I II
Polarised II Angle
light Reflected
light Resonance
Prism II signal
Sensor chip
with gold film Time
I
Sensorgram
Flow channel
Fig. 9.1 Principles of surface plasmon resonance detection. Due to the SPR
phenomenon the reflected light will have an intensity minimum at a specific angle that
is continuously monitored. Binding of biomolecules to the sensor surface results in a
shift in the resonance angle.
Sensor response
Time
Buffer Buffer Buffer Buffer
Sample Secondary Regeneration
antibodies
Fig. 9.2 Sensorgram for one analysis cycle. This example shows a typical injection
sequence for a sandwich format assay, with secondary antibody injection after the
sample injection. Switching of solutions is indicated by arrows. The pictures above
illustrate schematically the surface coverage at different stages.
food allergens (two minutes, 10 μg mL–1). The next essential step is to find
a suitable regeneration solution able to remove all the bound analyte without
damaging the surface. This solution is injected after each sample. When a
good regeneration solution has been found, the same surface can be used to
analyze 100 or more samples, resulting in a very economical use of antibodies
and sensor chips. In the allergen assays developed so far, 0.1 M hydrochloric
acid has been proved to regenerate the surface quite well and can be used
initially for newly-developed assays. To further optimize the regeneration
for the specific antibody used, it is advisable to test the regeneration efficiency
with 10 mM glycine-HCl with pH ranging from 3 down to 1.5 in a series of
analysis cycles run on a newly immobilized surface. In each cycle, a high
concentration of allergen (e.g. 10 μg mL–1) is injected followed by the
regeneration solution. The mildest regeneration solutions (highest pH) should
be run first, and each condition should be repeated for five cycles before
switching to the next. A plot showing the response increase during the allergen
injection versus cycle number can then be used to determine the optimal
regeneration solution. The regeneration solution that gives the highest and
most stable analyte response should be chosen for the assay. Software guides
are available for this type of assay development in the Biacore Q system.
Surface preparation
Allergen-specific rabbit IgG from the immune sera was isolated by
immunoaffinity chromatography on a NHS-activated HiTrap™ column
(GE Healthcare, Uppsala, Sweden) coupled with the respective allergens
(affinity purification). In some of the initial experiments total IgG, as obtained
166 Detecting allergens in food
150
100
50
0
0 200 400 600 800 1000 1200
Concentration (ng/mL–1)
Fig. 9.3 Calibration curves for beta-lactoglobulin, ovomucoid and hazelnut proteins.
Two-minute injections were used.
solutions. This initial investigation showed that the sensitivity of the SPR
detector is sufficient for detection of food allergens. Compared to rocket
immunoelectrophoresis using the same antibodies the sensitivity was at least
ten times more sensitive.
Analysis of food extracts, however, showed that the assays were not yet
specific enough. Some types of food extracts bound non-specifically to the
surface. Since everything that binds to the surface will contribute to the
sensor signal in SPR, non-specific binding will result in false-positive results
in measurements based on total sample binding. By comparing the response
on surfaces with or without immobilized antibodies, it was shown that most
of the non-specific binding occurred to the antibodies, not to the surface. The
dextran-coated surface was quite inert. Extracts from bread and other bakery
products never gave high non-specific binding. Chocolate extracts, on the
other hand, were very important for the hazelnut assay, often bound to a very
high degree making sensitive measurements of hazelnuts impossible.
One reason for the non-specific binding to the immobilized antibodies is
that the Protein G-purified antiserum contains a mixture of IgG towards all
food proteins that rabbits have encountered during their life. One way to
enhance the specificity is to isolate only specific antibodies (see below).
the specific binding of hazelnut proteins remained almost the same. Still, the
non-specific binding was too high for several types of sample extracts.
Increasing the concentration of salt and increasing the pH of the buffer
further reduced the non-specific binding from chocolate extracts. However,
these buffer modifications also reduced the specific binding. Increasing the
amount of surfactant (P20) reduced the non-specific binding for dark chocolate
extracts (Jonsson and Hellenäs, 2001). In contrast, increasing the surfactant
concentration increased the non-specific binding of milk chocolate extracts.
The best way of reducing the high non-specific binding for dark chocolate
extracts was found to be addition of PVP during the extraction. This also
greatly increased recovery of hazelnut proteins in spiked samples.
This examination indicates that it is possible to optimize the assay conditions
in the SPR assays so that sensitive measurements can be performed even in
food matrices that contain many interfering substances. When the composition
of the analyzed food extract is known and similar from time to time, this can
be used to obtain a very rapid analysis of samples. For instance, this could
be used in continuous monitoring of a manufacturing process. In most situations,
however, the compositions of the samples are not very well known and differ
much from sample to sample. Under such assay circumstances, it is very
difficult to find conditions that will give very little non-specific binding for
all types of sample extracts. The best way of circumvent this problem was
found to be extending the assay slightly by adding an injection of antibodies
after the sample, creating a ‘sandwich’ format.
Table 9.1 Cross-reactivity expressed as ppm hazelnut protein in solid food sample. All
samples were diluted a hundred times before analysis. Concentrations assigned as >100
indicate that the level was above the measurement range when normal assay conditions
were applied. Only hazelnut extract was further diluted to allow higher concentrations to
be determined.
currently used for routine analysis at the Swedish National Food Administration,
the SPR hazelnut assays were ten-fold more sensitive. The limit of detection
was in the same range as commercially available kits for hazelnuts.
Sample Peanut butter Chocolate Calculated RIE peanut SPR BioKits™ EIA Ridascreen
amount of protein immunoassay peanut protein EIA peanut
peanut protein (ppm) peanut protein (ppm) protein
(ppm) (ppm) (ppm)
Tropomyosin assay
Recently, crab tropomyosin was purified and antisera raised in a rabbit as
well as in eggs (egg yolk) by immunizing two hens. The rabbit IgG as well
as the IgY from egg yolks was immunopurified on a HiTrap™ column,
coupled with tropomyosin. Rabbit IgG or hen IgY was coupled to CM3
chips according to the established procedure. The chips were tested both in
a direct and in a sandwich assay with different solutions of tropomyosin,
with crab, shrimp and model foods, composed of surimi containing different
amount of crab meat. The lowest concentration detected in a tropomyosin
solution was 0.2 μg mL–1. In model products, a level of 0.063% crab meat
in surimi could be detected, corresponding to a tropomyosin concentration
of 13 ppm tropomyosin in solid food. The response both in the direct as well
as in the sandwich assay was virtually the same. By using IgY bound to the
surface and rabbit IgG as the secondary antibody, the non-specific binding to
the chip surface was reduced.
9.6 Conclusions
The SPR technique has proven to be a useful tool for the analysis of allergens
in food. Analysis of six important food allergens has been demonstrated.
SPR provides non-destructive detection that requires no labeling or any other
type of modification of the analytes detected. This gives unsurpassed control
during assay development and routine analysis, not available in established
methods of food allergen detection. Limits of detection in the low ppm level
can be obtained and with the sandwich format, very specific assays can be
achieved.
The main advantage of the SPR methodology is the short analysis time
per sample. This is important for large series of samples and for real-time
process monitoring. In the future, SPR analysis of food allergens may be
extended to analyze several allergens in the same extract simultaneously by
injection over several surfaces immobilized with antibodies raised against
different species.
Detecting with a surface plasmon resonance immunoassay 173
9.8 References
Byun, M W, Kim, J H, Lee, J W, Park, J W, Hong, C H and Kang, I J (2000) ‘Effects of
gamma radiation on the conformational and antigenic properties of a heat-stable major
allergen in brown shrimp’, J Food Prot, 63, 940–944.
Daly, S J, Keating, G J, Dillon, P P, Manning, B M, O’Kennedy, R, Lee, H A and Morgan,
M R (2000) ‘Development of surface plasmon resonance-based immunoassay for
aflatoxin B(1)’, J Agric Food Chem, 48, 5097–5104.
Feriotto, G, Borgatti, M, Mischiati, C, Bianchi, N and Gambari, R (2002) ‘Biosensor
technology and surface plasmon resonance for real-time detection of genetically modified
roundup ready soybean gene sequences’, J Agric Food Chem, 50, 955–962.
Feriotto, G, Gardenghi, S and Gambari, R (2003) ‘SPR-based assays for real-time detection
of genetically modified organisms’, Biacore J, 2, 5–8.
Haasnoot, W, Olieman, K, Cazemier, G and Verheijen, R (2001) ‘Direct biosensor
immunoassays for the detection of nonmilk proteins in milk powder’, J Agric Food
Chem, 49, 5201–5206.
Holzhauser, T and Vieths, S (1999) ‘Quantitative sandwich ELISA for determination of
traces of hazelnut (Corylus avellana) protein in complex food matrixes’, J Agric Food
Chem, 47, 4209–4218.
Indyk, H E and Filonzi, E L (2003) ‘Determination of immunoglobulin G in bovine
colostrum and milk by direct biosensor SPR-immunoassay’, J AOAC Int, 86, 386–393.
Jongerius-Gortemaker, B G M, Goverde, R L J, van Knapen, F and Bergwerff, A A (2002)
‘Surface plasmon resonance (BIACORE) detection of serum antibodies against
Salmonella enteritidis and Salmonella typhimurium’ J Immunol Methods, 266, 33–44.
Jonsson, H and Hellenäs, K-E (2001) ‘Optimizing assay conditions in the detection of
food allergens with Biacore’s SPR technology’, Biacore J, 1(2), 16–18.
Jönsson, U, Fägerstam, L, Ivarsson, B, Johnsson, B, Karlsson, R, Lundh, K, Löfås, S,
Persson, B, Roos, H and Rönnberg, I (1991) ‘Real-time biospecific interaction analysis
using surface plasmon resonance and a sensor chip technology’, Biotechniques, 11,
620–627.
Klein, E, Baudner, S and Günther, H O (1985) ‘Immunochemische Bestimmung des
Hasselnussproteins mit Hilfe der Elektroimmundiffusion nach Laurell. I. Metteilungen
der optimierten Methode’, Z Lebensm Unters Forsch, 180, 30–35.
Koppelman, S J, Knulst, A C, Koers, W J, Penninks, A H, Peppelman, H, Vlooswijk, R,
Pigmans, I, Van Duijn, G and Hessing, M (1999) ‘Comparison of different
immunochemical methods for the detection and quantification of hazelnut proteins in
food products’, J Immunol Methods, 229, 107–120.
Liedberg, B, Nylander, C and Lundström, I (1983) ‘Surface plasmon resonance for gas
detection and biosensing’, Sensors and Actuators, 4, 299–304.
Löfås, S (1995) ‘Dextran modified self-assembled monolayer surfaces for use in
biointeraction analysis with surface plasmon resonance’, Pure Appl Chem, 67, 829–
834.
174 Detecting allergens in food
10.1 Introduction
In recent years, the number of solid phase-based immunoassays available
has further increased. Most of these are enzyme-linked immunosorbent assays
(ELISAs). Among the newly introduced tests, a significant number have
been developed either to identify the presence of allergenic substances in
e.g. food preparations, such as peanuts or hazelnuts in cookies, or to measure
in sera the antibodies (IgE) that patients may have developed in response to
repeated exposure to such allergens. These types of tests show a very high
specificity and a high sensitivity.
In general, an analytical methodology is sought where the detection of
offending foods or allergenic proteins needs to be managed with high specificity,
sensitivity and rapidity. In the following, typical demanded characteristics
for the methodology are listed.
• Sufficient specificity – should allow the detection of the analyte in question
in a wide variety of matrices including processed foods.
• Satisfactory sensitivity – detection in the range of 1–5 mg/kg is necessary.
• Rapid detection of contamination – short reaction time for correction
before large volumes of mislabelled products have been made or to give
the consumer a yes/no decision within an adequate time span.
• Ease-of-use – no specially trained employees needed or less experienced
persons can perform the assay.
• For industrial purposes – continuous sampling could be advantageous.
Most of the immunochemical methods mentioned above are considered
rapid, but there is still great demand for easy-to-use immunochemical tests
176 Detecting allergens in food
for the detection of traces of allergenic proteins in food. The faster commercial
ELISA kits for allergens on the market take only 30 minutes, and some of
them (qualitative determinations) do not require a high level of expertise, as
they have dropper bottles and visual comparisons. However, the more sensitive,
quantitative tests take a trained technician to carry them out; the desire to
improve on speed and ease-of-use, especially in non-research settings, has
prompted researchers to look for faster and simpler procedures. Thus, new
rapid test platforms have been developed, such as the chromatographic test
strip (lateral flow test). This is widely used for example in so-called ‘home
tests’, such as hCG measuring pregnancy tests, which do not require assistance
from trained technicians, but can be carried out by the consumers themselves.
The first generation of such tests was more or less based on variations on the
classic ‘dot blot’ test. From the experience gained with these early rapid
assays, much was learned regarding attainable sensitivities, and suitable
read-out systems. However, they did not take full advantage of the increased
kinetics, due to close contact of high concentrations of the analyte with
antibody. More recently, a second generation of immunomigration media
has been developed which exploit the micropores in nitrocellulose or nylon
membranes. The success of this new generation of so-called ‘lateral flow’
tests is based on the fast flow of fluids running through the membrane
enabling the application of various immunoreactants at different locations
along the membrane strip. There are currently four dipstick-type tests
commercially available, two for detection of peanut and two for detection of
gluten – these will be discussed later in the chapter.
10.2 Antibodies
10.2.1 Antibody requirements
There is more to designing one-step tests than meets the eye. Components
proven to be useful in ELISA are not necessarily adequate for one-step tests.
The environment in which they have to bind in ELISA is totally different
from that in one-step tests – in the latter all components are acting
simultaneously and within a very short time-frame. Thus, selecting antibodies
for a one-step test requires their affinities to be sufficient and yet mutually
tolerant so that they will allow each other to participate in forming a visible
complex within the short time that they flow across the test and control lines.
Also, it is more critical that they do not compete for the same epitopes. In
addition, gold labeling has a greater interference with antibodies, where
binding affects overall charge and secondary and tertiary structure more than
labeling with enzymes or biotin. As an example, rabbit antibodies do not
retain gold for more than a few days, while gold-labeled goat antibodies are
very stable.
The use of lateral flow devices to detect food allergens 177
Sample filter
Gold pad
Test line
Control line
• Sample filter
• Gold pad
• Membrane
• Test line
• Control line Membrane
• Absorption filter
• Store at 10–28 °C
Absorption filter
S S S
• Antigen-detection
• Antibody-detection
• Inhibition assay
10.7 References
AOAC Research Institute (2003) ‘Performance tested methods’, Inside Laboratory
Management, Sept/Oct, 19–22.
Mills C E N, Potts, A, Plumb, G W, Lambert, N and Morgan, R A (1997) ‘Development
of a rapid dipstick immunoassay for the detection of peanut contamination of food’,
Food Agric Immunol, 9, 37–50.
Part III
11.3.3 IgE-immunoblotting
An IgE-dot-immunoblotting method using peanut-allergic sera was described
by Schappi et al. (2001) to detect the presence of peanut proteins in foods.
The standards were made from roasted peanuts. The detection limit was
approximately 50 ppm, too high for practical allergen management. The use
of human sera and of urea in the extraction procedure limits the practical use
of this type of method in food-processing facilities. The authors found that
19 of 46 commercial food products had undeclared peanut residues, ranging
from 0.05 – >1% peanut (500 ppm – >10 000 ppm).
While IgE-based techniques such as RAST and enzyme allergosorbent
test (EAST inhibition) and IgE-immunoblotting are useful techniques that
can be used in many clinical and research settings (see Chapters 4 and 5), it
must be stressed that they require well-characterized peanut-allergic human
sera, a substance which needs special handling, and the use of this reagent is
precluded in food processing environments. Also, the specificity of IgE from
allergic individuals can vary considerably, making standardization difficult.
plate, and the authors indicated that they could store coated plates for
24 months. Yeung and Collins also spiked food samples at 2.5, 5, 10, and
20 ppm; the spiking resulted in observed recoveries of 68–90%. They reported
no cross-reactivity but tested samples for this in very low protein amounts
(< 40 ug/mL). They found that their rabbit anti-roasted peanut antibody
produced the most effective ELISA. They analyzed 15 non-peanut chocolate
bars and three types of sesame crackers and found that none of them had
undeclared peanut residues. This ELISA method reportedly had a detection
limit of 400 ppb.
Koppelman et al. (1996) described a sandwich-type ELISA using rabbit
polyclonal antibodies against partially purified Ara h 1 derived from raw
peanuts. This sandwich ELISA utilized the same antibody for detector and
capture; part of the antibody was labeled with horseradish peroxidase to be
the detector antibody. In studies, it was observed that fried peanuts were
somewhat less reactive in the ELISA, but the method did not show cross-
reactivity with the majority of legumes and other foods tested. The highest
concentration of almond and cashew tested (500 ug/mL) showed a slightly
positive signal, but a ten-fold dilution removed it. Recoveries in various
matrices ranged from 35–75%. The investigators also found that of 25 non-
peanut foods surveyed, none had detectable peanut content. The assay was
capable of measuring raw or roasted peanut, and the reported detection limit
was 0.1 ppm.
Much later, Pomes et al. (2003) published another sandwich ELISA based
on monoclonal antibody against Ara h 1. In this study, Ara h 1 could not
always be recovered in spiked chocolate samples nor always detected in the
presence of chocolate. However, it was found that in general, Ara h 1 content
correlated with a commercial ELISA method (Veratox® for Peanut – Neogen
Corporation, Lansing, MI), but the results were not compared to any other
commercial ELISA kits. The investigators ‘spiked’ ground chocolate with
ground peanut flour, but without using industrially manufactured standards,
the true extraction efficiency of the method cannot be truly ascertained. The
best extraction of Ara h 1 from chocolate was found to be with 5% non-fat
dry milk in phosphate-buffered saline for 2.5 hours at room temperature, an
approach that is too long for the rapid testing needs of the food industry, but
may be applicable elsewhere.
Keck-Gassenmeier et al. (1999) used a commercial sandwich-type ELISA
kit for peanut (Cortecs, now Tepnel Biosystems, Deeside, UK) and found
that adding 12.5% fish gelatin to the extraction buffer allowed recoveries in
chocolate of 60–90% and other food products of 48–88%; foods other than
chocolate in most cases did not require the use of the fish gelatin for improved
recoveries. A detection limit of 2 ppm could be obtained in chocolate. The
commercial test was qualitative only, but the investigators could make it
semi-quantitative by making their own standards; dark chocolate was melted
and then spiked with peanut butter. Cross-reactions were only assessed for
almonds, hazelnut, and milk and dark chocolate. Raw peanuts were found to
190 Detecting allergens in food
have a higher response in the assay. The detection limit of the modified assay
for detection of peanut proteins in dark chocolate was better than that claimed
by the manufacturer for the commercial kit.
Newsome and Abbott (1999) coupled an immunoaffinity column to capture
peanut proteins from foods, with a subsequent ELISA. The antiserum used
was raised against roasted peanuts. The antiserum was coupled to a commercial
gel preparation and they then did an ELISA according to the protocol of
Yeung and Collins (1996). The authors said that spiked chocolate gave
recoveries of 72–84%, and that the minimum detection limit was 0.1 ppm,
but they did not provide many details on the spiking.
Holzhauser and Vieths (1999) also developed an indirect ELISA method
using commercial anti-raw peanut rabbit antisera (absorbed to remove possible
soy reactivity) and ground roasted peanuts as a standard. This method was
reported to have a detection limit of 2 ppm and showed no cross-reaction
concerns with the food ingredients tested. In five of 17 commercial food
products not declaring peanut, levels of peanut ranging from 2–18 ppm were
found.
Later, the same research group (Stephan and Vieths, 2004) reported both
a sandwich ELISA and a PCR method for detection of peanut residues. Both
sheep and rabbit anti-peanut antibodies were used in the development of the
ELISA and, while the authors did indicate that the antibodies were raised
against peanut extract, it was not reported whether the peanuts used were
raw or roasted. The methods were developed primarily for confection products,
and so cross-reactivity with some types of food ingredients, such as spices,
was not done. Industrially-manufactured standards of 10, 40, and 200 ppm
peanut were made in semi-sweet and whole milk chocolate. For the sandwich
ELISA, recoveries from whole milk chocolate were 80.6–141.9%, and for
semi-sweet, 64.3–110.9%. Fish gelatin was added to the extraction buffer as
recommended by Keck-Gassenmeier et al. (1999); however, interference
still was observed with chocolate even when this was employed. In a survey
of 33 products, two were found to have undeclared peanut residue, in the
range of 6.3–74 ppm.
A cloth-based immunoassay was described by Blais and Phillippe (2000).
The method utilized polyester cloth as the solid phase and the capture and
detector (peroxidase-labeled) antibody was chicken anti-peanut IgY. Peanut
immunogen prepared according to the Yeung and Collins (1996) method was
used to immunize chickens, followed by a fairly lengthy purification of IgY
from egg yolks. The authors felt that the cloth solid phase provided the
advantages of conferring a high surface area for rapid immunoreactions and
ease of washing between steps. Food samples connected with consumer
illnesses were analyzed using the cloth-ELISA; of 11 samples not declaring
peanut, nine contained peanut residues and were positive in the cloth-ELISA.
Levels were reported to be 1.2–116 ppm (determined using the Yeung and
Collins (1996) quantitative ELISA method). Cross-reactivity data was not
shown; chocolate seemed to give significant interference in the method. The
Methods for detecting peanuts in food 191
investigators found that skim milk powder alleviated the problem, but some
samples were positive when analyzed in a commercial peanut kit but not
detected using the Yeung and Collins (1996) method – however, results from
the cloth-ELISA and the commercial peanut kit did match. The authors
reported that the detection limit of the cloth-ELISA was less than 1 ppm
peanut protein for some matrices.
As an outgrowth of Koppelman et al.’s (1996) work (discussed above),
R-Biopharm AG (Darmstadt, Germany) has developed a faster version of
their peanut ELISA assay (RIDASCREEN®), which can be completed in
about 30 minutes (Immer et al., 2004). Other commercial kit companies
have also reduced the assay time of their peanut ELISA peanut kits to a
similar level.
Test), and other diagnostic kit companies are planning to produce kits in this
type of format also.
Two multiplex methods have been described, based on earlier work by the
authors. Ben Rejeb et al. (2002) developed a multi-analysis semi-quantitative
ELISA for the detection of peanut, almond, cashew, Brazil nut, and hazelnut.
The reported detection limit was 2 ppm for all allergens. Blais et al. (2003)
continued with their use of IgY and cloth in a multiple immunoassay method
(as described above) for peanut, Brazil nut, and hazelnut. The claimed detection
limit for peanut for this multiplex method was 0.1 ppm. The multiplex approach
offers the advantages of simple and rapid testing for various allergens
simultaneously, allowing for an initial preliminary screening to identify which
allergenic residues are present, to be followed up with a confirmatory test,
such as a specific ELISA method. The practicality of and need for this type
of approach for food manufacturers have yet to be evaluated fully.
In the only study to date addressing simply chocolate contamination,
Vadas and Perelman (2001) performed a limited survey of retail chocolate
bars using the Neogen Veratox® for Peanut test kit; results showed that eight
of 46 samples contained undeclared peanut residues, ranging from 7.9–69.6
ppm peanut.
Table 11.1 Comparison of four commercial quantitative peanut kits using manufactured
peanut in dark chocolate samples
0 ppm < 2.5 ppm < 2.5 ppm 0.33 ± 0.1 ppm 0 ppm
0.5 ppm < 2.5 < 2.5 0.93 ± 0.25 0.16 ± 0.02
1 ppm < 2.5 < 2.5 1.57 ± 0.29 0.11 ± 0.11
2 ppm < 2.5 < 2.5 2.82 ± 0.38 0± 0
10 ppm 12.87 ± 5.6 6.67 ± 0.31 12.1 ± 2.8 0.16 ± 0.1
20 ppm 23 ± 6.1 12.0 ± 0.67 25.1 ± 7.1 0.27 ± 0.1
50 ppm 52.5 ± 16.7 66.4 ± 15.0 63.5 ± 9.7 1.62 ± 1.4
100 ppm 121.0 ± 27.6 117.4 ± 34.2 127.1 ± 28.5 3.65 ± 3.9
one more positive sample than their sandwich ELISA did. The authors
concluded that PCR and ELISA gave similar results for detection of peanut
residues in foods, but pointed out that in some cases, such as pickled foods,
oils, and canned foods, the results will not be comparable due to DNA
degradation during processing. There are two commercial PCR methods
currently on the market for the detection of peanut residues (Table 11.2).
Indirect approaches such as PCR-based methods detect a marker for the
allergenic fraction rather than allergen itself. For food ingredients that are
only slightly processed, such as peanuts as topping on cookies, these indirect
approaches can be useful. However, when food ingredients undergo
fractionation steps such as concentration of the protein fraction as is done for
defatted peanut meal, indirect approaches run the risk that the outcome of
the analytical results could be underestimated. On the contrary, for peanut
PCR, results may be overestimated for food products from which the protein
fraction is removed, such as refined, bleached, deodorized peanut oils, which
are essentially devoid of protein, but can have DNA potentially present.
Company Method Name of kit Type LOD1 LLOQ2 Time3 Contact information
4
ELISA Systems ELISA Peanut Protein Residue Quantitative 0.5 ppm – 30 min www.elisasystems.net
ELISA Systems ELISA Peanut Residue Mass Quantitative 1 ppm 2.5 ppm 60 min www.elisasystems.net
Neogen ELISA Veratox® Quantitative5 1 ppm 2.5 ppm 30 min www.neogen.com
Neogen ELISA Alert® Qualitative 5 ppm – 30 min www.neogen.com
Neogen LFD6 Reveal® Qualitative 5 ppm – 10 min www.neogen.com
Pro-Lab ELISA Quantitative www.pro-lab.com
Diagnostics
R-Biopharm ELISA RIDASCREEN® Quantitative < 2.5 ppm 3.3 ppm 1.5 hr www.r-biopharm.com
Peanut
R-Biopharm ELISA RIDASCREEN® Quantitative 1.5 ppm 2.5 ppm 30 min www.r-biopharm.com
FAST Peanut
R-Biopharm PCR7 SureFood® Peanut Quantitative < 10 ppm – 2–3 hr www.r-biopharm.com
PCR-ELISA
R-Biopharm PCR SureFood® Peanut Qualitative 10–50 ppm – 30 min www.r-biopharm.com
Real-time PCR
Tepnel ELISA BioKits Quantitative < 0.1 ppm 1 ppm 1.25 hr www.tepnel.com
BioSystems Peanut Assay
Tepnel LFD BioKits RAPID Qualitative Low ppm < 50 ppm 6 min www.tepnel.com
BioSystems Peanut Test
Tepnel PCR BioKits Peanut Qualitative < 10 ppm – < 2 hr www.tepnel.com
BioSystems PCR Mastermix Pod
1
LOD = limit of detection; LOD is defined in different ways (whole food or protein); check with manufacturers for specifics
2
LLOQ = lower limit of quantitation
3
Time = excludes sample and reagent preparation times
4
ELISA = enzyme-linked immunosorbent assay
5
Any quantitative ELISA can be run qualitatively or semi-quantitatively
6
LFD = lateral flow device
7
PCR = polymerase chain reaction
Notes: Claims are as specified by manufacturers; all test performance is sample-dependent
Units of measurement are different for different kits; please check with manufacturers for specifics
Methods for detecting peanuts in food 197
11.6 References
Bannon, G A, Besler, M, Heflé, S L, Hourihane, J O’B and Sicherer, S H (2000) ‘Allergen
data collection: peanut (Arachis hypogea)’, Internet Symposium on Food Allergens, 2,
87–122.
Barnett, D and Howden, M E H (1984) ‘A rocket immunoelectrophoretic method for
the detection of heat-treated peanut protein’, Food Technology in Australia, 36, 510–
511.
Ben Rejeb, S, Abbott, M, Davies, D, Cleroux, C and Streng, D (2002) ‘Multi-screening
immunoassay for the detection of nut proteins in chocolate’, Proc. TNO International
Food Allergy Forum, Noordwijkerhout, The Netherlands, April 15–16, 58.
Blais, B W and Phillippe, L M (2000) ‘A cloth-based enzyme immunoassay for detection
of peanut proteins in foods’, Food Agric Immunol, 12, 243–248.
Blais, B W, Gaudreault, M and Phillippe, L M (2003) ‘Multiplex enzyme immunoassay
system for the simultaneous detection of multiple allergens in foods’, Food Control
14, 43–47.
Bock, S A and Atkins, F M (1990) ‘Patterns of food hypersensitivity during sixteen years
of double-blind, placebo-controlled food challenges’, J Pediatrics, 117, 561–567.
Bock, S A, Munoz-Furlong, A and Sampson, H A (2001) ‘Fatalities due to anaphylactic
reactions to foods’, J Allergy Clin Immunol, 107, 191–193.
Burks, A W, Sampson, H A and Bannon, G A (1998) ‘Peanut allergens’, Allergy, 53, 725–
730.
Burks, A W, Bannon, G A, Sicherer, S and Sampson, H A (1999) ‘Peanut induced
anaphylactic reactions’, Int Arch Allergy Immunol, 119, 165–172.
Clarke, M C A, Kilburn, S A, Hourihane, J O’B, Dean, K R, Warner, J O and Dean, T P
(1998) ‘Serological characteristics of peanut allergy’, Clin Exp Allergy, 28, 1251–
1257.
Emmett, S E, Angus, F J, Fry, J S and Lee, P N (1999) ‘Perceived prevalence of peanut
allergy in Great Britain and its association with other atopic conditions and with
peanut allergy in other household members’, Allergy, 54, 380–385.
Ewan, P W (1996) ‘Clinical study of peanut and nut allergy in 62 consecutive patients:
new features and associations’, Br Med J, 312, 1074–1078.
Freeman, A K, Morris, N J and Willich, R K (1954) ‘Peanut butter’, U.S. Dept. of
Agriculture Bulletin AIC–370.
Hefle, S L, Chu, F S, Yunginger, J W and Bush, R K (1994a) ‘A sandwich-type enzyme-
linked immunosorbent assay (ELISA) for quantitation of selected peanut proteins in
foods’, J Food Prot, 57, 419–423.
198 Detecting allergens in food
12.1 Introduction
Food allergy is an increasing health problem, and tree nuts belong to the ‘big
eight’ food allergens related to 90% of the food allergies (FAO, 1995). Tree
nut allergens are stable towards food processing and digestion. Similar stable
allergens have been identified in several seeds, and both seeds and nuts are
often used by the food industry owing to their taste. Food product diversification
since the 1990s has led to a vast number of recipes with nuts and seeds. This
was further stimulated by the observation that nuts have the beneficial health
effect of lowering cholesterol levels (Hu and Stampfer, 1999). Products that
do not contain nuts or seeds may be manufactured in facilities that also
process nuts and seeds, potentially forming a risk of cross-contact. Traces of
these allergens present in assumed nut- or seed-free products have led to
several severe allergic reactions. This indicates that methods to detect nut
and seed allergens are important tools for the food industry to control their
manufacturing and cleaning procedures. This chapter deals with the prevalence
of nut and seed allergy, threshold levels of allergens, the types of allergens
found in nuts and seeds, the effect of food processing on allergenicity, and
with tools available for detecting nuts and seeds. An overview of individual
methods will be given as well as a list of commercially available methods
that are presently used by the food industry. Since hazelnuts have been used
for a long period of time, especially in Western Europe, a lot of effort was
put into hazelnut detection. Pros and cons of the hazelnut detection methods
will be discussed, and we will look ahead to how detection of nut and seed
allergens may help the food industry in the future.
202 Detecting allergens in food
Nuts Seeds
Almond Celery
Brazil nut Linseed
Cashew Mustard
Chestnut Poppy seed
Hazelnut Rape seed
Macadamia Sesame seed
Pecan Sunflower seed
Pine nut
Pistachio
Walnut
Detecting tree nuts and seeds in food 203
to poppy seed, rape seeds, and linseed are less frequently described (Axelsson
et al., 1994; Kolopp-Sarda et al., 1997; Lezaun et al., 1998; Frantzen et al.,
2000; Galloway, 2000). Reactions vary for seeds, as for tree nuts, from mild
to severe, with anaphylaxis often described. Interestingly, the majority of
studies on the allergenicity of seeds and reports of allergic reactions come
from Europe. The European Commission published data on the prevalence
of seed allergies in 1998, and seeds were involved 1–2% of food-induced
anaphylaxis. There are no studies reporting prevalence of seed allergy in a
cohort of randomly selected individuals as has been done for tree nuts.
12.3 Thresholds
A lot of cases of unintentional ingestion of tree nuts and seeds have been
described, and traces of nuts and seeds are able to induce allergic reactions.
In some cases, the amount of offending allergen has been estimated. However,
double-blind, placebo-controlled food challenge studies are required to judge
how much allergen is too much (Hefle and Taylor, 2002). For hazelnut,
several researchers have investigated threshold in this way. Ortolani et al.
(2000) described thresholds of 168 mg–1.8 g hazelnut protein (Ortolani et
al., 2000) and Wensing et al. (2002) found thresholds as low as 1 mg hazelnut
protein (Wensing et al., 2002), indicating a considerable difference between
patients. The published thresholds are in line with case reports concerning
allergic reaction to hazelnut traces; milligram amounts of hazelnut protein
have been found to be enough to induce allergic reactions (Malmheden
Yman et al., 1994; Wensing et al., 2001a). Wensing et al. (2001b) showed
that the threshold for roasted hazelnuts was higher compared than that for
raw hazelnuts (Wensing et al., 2001b). This was further investigated by
Hansen et al. (2003) who found that only five of 17 hazelnut-allergic patients
that reacted clinically to raw hazelnut reacted to roasted hazelnut. Differences
in threshold levels were not determined (Hansen et al., 2003). Table 12.2
gives a brief overview of threshold data for some nuts and seeds obtained
using double blind, placebo-controlled food challenges. Note that for most
tree nuts and seeds, thresholds have not been established. Considering
thresholds in the mg range, and an average food intake of 50 g, as in the case
of a chocolate bar, a relevant limit for food allergen detection is in the order
of 10 ppm (Taylor and Nordlee, 1996). It should be noted, however, that
there may be food-allergic individuals who react to smaller amounts of
allergenic food. Clearly, there is an urgent need for threshold data, and the
Food Allergy Research and Resource Program of the University of Nebraska
(FARRP) coordinates multi-center studies on this topic (Hefle and Taylor,
2002; Taylor et al., 2002). A standardized protocol for performing threshold
studies has also been developed (Taylor et al., 2004). An overview of threshold
studies and the progress thereof is described in Chapter 1.
204 Detecting allergens in food
Table 12.2 Thresholds for nuts and seeds determined with double-blind, placebo controlled
food challenges
2.5
2
A 490
1.5
0.5
0
1 10 100 1000 10 000
Hazelnut protein (ng/ml)
Hazelnut protein Electroimmunodiffusion Polyclonal animal serum 100–1000 ppm Klein and Guenther, 1985;
Klein et al., 1985;
Malmheden Yman et al., 1994
Hazelnut allergens Immunoblot Hazelnut-allergic serum Not reported Teuber et al., 1997
Hazelnut protein Immunoblot Polyclonal rabbit serum 300 ppm Koppelman et al., 1999
Hazelnut allergens Inhibition ELISA Hazelnut-allergic serum 0.1 ppm Koppelman et al., 1999
Hazelnut protein Sandwich ELISA Polyclonal rabbit serum 0.1 ppm Koppelman et al., 1999
Hazelnut protein Sandwich ELISA Polyclonal rabbit serum 0.2 ppm Holzhauser and Vieths, 1999
Hazelnut DNA PCR-hybridization Specific DNA primers 10 ppm Holzhauser et al., 2000
Hazelnut protein Dot blot Egg yolk IgY 1 ppm Blais and Phillippe, 2000
Hazelnut protein Immunoblot Polyclonal rabbit serum 5 ppm Scheibe et al., 2001
Hazelnut protein Sandwich ELISA Egg yolk IgY 1 ppm Blais and Phillippe, 2001
Hazelnut protein Biacore immunoassay Polyclonal rabbit serum 2 ppm Jonsson and Hellenas, 2001
Hazelnut protein Dipstick (sandwich) Polyclonal rabbit serum 1 ppm (only Stephan et al., 2002
qualitative)
Hazelnut 2S albumin Basophil histamine release Hazelnut-allergic serum < 0.1 ppm Akkerdaas et al., 2002a
Hazelnut protein Sandwich ELISA Polyclonal rabbit serum < 0.1 ppm Akkerdaas et al., 2002b
Detecting tree nuts and seeds in food 209
food products that contain nuts other than hazelnut will also be avoided by
hazelnut-allergic consumers. Nevertheless, specificity is an important aspect
and, since tree nuts are phylogentically related, immunochemical cross-
reactivity leading to non-specificity is expected. The theoretical explanation
for immunochemical cross-reactivity is found in the antibodies recognizing
specific parts of the target proteins. Amino acid sequences of nut proteins are
sometimes very similar and homologous to a high extent as in the case for
the family of 2S storage proteins (Ampe et al., 1986; Teuber et al., 1998).
Antiserum may recognize epitopes on the immunogen as well as on homologous
proteins in other species. Cross-reacting epitopes usually have lower affinities
compared to the original epitopes, and the recognition of other species can
be orders of magnitude less. It is interesting to note that four different research
groups who raised hazelnut antisera independently describe the same cross-
reacting species; walnut appeared to cross-react in all raised antibodies,
while almond and cashew cross-reacted with three and two antisera, respectively
(Holzhauser and Vieths, 1999; Koppelman et al., 1999; Blais and Phillippe,
2001; Scheibe et al., 2001). One antiserum recognized pumpkin seed, and
another reacted with pine nut and peanut, although the latter two were a
100 000 fold less compared to hazelnut (Koppelman et al., 1999) and therefore
considered to be negligible. Other tree nuts, seeds, and legumes were tested
for only two of the sera, and appeared to be negative (Holzhauser and Vieths,
1999; Koppelman et al., 1999), while for the other sera an extensive
investigation of cross-reactivity was not performed (Blais and Phillippe,
2001; Scheibe et al., 2001). One antiserum was not tested for possible cross-
reactivity (Jonsson and Hellenas, 2001).
Recovery of analytes, in this case hazelnut protein, from food matrices is
sometimes difficult because of food processing steps. Heat treatment especially
can denature proteins, thereby decreasing the solubility. This aspect is discussed
in detail elsewhere in this book. For hazelnut detection, the main areas of
application are chocolates and bakery products. Extraction from chocolate
can be particularly difficult due to binding of tannins to proteins, limiting the
protein solubility. Holzhauser and Vieths investigated the recovery of hazelnut
protein from various spiked samples as analyzed with their assay. They
found an average recovery of 106 ± 17%, although for samples spiked with
lower amounts of hazelnut, the recovery varied between 62 and 132%
(Holzhauser and Vieths, 1999). Recoveries of commercial kits are optimized
by adding tannin-binding components such as skimmed milk powder or fish
gelatin (Keck-Gassenmeier et al., 1999). Another aspect that may influence
the quantification of hazelnut in food is roasting. Roasting of hazelnut has
two effects on immunochemical analysis: (i) changing the immunochemical
properties of the target protein, for example by protein denaturation and
Maillard reactions; and (ii) limitation of solubility. It is therefore important
to assess the effect of roasting on the quantitative determination of hazelnuts.
One report shows that dilution curves of raw and roasted hazelnut protein
were almost identical with an estimated maximum difference of about 10%
210 Detecting allergens in food
Almond
For almond, polyclonal antibodies were raised in rabbits, and successfully
applied in immunoblot and competition ELISA (Acosta et al., 1999). The
reactivity was strongly influenced by heat treatment and pH treatment, and
some cross-reactivity with other nuts and legumes was observed (Acosta
et al., 1999). The same group reported an optimized ELISA for almond and
recovery from different matrices, and in addition the effect of heat treatment
was studied (Roux et al., 2001). The detection limit of the method was 5 ppm
in processed foods, and neither blanching, roasting, nor autoclaving markedly
decreased the reactivity (Roux et al., 2001). Meanwhile Hlykwa et al. published
a method to detect almond residues in food as well (Hlywka et al., 2000).
This sandwich ELISA detected down to 1 ppm, and did not cross-react
substantially with other food ingredients, although some reactivity was observed
for sesame seeds, and to a lesser extent for walnuts and macademia, (Hlywka
et al., 2000). The assay was used in a survey of 20 brands of dry breakfast
cereal, and in these products no almond cross-contact was observed.
Detecting tree nuts and seeds in food 211
Brazil nut
Blais et al. (2002) developed an ELISA for the detection of Brazil nut. This
assay was based on egg yolk antibodies (IgY) and reached a sensitivity of
well below 1 ppm. Cross-reactivity with other ingredients was investigated
and appeared to be negligible for peanut, hazelnut, sunflower seed, cucumber
seed, and castor beans (Blais et al., 2002). The antibody was later applied in
a multiplex enzyme immunoassay for the detection of peanut, hazelnut, and
Brazil nut with a sensitivity of around 1 μg protein per gram (1 ppm) (Blais
et al., 2003). The advantage of this test is that important food allergens that
are often used in chocolate manufacturing are assayed simultaneously. Clemente
et al. (2004) developed an inhibition ELISA for Brazil nut 2S albumin, with
a limit of detection of 1 ppm. Cross-reactivity with other nuts and legumes
including peanuts was negligible, and the food products with Brazil nut
declared were judged positive (Clemente et al., 2004).
Walnut
An ELISA for walnut detection has been described by Niemann and Hefle
(2003). The antibodies were raised against a mixture of several varieties of
walnut, both raw and roasted, in sheep and rabbits. Levels of 1 ppm were
detected, and recovery from different food matrices including chocolate was
between 85 and 96% even when low concentrations of walnut were spiked.
Of a set of 50 main food ingredients, only pecan which is a close relative of
walnut cross-reacted significantly (Niemann and Hefle, 2003).
Cashew
Recently, the first assay for cashew nut detection was published. Antibodies
were raised in rabbits and goats against major storage protein. The resulting
antisera were immunoadsorbed with immobilized protein of several nuts,
legumes, and seeds in order to decrease possible cross-reaction (Wei et al.,
2003). Immunoblotting studies demonstrated that the polyclonal sera recognized
proteins similar to those recognized by IgE from cashew nut-allergic patients.
Cross-reactivity of other food ingredients was low, although pistachio and
sunflower seed showed significant cross-reaction. A sensitivity of 0.02 ppm
was reached and recoveries from divers food matrices varied from 64 to
136%, while heating did not affect the reactivity of cashew in the test (Wei
et al., 2003).
212 Detecting allergens in food
2
1.8
1.6
1.4
1.2
A 490
1
0.8
0.6
0.4
0.2
0
0.001 0.01 0.1 1 10 100
Mustard protein (μg/ml)
Fig. 12.2 Calibration curve of mustard seed protein in an inhibition ELISA for
mustard using rabbit polyclonal antibodies. The line represents serial dilutions of
mustard seed protein.
Sesame
Brett et al. (1998) purified the 13S protein fraction of sesame seeds and
raised polyclonal antibodies in rabbits towards it. IgG-immunoblots indicated
that the serum was specific although the authors did not provide a complete
list of tested food ingredients. Using ELISA, detection of sesame was possible
(no detection limit described) (Brett et al., 1998).
Mustard
EU legislation on labeling of food allergens also includes mustard and products
thereof (EU, 2003). Up to now, no methods are either commercially available
or published. Recent work from our group led to a polyclonal antiserum for
mustard seed proteins, and an inhibition ELISA was constructed. The test
performance has been sorted out for some food applications, and Fig. 12.2
shows a calibration curve for mustard seed protein. Mustard seed protein at
a concentration of around 50 ng/ml results in significant inhibition. Taking
into account the necessary dilution steps, the sensitivity of this ELISA is
around 1–2 ppm.
Table 12.4 gives an overview of the published methods for nuts and seeds.
12.7 Conclusions
For hazelnut detection a large number of (ELISA) tests has been described,
and there are test kits commercially available. For other nuts and seeds, there
remains a lot of work to do. Some tests are already commercially available
and used by the food industry (Table 12.5). The assays described up to now
have detection limits around 1 ppm. This is suitable for allergen detection, as
clinical case reports and threshold studies indicate that allergen levels of
Table 12.4 Detection methods for nuts and seeds other than hazelnut
Almond protein Inhibition ELISA Polyclonal rabbit serum 5 ppm Acosta et al., 1999;
Roux et al., 2001
Almond protein Sandwich ELISA Polyclonal rabbit and 1 ppm Hlywka et al., 2000
sheep serum
Brazil nut protein Sandwich ELISA Egg yolk IgY < 0.1 ppm Blais et al., 2002
Walnut protein Sandwich ELISA Polyclonal rabbit and 1 ppm Niemann and Hefle, 2003
sheep serum
Cashew storage protein Sandwich ELISA Polyclonal rabbit and < 0.1 ppm Wei et al., 2003
goat serum
Sesame 13S protein Inhibition ELISA Polyclonal rabbit serum Not specified Brett et al., 1998
214 Detecting allergens in food
Table 12.5 Commercially available detection methods for nut and seed allergens1
10 ppm are relevant (discussed in Chapter 1). A major problem with test
development for allergen detection from nuts and seeds is that nuts and seeds
are closely related phylogenetically. Therefore, raising antibodies that are
specific is a challenge. On the other hand, products tested for certain analytes,
for example traces of hazelnut, should in most cases also be free of other
nuts. In the case that nut speciation is desired, PCR techniques may be
helpful as they are usually more specific compared to immunochemical
techniques.
Commercially available ELISA kits have been improved with respect to
performance. Most optimized tests use three incubation steps of 10 minutes
only (Immer et al., 2004). Compared to sample preparation which may take
considerable time (milling, mixing, heating, and centrifugation) the test itself
is no longer the rate-limiting step. We foresee a future for ELISA-based
methods in the detection of nut and seed allergenic residues. With new
polyclonal antibodies produced and characterized by several groups around
the world, the availability of new kits will increase. It is, however, difficult
to predict which nuts and seeds will be considered the most important allergens
in the future as clinical reactions for a wide variety of nuts and seeds have
been described. For the nuts and seeds that are common food ingredients in
Western diets, we believe that a complete set of ELISA-based kits will be
available in the coming years.
12.8 References
Acosta, M R, Roux, K H, Teuber, S S and Sathe, S K (1999) ‘Production and characterization
of rabbit polyclonal antibodies to almond (Prunus dulcis L.) major storage protein’, J
Agric Food Chem, 47, 4053–4059.
Akkerdaas, J, Schocker, F, Alcocer, M J, Schilte, P, Knulst, A C, Hefle, S L, Aalberse, R
and van Ree, R V (2002a) ‘Detection and immunological characterization of hazelnut
2S albumin’, J Allergy Clin Immunol, 109, s328.
Akkerdaas, J, Wensing, M, Knulst, A C, Hefle, S L, Aalberse, R and van Ree, R (2002b)
‘Sensitive and specific measurement of crude and processed hazelnut protein by a
sandwich-type ELISA’, J Allergy Clin Immunol, 109, s302.
Ampe, C, Van Damme, J, de Castro, L A, Sampaio, M J, Van Montagu, M and
Detecting tree nuts and seeds in food 215
Hefle, S L and Taylor, S L (2002) ‘How much food is too much? Threshold doses for
allergenic foods’, Curr Allergy Asthma Rep, 2, 63–66.
Hlywka, J J, Hefle, S L and Taylor, S L (2000) ‘A sandwich enzyme-linked immunosorbent
assay for the detection of almonds in foods’, J. Food Prot 63, 252–257.
Holzhauser, T and Vieths, S (1999) ‘Quantitative sandwich ELISA for determination of
traces of hazelnut (Corylus avellana) protein in complex food matrixes’, J Agric Food
Chem, 47, 4209–4218.
Holzhauser, T, Wangorsch, A and Vieths, S (2000) ‘Polymerase chain reaction (PCR) for
detection of potentially allergenic hazelnut residues in complex food matrixes’, Eur
Food Res Technol, 211, 360–365.
Hourihane, J O, Bedwani, S J, Dean, T P and Warner, J O (1997) ‘Randomised, double
blind, crossover challenge study of allergenicity of peanut oils in subjects allergic to
peanuts’, Br Med J, 314, 1084–1088.
Hu, F B and Stampfer, M J (1999) ‘Nut consumption and risk of coronary heart disease:
a review of epidemiologic evidence’, Curr Atheroscler Rep 1, 204–209.
Immer, U, Reck, B, Lindeke, S and Koppelman, S (2004) ‘RIDASCREEN ® FAST
PEANUT, a rapid and safe tool to determine peanut contamination in food’, Intl J
Food Sci Tech, 39, 869–871.
Jonsson, H and Hellenas, K E (2001) ‘Optimizing assay conditions in the detection of
food allergens with Biacore’s SPR technology’, Biacore J, 2, 16–18.
Kanny, G, de Hauteclocque, C and Moneret-Vautrin, D A (1996) ‘Sesame seed and
sesame seed oil contain masked allergens of growing importance’, Allergy, 51, 952–
957.
Keck-Gassenmeier, B, Benet, S, Rosa, C and Hischenhuber, C (1999) ‘Determination of
peanut traces in food by a commercially-available ELISA test’, Food Agric Immunol,
11, 243–250.
Klein, E and Guenther, H O (1985) [Determination of hazelnut proteins by
electroimmunodiffusion. II. Dependence of protein yield on the degree of roasting.]
‘Bestimmung von Haselnussprotein (Corylin) mit Hilfe der Elektroimmundiffusion
nach Laurell. II. Abhaengigkeit der Proteinausbeute vom Roestgrad der Produkte’, Z
Lebens Unters Forsch, 180, 36–40.
Klein, E, Baudner, S and Guenther, H O (1985) ‘[Determination of hazelnut proteins by
electroimmunodiffusion. I. Optimized methodology.] ‘Immunchemische Bestimmung
des Haselnussproteins mit Hilfe der Elektroimmundiffusion nach Laurell. I. Mitteilung
der optimierten Methode’, Z Lebens Unters Forsch, 180, 30–35.
Kolopp-Sarda, M N, Moneret-Vautrin, D A, Gobert, B, Kanny, G, Brodschii, M, Bene, M
C and Faure, G C (1997) ‘Specific humoral immune responses in 12 cases of food
sensitization to sesame seed’, Clin Exp Allergy, 27, 1285–1291.
Koppelman, S J, Knulst, A C, Koers, W J, Penninks, A H, Peppelman, H, Vlooswijk, R,
Pigmans, I, van Duijn, G and Hessing, M (1999) ‘Comparison of different
immunochemical methods for the detection and quantification of hazelnut proteins in
food products’, J Immunol Methods, 229, 107–120.
Laurell, C B (1972) ‘Electroimmuno assay’, Scand J Clin Lab Invest Suppl, 124, 21–37.
Lezaun, A, Fraj, J, Colas, C, Duce, F, Dominguez, M A, Cuevas, M and Eiras, P (1998)
‘Anaphylaxis from linseed’, Allergy, 53, 105–106.
Malanin, K, Lundberg, M and Johansson, S G (1995) ‘Anaphylactic reaction caused by
neoallergens in heated pecan nut’, Allergy, 50, 988–991.
Malmheden Yman, I, Eriksson, A, Everitt, G and Yman, L (1994) ‘Analysis of food
proteins for verification of contamination or mislabelling’, Food Agric Immunol, 6,
167–172.
Muller, U, Luttkopf, D, Hoffman, A, Petersen, A, Becker, W M, Schocker, F, Niggeman,
B, Altmann, F, Kolarich, D, Haustein, D and Vieths, S (2000) ‘Allergens in raw and
roasted hazelnuts (Corylus avellana) and their cross-reactivity to pollen’, Eur Food
Res Technol, 212, 2–12.
Detecting tree nuts and seeds in food 217
Niemann, L and Hefle, S L (2003) ‘Validated ELISA for detection of undeclared walnut
residues in food’, J Allergy Clin Immunol, 109, s248.
Nordlee, J A, Taylor, S L, Townsend, J A, Thomas, L A and Bush, R K (1996) ‘Identification
of a Brazil-nut allergen in transgenic soybeans’, N Engl J Med, 334, 688–692.
Ortolani, C, Ballmer-Weber, B K, Hansen, K S, Ispano, M, Wüthrich, B, Bindslev-
Jensen, C, Ansaloni, R, Vannucci, L, Pravettoni, V, Scibilia, J, Poulsen, L K and
Pastorello, E A, (2000) ‘Hazelnut allergy: a double-blind, placebo-controlled food
challenge multicenter study’, J Allergy Clin Immunol, 105, 577–581.
Pastorello, E A, Pompei, C, Pravettoni, V, Brenna, O, Farioli, L, Trambaioli, C and Conti,
A (2001) ‘Lipid transfer proteins and 2S albumins as allergens’, Allergy, 56 (Suppl
67), 45–47.
Roux, K H, Teuber, S S, Robotham, J M and Sathe, S K (2001) ‘Detection and stability
of the major almond allergen in foods’, J Agric Food Chem 49, 2131–2136.
Roux, K H, Teuber, S S and Sathe, S K (2003) ‘Tree nut allergens’, Int Arch Allergy
Immunol, 131, 234–244.
Scheibe, B, Weiss, W, Rueff, F, Przybilla, B and Gorg, A (2001) ‘Detection of trace
amounts of hidden allergens: hazelnut and almond proteins in chocolate’, J Chromatogr
B Biomed Sci Appl, 756, 229–237.
Schocker, F, Luttkopf, D, Muller, U, Thomas, P, Vieths, S and Becker, W M (2000) ‘IgE
binding to unique hazelnut allergens: Identification of non pollen-related and heat-
stable hazelnut allergens eliciting severe allergic reactions’, Eur J Nutr, 39, 172–180.
Sicherer, S H, Munoz-Furlong, A, Burks, A W and Sampson, H A (1999) ‘Prevalence of
peanut and tree nut allergy in the US determined by a random digit dial telephone
survey’, J Allergy Clin Immunol, 103, 559–562.
Sporik, R and Hill, D (1996) ‘Allergy to peanut, nuts, and sesame seed in Australian
children [letter]’, Br Med J, 313, 1477–1478.
Sten, E, Stahl, S P, Andersen, S B, Torp, A M, Olesen, A, Bindslev-Jensen, U, Poulsen,
L K and Bindslev-Jensen, C (2002) ‘Allergenic components of a novel food, Micronesian
nut Nangai (Canarium indicum), shows IgE cross-reactivity in pollen allergic patients’,
Allergy, 57, 398–404.
Stephan, O, Moller, N, Lehmann, S, Holzhauser, T and Vieths, S (2002) ‘Development
and validation of two dipstick type immuno assays for the determination of trace
amounts of peanut and hazelnut in processed food’, Eur Food Res Technol, 215, 431–
436.
Tariq, S M, Stevens, M, Matthews, S, Ridout, S, Twiselton, R and Hide, D W (1996)
‘Cohort study of peanut and tree nut sensitisation by age of 4 years’, Br Med J, 313,
514–517.
Taylor, S L and Nordlee, J A (1996) ‘Detection of food allergens’, Food Technol, 50, 231–
238.
Taylor, S L, Hefle, S L, Bindslev-Jensen, C, Bock, S A, Burks, Jr, A W, Christie, L, Hill,
D J, Host, A, Hourihane, J O, Lack, G, Metcalfe, D D, Moneret-Vautrin, D A, Vadas,
P A, Rance, F, Skrypec, D J, Trautman, T A, Yman, I M and Zeiger, R S (2002)
‘Factors affecting the determination of threshold doses for allergenic foods: how
much is too much?’ J Allergy Clin Immunol, 109, 24–30.
Taylor, S L, Hefle, S L, Bindslev-Jensen, C, Atkins, F M, Andre, C, Bruijnzeel-Koomen,
C, Burks, A W, Bush, R K, Ebisawa, M, Eigenmann, P A, Host, A, Hourihane, J O,
Isolauri, E, Hill, D J, Knulst, A, Lack, G, Sampson, H A, Moneret-Vautrin, D A,
Rance, F, Vadas, P A, Yunginger, J W, Zeiger, R S, Salminen, J W, Madsen, C and
Abbott, P. (2004) ‘A consensus protocol for the determination of the threshold doses
for allergenic foods: how much is too much?’ Clin Exp Allergy, 34, 689–695.
Teuber, S S, Brown, R L and Haapanen, L A (1997) ‘Allergenicity of gourmet nut oils
processed by different methods’, J Allergy Clin Immunol, 99, 502–507.
Teuber, S S, Dandekar, A M, Peterson, W R and Sellers, C L (1998) ‘Cloning and
sequencing of a gene encoding a 2S albumin seed storage protein precursor from
218 Detecting allergens in food
English walnut (Juglans regia), a major food allergen’, J Allergy Clin Immunol, 101,
807–814.
van Ree, R (2002) ‘Carbohydrate epitopes and their relevance for the diagnosis and
treatment of allergic diseases’, Int Arch Allergy Immunol, 129, 189–197.
Wei, Y, Sathe, S K, Teuber, S S and Roux, K H (2003) ‘A sensitive sandwich ELISA for
the detection of trace amounts of cashew (Anacardium occidentale L.) nut in foods’,
J Agric Food Chem, 51, 3215–3221.
Wensing, M, Koppelman, S J, Penninks, A H, Bruijnzeel-Koomen, C A F M and Knulst,
A C (2001a) ‘Hidden hazelnut is a threat to allergic patients’, Allergy, 56, 191–192.
Wensing, M, Penninks, A H, Hefle, S L, Akkerdaas, J, Ree, R V, Koppelman, S J,
Bruijnzeel-Koomen, C A F M and Knulst, A C (2001b) ‘Determination of threshold
levels of patients with hazelnut allergy using double blind placebo controlled food
challenges (DBPCFC’s) 8th International Symposium on Immunological and Clinical
Problems of Food Allergy, Venice, March 11–13, 63.
Wensing, M, Penninks, A H, Hefle, S L, Akkerdaas, J H, van Ree, R, Koppelman, S J,
Bruijnzeel-Koomen, C A and Knulst, A C (2002) ‘The range of minimum provoking
doses in hazelnut-allergic patients as determined by double-blind, placebo-controlled
food challenges’, Clin Exp Allergy, 32, 1757–1762.
Wigotzki, M, Steinhart, H and Paschke, A (2000) ‘Influence of varieties, storage and heat
treatment on IgE-binding proteins in hazelnuts’, (Corylus avellana), Food Agric Immunol,
12, 217–229.
Yunginger, J W, Sweeney, K G, Sturner, W Q, Giannandrea, L A, Teigland, J D, Bray, M,
Benson, P A, York, J A, Biedrzycki, L and Squillace, D L (1988) ‘Fatal food-induced
anaphylaxis’, JAMA, 260, 1450–1452.
13
13.1 Introduction
The major sources of food allergens having animal origin are milk, egg
(especially egg white), crustaceans and fish. Allergies to meat are less frequent.
Crustaceans and fishes are less often used as processed ingredients, except in
surimi, flavors and spices using shrimp powders. They are generally consumed
as clearly recognizable and labelled ingredients. Moreover, they are usually
eaten later in life. In contrast, milk and egg are widely consumed as basic
food products, as milk is the first food for babies. They constitute a good
source of animal protein for a modest financial cost. They are also traditionally
used around the world as food ingredients due to their useful technological
properties such as foaming, jellification, thickening, emulsifying and binding.
They are used in bakery and cakes, delicatessen and processed meat products,
soups, dressings and sauces, among other applications.
Food allergies to cows’ milk and hen’s egg white are frequent in man, and
they are the two prevalent food allergies in children. Symptoms of egg and
milk allergy are identical to those for other food allergies (see Chapter 1).
Many studies have been published concerning the prevalence of egg and
milk allergies; results may differ depending on the selection (food-allergic
patients or unselected population) or on the age of the population studied,
but they show that egg and milk allergies are the most prevalent among food
allergy. In studies on unselected (general) populations, milk allergy could
represent about 2.0% in Australia (Hill et al., 1997, 1999), 1.9% in Finland
(Saarinen et al., 2000), 3.2% in France (Kanny et al., 2001), 3.9% in Germany
(Schäfer et al., 1999), 4.0% in Japan (Iikura et al., 1999) and 1.4% in the US
(Rhim and McMorris, 2001) and egg allergy could represent about 3.2% in
220 Detecting allergens in food
Australia (Hill et al., 1999) and 2.8% in Germany (Schäfer et al., 1999).
Allergic reactions due to the presence of egg white proteins in vaccines were
also shown by Davies and Pepys (1976). Pichler and Campi (1992) reported
an allergy case due to the presence of lysozyme contained in vaginal
suppositories.
The amount of allergenic food that can provoke allergic reactions is not
well known. There is variability in patient sensitivity and in allergen specificity.
Food ingredients can be enriched with allergens due to the production methods
or processing. Heat treatment may decrease or increase the allergenic potential
of food allergens. For all these reasons, according to studies performed by
clinicians, it is considered that detection techniques for allergenic sources
should be around 1–10 ppm, i.e. 1–10 mg/kg (Moneret-Vautrin et al., 2003;
Morisset et al., 2003; Osterballe and Bindslev-Jensen, 2003). Most commercial
methods have detection limits encompassing this range.
Taken together, these reasons indicate the need for quick, sensitive,
economical and available detection methods for milk and egg. Analyses
using these methods are carried out in order to ensure the fairness of commercial
transactions and the protection of food-allergic consumers.
13.2 Milk
13.2.1 Cows’ milk allergy
Cows’ milk allergy (CMA) is a common disease of infancy and early childhood,
with a prevalence of 8% reported in a population of 1121 people with food
allergies (Kanny et al., 2001). CMA affects 2.5% of children under two
years of age, but about 80% become clinically tolerant within the first three
years of life (Høst, 1997). Symptoms of CMA are the usual symptoms of
food-allergic reactions. They are mainly urticaria (76%), atopic dermatitis
(28%), vomiting (9%) appearing in less than two hours (68%) at cumulative
reactive doses below 20 mL (59%) and between 20 mL and 200 mL (32%)
(Saarinen et al., 2000).
More recent studies have tried to evaluate the threshold doses of clinical
reactivity to milk allergens. Clinical allergists have published existing data
on threshold doses for several food allergens such as milk, egg, peanut, fish
and mustard (Hefle and Taylor, 2002; Taylor et al., 2002; and Taylor et al.,
2004). For milk, the lowest amount which provoked symptoms by double-
blind placebo-controlled, single-blind or open challenge ranged from 0.02–
5 mL corresponding, respectively, to 0.6–180 mg of protein. Morisset et al.
(2003) presented results of 59 positive oral challenges to milk. A low threshold
inferior or equal to 0.3 mL of milk was observed in 1.7% of milk allergies
and a cumulative reactive dose inferior or equal to 0.8 mL of milk characterized
5% of milk allergies. The lowest reactive dose found was 0.1 mL of milk.
Sicherer et al. (1999) reported that 25% of milk allergics reacted to doses of
100 mg.
Detecting dairy and egg residues in food 221
Whey 20%
α-lactalbumin Bos d 4 5% 14.2 4.8 P00711 4 disulfide bonds Glycosyl
glycosylated hydrolase
β-lactoglobulin Bos d 5 10% 18.3 5.3 P02754 2 disulfide bonds, Lipocalin
dimer, not Retinol
present in binding
human milk protein
(RBP)
Bovine serum Bos d 6 1% 67 4.9–5.1 P02769 9 disulfide bonds Plasma
albumin protein
Immunoglobulins Bos d 7 3% 160 6–8
Lactoferrin < 1% 80 8.7 P24627 Iron
transport
Caseins Bos d 8 80%
αS1-casein 32% 23.6 4.9–5 P02662 Phosphorylated Calcium
transport
αS2-casein 10% 25.2 5.2–5.4 P02663 Phosphorylated +
1 disulfide bond
β-casein 28% 24.0 5.1–5.4 P02666 Phosphorylated
and glycosylated
κ-casein 10% 19.0 5.4–5.6 P02668 Phosphorylated + Miscelles
1 disulfide bond stability
γ-casein
Detecting dairy and egg residues in food 223
sensitized to cows’ milk proteins. However, cows’ milk proteins have been
demonstrated to be present in breast milk (Axelsson et al., 1986). An increasing
population of infants receives hydrolyzed cows’ milk derived formulae for
the prevention and treatment of CMA. However, for primary prevention in
non-sensitized infants and protection from CMA, partially hydrolyzed
hypoallergenic formulae can be recommended. These formulae have reduced
allergenic activity (Restani et al., 1996).
Evaluation of the residual antigenicity and allergenicity of cows’ milk
substitutes was performed using several in vitro tests (Docena et al., 2002).
Immunoenzymatic methods such as enzyme-linked immunosorbent assay
(ELISA) using anti-cows’ milk protein antibodies were used to detect the
presence of residual antigenic epitopes. Direct EAST (enzyme allergosorbent
test) enables the detection of a potential decrease of IgE-binding to peptides
from moderately and extensively hydrolyzed milk preparations. SDS-PAGE
followed by IgE-immunoblotting using sera from CMA patients or cows’
milk protein-specific IgG antibodies showed that extensively hydrolyzed
preparations do not contain residual components from cows’ milk proteins.
While extensively hydrolyzed milk substitutes produced from whey or caseins
were reported to be safe for some CMA consumers (Halken et al., 1993;
Martin-Esteban et al., 1998), adverse reactions were also reported (Ragno et
al., 1993; Nilsson et al., 1999; Sotto et al., 1999; Carroccio et al., 2000). In
these cases of extreme sensitivity, an elemental amino acid formula should
be used.
milk samples (Axelsson et al., 1986). On the other hand, Restani et al.
(2000) did not detect any β-lactoglobulin or casein in human milk from
breast-feeding mothers using SDS-PAGE, immunoblotting, and β-lactoglobulin
or caseins-specific monoclonal antibodies. However, monoclonal antibodies
might not be able to detect the proteins due to proteolytic changes.
Using RAST inhibition, α-lactalbumin was found to contaminate food-
grade lactose at levels of 1–5 ppm (Frémont et al., 1996). A 30 year old
woman with CMA and without fish allergy experienced anaphylaxis after
ingestion of reconstituted salmon containing undeclared casein, which was
used as a gelation agent combined with microbial transglutaminase, which
serves to cross-link casein to meat proteins for restructuring (Koppelman et
al., 1999).
Cows’ milk proteins can be found in other types of products. Ylitalo et al.
(1999) detected the presence of cows’ milk casein in 13 of 30 commercial
natural latex glove brands by rocket immunoelectrophoresis (RIE) and RAST
inhibition with casein-specific IgE from pooled human sera. Caseins can
also be an aero-allergen: a case of occupational asthma and rhinoconjunctivitis
has been described in a chocolate candy worker in contact with dried cows’
milk. α-lactalbumin was implicated as the cause of the sensitization (Bernaola
et al., 1994). Mäkinen-Kiljunen and Mussalo-Rauhamaa (2002) showed the
presence of caseins in 90 out of 91 house dust samples. The median casein
level was 30 μg/g (range 3–3300 μg/g), which was higher than those of pets
or mites (0.11 μg/g of Fel d 1 and < 0.01 μg/g of Der p 1 or Der f 1). The
authors explain these findings by the use of casein in indoor plasters to
improve their handling properties in practice since the 1960s. Some paints
can also contain casein proteins. In one case, inhalation of milk proteins was
reported to cause death (Bosetti et al., 1997).
13.2.6 Cross-reactions
Cross-reactions between ovine (sheep or goat) and bovine milk have been
studied by several authors (Dean et al., 1993; Sabbah et al., 1997; Bellioni-
Businco et al., 1999; Restani et al., 1999). People allergic to cows’ milk
proteins will often have serum IgE to goat or sheep’s milk proteins (Besler
et al., 2002b,c). β-lactoglobulin and caseins have been described as the main
cross-reacting allergens. Comparison between milk from the different ruminant
species reveals high homologies in both protein composition and structure,
particularly for the four caseins (αS1, αS2, β and κ) that are found in cows’,
sheep’s and goat’s milk. Moreover, important sequence homologies are
observed. Using CMA patient sera by ELISA with purified caseins, Bernard
et al. (1999) showed that important levels of cross-reactivity occur between
bovine, caprine and ovine caseins.
In a study on cows’ milk and human α-lactalbumin, proteins showing a
high degree of sequence homology demonstrated IgE cross-reactivity by
ELISA inhibition (Maynard et al., 1999). The authors suggested low clinical
Detecting dairy and egg residues in food 225
13.3 Egg
13.3.1 Egg allergy
The prevalence of egg allergy, as studied by skin prick tests, was reported to
be about 3.2% in a group of 620 Australian children at risk of atopy (Hill et
al., 1999), 53% by labial food challenge in a population of 142 food-allergic
children (Rancé and Dutau, 1997), and 0.4% in an unselected population of
502 adults (Gislason et al., 1999). Cumulative reactive doses were reported
as ranging from 0.13 mg (raw whole egg) to 200 mg of proteins (dried whole
egg) (Taylor et al., 2002). A recent report (Morisset et al., 2003) showed that
a cumulative reactive dose inferior or equal to 65 mg was observed in 16%
of egg allergic patients with 125 oral challenges performed. The lowest
reactive dose was observed at less than 2 mg of egg. The conclusion of the
study was that the detection tests for egg should ensure a sensitivity of at
least 10 ppm taken from minimal quantities as a 95% guarantee for the
safety of patients allergic to egg on the basis of consumption of 100 g of
food.
Egg white
Ovomucoid Gal d 1 11 28 4.1 P01005 glycoprotein trypsin (serine
protease)
inhibitor
Ovalbumin Gal d 2 54 43 4.5 P01012 phosphoglyco- serpin
protein
Ovotransferrin Gal d 3 12 77.7 6.0 P02789 glycoprotein (15 iron
disulfide bonds) transport
Lysozyme Gal d 4 3.4 14.3 10.7 P00698 (4 disulfide bonds) glycosidase
bacteriolytic
function
Ovomucin 1.5 0.2–8 106
Egg yolk
α-livetin Gal d 5 67 P19121 glycoprotein (17 serum
disulfide bonds) albumin
Detecting dairy and egg residues in food 227
1 2 3 4 1 2 3 4 1 2 3 4 1 2 3 4 1 2 3 4
kDa
148
Lf
BSA 60
42
Cas
30
22
β-Lg
17
α-Lac
A B C D E
Fig. 13.1 SDS-PAGE of milk products (Leduc et al., 1997). A: silver staining; B:
antigens revealed with rabbit antimilk antiseruml C: antigens revealed with rabbit
antiwhey antiserum; D: allergen revealed with patient’s serum H[15] × M; E: allergen
revealed with patient’s serum H[17] × M; 1: proteic binder milk by-product; 2:
skimmed milk powder; 3: whey; 4: sodium caseinate; Lf: lactoferrin; BSA: bovine
serum albumin; Cas: caseins; β-Lg: β-lactoglobulin; α-Lac: α-Lactalbumin.
Detecting dairy and egg residues in food 231
whey proteins, and that the whey fractions contain caseins, as also previously
observed by Janssen et al. (1987). The use of two milk-allergic patient sera
showed that their IgE recognized allergens in the four milk ingredients studied.
For these two sera, major allergens were caseins and, to a lesser extent, lactoferrin.
2D-electrophoresis
Two-dimensional (2D)-electrophoresis, initially described by O’Farrell (1975)
combines IEF and SDS-PAGE. Holen and Elsayed (1990) used this technique
to characterize egg white allergens and showed that in their population of
egg-allergic patients, ovalbumin, ovomucoid, ovotransferrin and lysozyme
were the major egg white allergens. Brodard et al. (1995) used this method
232 Detecting allergens in food
Paste C Paste E
Paste C Paste E R P S R P S
R P S R P S E3
E3
pH 9
Lys pH 9
AP
Ot Ot
Ova
pH 3
pH 3
(a) (b)
Paste C Paste E
R P S R P S
E3
pH 9
Ova
pH 3
(c)
Fig. 13.2 Immunoblots from IEF separation of meat extracts (Leduc et al., 1999). (a)
egg white antigens revealed with rabbit antiserum; (b) allergens revealed with human
serum H [4] × M; (c) allergens revealed with human serum H [1] × M; paste C:
negative control meat product; paste E: meat product containing egg white; R: raw; P:
pasteurized; S: sterilized; E3: frozen pasteurized egg white; Lys: lysozyme; AP:
alkaline phosphatase; Ot: ovotoransferrin; Ova: ovalbumin; 䉰: sample application.
13.4.6 ELISA
Many ELISA methods have been described for detection of milk proteins
(Gern et al., 1991; Mäkinen-Kiljunen and Palosuo, 1992; Mariarger et al.,
1994; Venien et al., 1997; Hefle and Lambrecht, 2004) and egg white proteins
Detecting dairy and egg residues in food 233
(Breton et al., 1988; Rauch et al., 1990; Turin and Bonomi, 1994;
Demeulemester and Guizard, 1996; Leduc et al., 1999; Yeung et al., 2000;
Hefle et al., 2001; Baumgartner et al., 2002; Immer et al., 2003). Only one
ELISA was reported for yolk proteins (Pressi et al., 1994). The specific
antibodies used in these ELISA methods are IgG; thus, they do not detect the
allergenic epitopes, but the antigenic epitopes. Some commercial kits are
available. A list of suppliers is available in Table 6.1 in Chapter 6. Currently
available ELISA methods are generally based on the detection of caseins
and/or β-lactoglobulin, while commercial egg ELISA kits are usually based
on the detection of ovalbumin and/or ovomucoid.
Limits of detection in processed food products depend on various parameters,
such as fat content, severity of heat processing, muscle origin, state of meat
maturation, etc. Hence, detection limits might be different from one product
to another. From a theoretical point of view, ELISA methods are quantitative
but, for the same reasons as the detection limit, results can be only
semi-quantitative or qualitative unless validated appropriately.
In a collaborative study performed by two independent laboratories (Immer
et al., 2003), 43 food products were assayed using a commercial ELISA kit
for egg white detection (see Table 13.3). Of the 43, 32 had labels which
mentioned ‘egg’, ‘egg white’ or ‘egg yolk’, egg white was detected in 27 of
these 32 products and was not detected in five (505, 512, 515, 539 and 543).
Among the 11 products whose labels did not mention egg, egg white was
detected in one product (528) at 5 ppm. The discrepancy between egg levels
found and the levels expected could be due to a number of reasons: (i) the
products do not contain egg; (ii) they contain less egg than mentioned on the
labels or less than the detection limits of the ELISA kit; (iii) the egg-based
ingredients used or the final products were submitted to high levels of heat
or other processing that led to the denaturation of egg molecules and to the
lack of detection by the commercial kit.
Hefle and Lambrecht (2004) developed an ELISA for casein determination
with a detection limit of less than 0.5 ppm. They analyzed retail non-milk-
containing foods such as chocolates, sorbets, fruit drinks and fruit juices and
found in many of them undeclared casein from 0.5 to more than 10 000 ppm.
They also analyzed products associated with milk-allergic consumer complaints:
casein was detected in all of them (5 500–44 500 ppm).
Detection of bovine milk proteins is also useful in quality control to
differentiate milk from other species. ELISA methods were published to
specifically detect cows’ milk in goats’ milk (Castro et al., 1992) or in ovine
milk (Garcia et al., 1991; Rodriguez et al., 1993) as cows’ milk is cheaper
than ovine and caprine milk.
Table 13.3 Detection of egg white proteins in food products using ELISA (Immer et al.,
2003). ELISA method used was the Ridascreen® Egg Protein kit (R-Biopharm, Darmstadt,
Germany). 501–505: cheese and cheese delicatessens; 506 and 507: rice puddings; 508 and
509: custards; 510 and 511: vegetables purees; 516–520: vacuum-packed meat products; 521–
524: fresh pasta; 525–533: dried pasta; 537–541: biscuits; 542 and 543: breads. *: aberrant
results. A and B are the laboratories where assays were performed. Some of the foods are
typical French foods.
et al., 2001; Hird et al., 2003). As milk contains DNA from epithelial cells
(Lipkin et al., 1993), López-Calleja et al. (2004) described a PCR method to
detect cows’ milk in sheep and goat’s milk. Eggs contain DNA which can be
found in commercial egg white ingredients. DNA methods could therefore
be used for detection of milk and egg. But in practice, they are not adapted
for detection of egg or milk allergens because the presence of chicken meat
and beef, respectively, could give false positive responses.
13.4.8 Biosensors
Surface plasmon resonance (SPR) technology (Biacore™, Biacore AB,
Uppsala, Sweden) is discussed in Chapter 9. Allergen detection by SPR
immunoassay is based on the measurement of total sample binding to
allergen-specific antibodies coupled to the sensor surface. Purified antibodies
raised against ovomucoid (egg) and β-lactoglobulin (milk) were covalently
bound to a gold-dextran chip (Jonsson and Hellenäs, 2001). Calibration
curves established with purified allergens showed a detection limit of
approximately 10 ng/mL. No data was given for detection limits in solid
food matrices. However, non-specific binding can cause false positive results
when allergens are analyzed in untreated samples and total sample binding
is used for quantification (Mohammed et al., 2001).
(Weinberger et al., 2000; Bashir et al., 2001; Lin et al., 2001; Zlatanova and
Mirzabekov, 2001; Talapatra et al., 2002; Moreno-Bondi et al., 2003) but, at
the present time, these are impractical for use in most manufacturing
environments.
Quality assurance (ISO, 1999) has become widely used in laboratories.
Good practice guidelines, acknowledged or normalized methods, collaborative
studies, reference materials and accreditation of analysis by independent
organisations are very useful tools for reliable results.
Development of milk and egg with low allergenic properties may be
useful (Besler and Mine, 1999). However, they present certain disadvantages:
the low allergenic products are quite expensive and do not always protect
from possible reactions. Indeed, low allergenic products, prepared to decrease
the number of certain allergenic proteins/peptides, may fail to protect people
allergic to other proteins/peptides.
13.6 Acknowledgements
The authors thank Dominique Bornhauser, Gilles-Antoine Couëtte (Allerbio),
Laurence Guérin (Allerbio), Cécile Guizard (CTSCCV), Pravina Nallatamby
(CTSCCV), Céline Tattegrain and Anne Weyer (Pasteur Institute, Paris) for
helpful discussions.
13.7 References
Adams, S L, Barnett, D, Walsh, B J, Pearce, R J, Hill, D J and Howden, M E (1991)
‘Human IgE-binding synthetic peptides of bovine beta-lactoglobulin and alpha-
lactalbumin. In vitro cross-reactivity of the allergens’, Immunol Cell Biol, 69, 191–
197.
Anet, J, Back, J F, Baker, R S, Barnett, D, Burley, R W and Howden, M E (1985)
‘Allergens in the white and yolk of hen’s egg. A study of IgE binding by egg proteins’,
Int Arch Allergy Appl Immunol, 77, 364–371.
Añibarro, B, Seoane, F J, Vila, C and Lombardero, M (2000) ‘Allergy to eggs from duck
and goose without sensitization to hen egg proteins’, J Allergy Clin Immunol, 105,
834–836.
Axelsson, I, Jakobsson, I, Lindberg, T and Benediktsson, B (1986) ‘Bovine beta-lactoglobulin
in the human milk. A longitudinal study during the whole lactation period’, Acta
Paediatr Scand, 75, 702–707.
Bashir, R, Gomez, R, Sarikaya, A, Ladisch, M R, Sturgis, J and Robinson, J P (2001)
‘Adsorption of avidin on microfabricated surfaces for protein biochip applications’,
Biotechnol Bioeng, 73, 324–328.
Baumgartner, S, Steiner, I, Kloiber, S, Hirmann, D, Krska, R and Yeung, J (2002) ‘Towards
the development of a dipstick immunoassay for the detection of trace amounts of egg
proteins in food’, Eur Food Res Technol, 214, 168–170.
Becker, W M (1989) ‘Reactivities of immunoglobulin E and immunoglobulin G subclasses
identified by isoelectric focusing-immunoprint in allergic patients’, Electrophoresis,
10, 633–639.
Detecting dairy and egg residues in food 237
Davies, R and Pepys, J (1976) ‘Egg allergy, influenza vaccine, and immunoglobulin E
antibody’, J Allergy Clin Immunol, 57, 373–383.
De Blay, F, Hoyet, C, Candolfi, E, Thierry, R and Pauli, G (1994) ‘Identification of alpha
livetin as a cross reacting allergen in a bird-egg syndrome’, Allergy Proc, 15, 77–78.
Dean, T P, Adler, B R, Ruge, F and Warner, J O (1993) ‘In vitro allergenicity of cows’
milk substitutes’, Clin Exp Allergy, 23, 205–210.
Demeulemester, C and Guizard, C (1996) ‘Détection des constituants protéiques par
ELISA (Enzyme-Linked ImmunoSorbent Assay)’, Bull Liaison CTSCCV, 6, 26–29.
Demeulemester, C, Peltre, G, Laurent, M, Panheleux, D and David, B (1987) ‘Cyanogen
bromide-activated nitrocellulose membranes: a new tool for immunoprint techniques’,
Electrophoresis, 8, 71–73.
Demeulemester, C, Lajon, A, Abramowski, V, Martin, J L and Durand, P (1991) ‘Improved
ELISA and dot-blot methods for the detection of whey proteins in meat products’, J
Sci Food Agric, 56, 325–333.
Demeulemester, C, Leduc, V, Polack, B, Huneau, J F, Le Guern, L, Guizard, C and Peltre,
G (1997) ‘Antigènes et allergènes de lait, de blanc d’œuf, de soja. 2 – Dans les
produits carnés’, Ann Fals Exp Chim, 90, 235–246.
Desvaux, F X, Peltre, G and David, B (1989) ‘Characterization of grass pollen specific
IgE, IgA, IgM classes and IgG subclasses in allergic patients’, Int Arch Allergy Appl
Immunol, 89, 281–287.
Desvaux, F X, David, B and Peltre, G (1990) ‘Multiple successive immunoprinting: a fast
blotting technique of a single agarose isoelectric focusing gel’, Electrophoresis, 11,
37–41.
Djurtoft, R, Pedersen, H S, Aabin, B and Barkholt, V (1991) ‘Studies of food allergens:
soybean and egg proteins’, Adv Exp Med Biol, 289, 281–293.
Docena, G, Rozenfeld, P, Fernandez, R and Fossati, C A (2002) ‘Evaluation of the
residual antigenicity and allergenicity of cow’s milk substitutes by in vitro tests’,
Allergy, 57, 83–91.
Eigenmann, P A (2000) ‘Anaphylactic reactions to raw eggs after negative challenges
with cooked eggs’, J Allergy Clin Immunol, 105, 587–588.
Frémont, S, Kanny, G, Bieber, S, Nicolas, J P and Moneret-Vautrin, D A (1996) ‘Identification
of a masked allergen, alpha-lactalbumin, in baby-food cereal flour guaranteed free of
cow’s milk protein’, Allergy, 51, 749–754.
Frémont, S, Kanny, G, Nicolas, J P and Moneret-Vautrin, D A (1997) ‘Prevalence of
lysozyme sensitization in an egg-allergic population’, Allergy, 52, 224–228.
Garcia, T, Martin, R, Rodriguez, E, Azcona, J I, Sanz, B and Hernandez, P E (1991)
‘Detection of bovine milk in ovine milk by a sandwich enzyme-linked immunosorbent
assay (ELISA)’, J Food Prot, 54, 366–369.
Gern, J E, Yang, E, Evrard, H M and Sampson, H A (1991) ‘Allergic reactions to milk-
contaminated ‘nondairy’ products’, N Engl J Med, 324, 976–979.
Gislason, D, Björnsson, E, Gislason, T, Janson, C, Sjöberg, O, Elfman, L and Boman, G
(1999) ‘Sensitization to airborne and food allergens in Reykjavic (Iceland) and Uppsala
(Sweden) – a comparative study’, Allergy, 54, 1160–1167.
Giovannacci, I, Guizard, C, Carlier, M, Duval, V, Martin, J L and Demeulemester, C
(2004) ‘Species identification of meat products by ELISA’, Int J Food Sci Tech, 39,
863–867.
Gjesing, B, Osterballe, O, Schwartz, B, Wahn, U and Lowenstein, H (1986) ‘Allergen-
specific IgE antibodies against antigenic components in cow milk and milk substitutes’
Allergy, 41, 51–56.
Halken, S, Høst, A, Hansen, L G and Osterballe, O (1993) ‘Safety of a new, ultrafiltrated
whey hydrolysate formula in children with cow milk allergy: a clinical investigation,’
Pediatr Allergy Immunol, 4, 53–59.
Hawkes, R, Niday, E and Gordon, J (1982) ‘A dot-immunobinding assay for monoclonal
and other antibodies’, Anal Biochem, 119, 142–147.
Detecting dairy and egg residues in food 239
Rhim, G S and McMorris, M S (2001) ‘School readiness for children with food allergies’,
Ann Allergy Asthma Immunol, 86, 172–176.
Rodríguez, E, Martín, R, García, T, González, I, Morales, P, Sanz, B and Hernández, P E
(1993) ‘Detection of cow’s milk in ewes’ milk and cheese by a sandwich enzyme-
linked immunosorbent assay ELISA’, J Sci Food Agric, 61, 175–180.
Rugo, E, Wahl, R and Wahn, U (1992) ‘How allergenic are hypoallergenic infant formulae?’
Clin Exp Allergy, 22, 635–639.
Rupa, P and Mine, Y (2003) ‘Immunological comparison of native and recombinant egg
allergen, ovalbumin, expressed in Escherichia coli’, Biotechnol Lett, 25, 1917–1924.
Saarinen, K M, Juntunen-Backman, K, Jarvenpaa, A L, Klemetti, P, Kuitunen, P, Lope, L,
Renlund, M, Siivola, M, Vaarala, O and Savilahti, E (2000) ‘Breast-feeding and the
development of cows’ milk protein allergy’, Adv Exp Med Biol, 478, 121–130.
Sabbah, A, Drouet, M, Lauret, M G, Giffard, G and Minkhar, M (1997) ‘Étude de
réactivité croisée entre le lait de vache (LV) et le lait de chèvre (LC)’, Allergie Immunol
(Paris), 29, 203–214.
Sajdok, J, Rauch, P, Paluska, E and Kas, J (1990) ‘Determination of egg and egg white
content of food products by means of immunochemical assessment of ovalbumin’, J
Sci Food Agric, 53, 253–259.
Schäfer, T, Kramer, U, Dockery, D, Vieluf, D, Behrendt, H and Ring, J (1999) ‘What
makes a child allergic? Analysis of risk factors for allergic sensitization in preschool
children from East and West Germany’, Allergy Asthma Proc, 20, 23–27.
Sicherer, S H, Munoz-Furlong, A, Burks, A W and Sampson, H A (1999) ‘Prevalence of
peanut and nut allergy in the US determined by a random digit dial telephone survey’,
J Allergy Clin Immunol, 103, 559–563.
Sotto, D, Tounian, P, Baudon, J J, Pauliat, S, Challier, P, Fontaine, J L and Girardet, J P
(1999) ‘Allergy to cow’s milk protein hydrolysates: apropos of 8 cases’, Arch Pediatr,
6, 1279–1285.
Szepfalusi, Z, Ebner, C, Pandjaitan, R, Orlicek, F, Scheiner, O, Boltz-Nitulescu, G, Kraft,
D and Ebner, H (1994) ‘Egg yolk alpha-livetin (chicken serum albumin) is a cross-
reactive allergen in the bird-egg syndrome’, J Allergy Clin Immunol, 93, 932–942.
Talapatra, A, Rouse, R and Hardiman, G (2002) ‘Protein microarrays: challenges and
promises’, Pharmacogenomics, 3, 527–536.
Taudou, G, Varin-Blank, N, Jouin, H, Marchand, F, Weyer, A and Blank, U (1993)
‘Expression of the alpha chain of human Fc epsilon RI in transfected rat basophilic
leukemia cells: functional activation after sensitization with human mite-specific IgE’,
Int Arch Allergy Immunol, 100, 344–350.
Taylor, S L, Hefle, S L, Bindslev-Jensen, C, Bock, S A, Burks, A W, Jr, Christie, L, Hill,
D J, Høst, A, Hourihane, J O, Lack, G, Metcalfe, D D, Moneret-Vautrin, D A, Vadas,
P A, Rancé, F, Skrypec, D J, Trautman, T A, Yman, I M and Zeiger, R S (2002)
‘Factors affecting the determination of threshold doses for allergenic foods: how
much is too much?’ J Allergy Clin Immunol, 109, 24–30.
Taylor, S L, Hefle, S L, Bindslev-Jensen, C, Atkins, F M, André, C, Bruijnzeel-Koomen,
C, Burks, A W, Bush, R K, Ebisawa, M, Eigenmann, P A, Høst, A, Hourihane, J O,
Isolauri, E, Hill, D J, Knulst, A, Lack, G, Sampson, H A, Moneret-Vautrin, D A,
Rancé, F, Vadas, P A, Yunginger, J W, Zeiger, R S, Salminen, J W, Madsen, C and
Abbott, P (2004) ‘A consensus protocol for the determination of the threshold doses
for allergenic foods: how much is too much?’ Clin Exp Allergy, 34, 689–695.
Turin, L and Bonomi, F (1994) ‘Immunochemical detection of ovalbumin in dairy-based
foods’, J Sci Food Agric, 64, 39–45.
Umpierrez, A, Quirce, S, Maranon, F, Cuesta, J, Garcia-Villamuza, Y, Lahoz, C and
Sastre, J (1999) ‘Allergy to goat and sheep cheese with good tolerance to cow cheese’,
Clin Exp Allergy, 29, 1064–1068.
Urisu, A, Ando, H, Morita, Y, Wada, E, Yasaki, T, Yamada, K, Komada, K, Torii, S, Goto,
M and Wakamatsu, T (1997) ‘Allergenic activity of heated and ovomucoid-depleted
egg white’, J Allergy Clin Immunol, 100, 171–176.
Detecting dairy and egg residues in food 243
14.1 Introduction
Although allergy to wheat is not uncommon, especially the occupational
aspect of it (baker’s asthma), the proteins and peptide motifs involved in
IgE-mediated allergy have not been identified in detail. It is known that a
broad spectrum of wheat proteins is reactive with sera from wheat-allergic
persons. Battais et al. (2003) found, by using radioallergosorbent tests (RAST),
that 60% of sera from their wheat-allergic subjects had IgE and IgG antibodies
against α /β-gliadins and low molecular weight (LMW) glutenin subunits,
55% to γ-gliadins, 48% to ω-gliadins and 26% to high molecular weight
(HMW) glutenins. These results were confirmed with immunoblotting, by
which it was also shown that 67% of patients have IgE antibodies to the
albumin/globulin fraction. Heat processing of wheat dough seems to render
these proteins resistant to breakdown. It has been shown by in vitro digestion
that, while unheated wheat doughs lost their IgE-binding properties during
digestion, the IgE-binding potential was retained in heat-processed dough
(Simonato et al., 2001).
Even although antibodies to whole wheat proteins (including albumins)
are commercially available, most of the research to detect wheat proteins is
directed towards proteins involved in the pathogenesis of quite another
affliction: coeliac disease, a non-IgE-related hypersensitivity in which the
toxic principle is the storage protein (gluten) from some cereal species.
Although this disease is different from IgE-mediated allergy, the eliciting
proteins are shared to a large extent and, as long as these proteins stick
together, testing for one of them will be proof that wheat is present. Because
the prevalence of coeliac disease is increasing tremendously, primarily due
Detecting wheat gluten in food 245
are used. Traditionally, cereal proteins have been classified into four types
according to their solubility (Osborne, 1907):
• albumins – soluble in water or dilute salt solutions;
• globulins – insoluble in water, but soluble in dilute salt concentrations
and insoluble at high salt concentrations;
• prolamins – soluble in aqueous alcohol;
• glutelins – soluble in neither water nor dilute salt solutions but soluble in
dilute acid or alkali.
An alternative classification to that described above has been proposed based
on chemical similarity of reported amino acid sequences of individual
components rather than on differential solubility.
A comparison between these nomenclatures is given by Shewry et al.
(1986). In fact the LMW glutenins can also be considered as aggregated
gliadins, because their composition is comparable to the α-gliadin fraction.
The difference is that they are present in a polymeric form.
The gliadins and glutenins are characterized by a high content of the
amino acids glutamine (Q) and proline (P), ca. 30 and 17 mol%, respectively.
They lack nutritionally important amino acids like tryptophan, lysine and
methionine which makes their nutritional score rather low. They also lack
any enzyme activity and are generally considered as a nitrogen source for the
germinating seed. Gliadins are classified into fractions, according to their
mobility in an electrophoretic field, as α-, β-, γ- and ω-gliadins: the differences
between the first two fractions are small, and they are commonly designated
as α/β. A unique property of ω-gliadin is that it does not contain cysteine and
is not involved in disulfide exchange.
where the proposed limit of 200 mg/kg has been rejected by coeliac
organizations.
Year Number Reference Layout Antibody1 Protein Source Epitope Sensitivity2 Reactivity3 Cross- Extraction Remark
reactivity4
1982 1 Windemann Sandwich pAB A-gliadin Wheat Unknown 1 ng/ml 70% ethanol,
et al., (A-gliadin) 40 °C
1982* 10 ng/ml
(gliadin)
1983 2 Ciclitira pAB α-gliadin Wheat Unknown 1 ng/ml α-gliadin, 5
Year Number Reference Layout Antibody1 Protein Source Epitope Sensitivity2 Reactivity3 Cross- Extraction Remark
reactivity4
1994 10 Ellis et al., pAB/mAB Gliadin Wheat 54 amino- 15 ng/ml Wheat = rye
1994 acid >> barley
peptid >> oats
N-terminal
1995 11 Chirdo Competitive pAB Gliadin Wheat Unknown 1 ng/ml Wheat >> 70% ethanol
et al., 1995 barley > rye after pre-
>> oats extraction
of albumins/
globulins by
0.15 M-NaCl
1995 12 Albrecht Sandwich Gliadin Wheat Unknown 25 ng/ml Maize
and Toth,
1995
1998 13 Ellis et al., Sandwich pAB/mAB Gliadin Wheat A-gliadin 4 ng/ml Wheat >> 40% ethanol
1998 31–49 rye > barley or 50%
= oats propane-1-ol
with 1%
M.E in buffer
1998 14 Chirdo Sandwich mAB Gliadin Wheat Unknown 20 ng/ml 70% ethanol
et al.,1998 after pre-
extraction of
albumins/
globulins by
0.15 M-NaCl
1998 15 Chirdo Competitive mAB Gliadin Wheat Unknown 5 ng/ml
et al., 1998
1998 16 Chirdo Capture mAB Gliadin Wheat Unknown 1 ng/ml
et al., 1998
Table 14.1 Continued
Year Number Reference Layout Antibody1 Protein Source Epitope Sensitivity2 Reactivity3 Cross- Extraction Remark
reactivity4
1998 17 Sorell Sandwich mAB Secalin Rye / Unknown 3 ng/ml Rye > wheat Ethanol 60%
et al., 1998 2mAB wheat = barley >>>
oats
8
2003 18 Valdez Sandwich mAB Secalin Rye QXPW/FP 1,5 ng/ml Barley = rye Ethanol 60%
et al. 2003 = wheat
>>> oats
2003 19 Henterich Direct, mAB Secalin Rye As 18 0,16 ng/ml As 18 Reducing
et al., 2003 signal agent with
amplified guanidine
with PCR hydrochloride
2004 20 Spaenij- Competitive mAB α-gliadin Wheat α-59-71, 12 ng/ml Barley = Ethanol 40% 9
* in German
1
pAB means polyclonal antibody; mAB means monoclonal antibody
2
sensitivity is given in ng/ml. Actual detection level in food is depending on amount of food extracted
3
reactivity: the sensitivity with respect to the various prolamins is stated
4
cross-reactivity: if unspecific, cross-reacting cereal species are stated
5
radioimmunoassay, Staphylococcus aureus used to separate bound and free125 iodinated antigen
6
designed to detect conformation-dependent epitopes in wheat
7
ringtested in AOAC framework
8
alternatively an extraction is carrried out in a reducing environment containing guanidine hydrochloride
9
uses ABs detecting T-cell epitopes
254 Detecting allergens in food
heat stable gliadin fractions or the use of reducing agents and/or chaotropic
agents to reverse the disulfide interactions that take place during dough
kneading and baking.
The latter approach was investigated by Sorell et al. (1998) and García et
al. (2005), and a patent has been applied for this extraction procedure (WO
02/092633). An amount of 0.25 g of a food is extracted with 2.5 ml of a
solvent containing 250 mM 2-mercaptoethanol and 2 M guanidine
hydrochloride in PBS. The extract is kept at 50 °C for 40 minutes and is then
diluted with 7.5 ml 80% ethanol, after which it is vortexed and incubated for
one hour at room temperature in a rotary shaker. After centrifugation, the
extracts are ready to be used. A panel of antibodies was generated against rye
prolamins. Out of this panel, two, R3 and R5, were chosen for the assay in
addition to another, 13B4, which has been described elsewhere (Ellis et al.
1998). A mixture of R5 and 13B4, which have complementary selectivities
(R5 reacting with secalins and hordeins and 13B4 with gliadins), was used
to coat the wells. This multiple coating consequently allowed the binding of
all three cereal species. A third antibody (R3) was conjugated with horseradish
peroxidase and used as a reporter antibody. The assay thus developed was
able to detect gliadins, hordeins and secalins at about the same sensitivity.
The detection level as reported by the authors was 1.5 ng/ml. No combination
of the R-monoclonals was sensitive enough to extend the sensitivity to oats:
a separate method was developed to detect this cereal.
A modification of this assay was described by Valdés et al. (2003). In this
assay (sandwich format) just one monoclonal (the R5) antibody was used for
capturing as well as detection. The molecular recognition pattern of this
antibody has been investigated by Osman et al. (2001) by phage display and
pepscan. With both techniques a pentapeptide consensus sequence was found
(QXPW/FP, resp. QQPFP). This sequence is one of the amino acid motifs
believed to be involved in the pathogenicity of coeliac disease. It is present
in a high number of copies in γ- and ω-gliadin, and in a smaller number in
α-gliadin and LMW glutenin. The presence of an epitope in LMW glutenin
poses problems with respect to correct calibration of the assay because,
when comparing to a gliadin standard, part of the glutenin will be reported
as gliadin. In this assay, however, using the same mAB for capture and
reporting, this might not lead to substantial errors because multiple epitopes
would be needed for detection of glutenin, and LMW glutenin contains only
one. In addition, the number of epitopes on γ- and ω-gliadin outweighs the
number of epitopes on the LMW glutenin. The authors report a sensitivity of
1.56 ng gliadin/ml corresponding to 1.5 mg/kg in food using a low dilution
of 1:25. The reproducibility and repeatability was stated to be 8.7% and the
repeatability 7.7% in a small scale investigation.
A collaborative trial was organized in 2002 by the WGPAT (see Section
14.7), the provisional results of which have been published (Immer et al.,
2003; Immer and Haas-Lauterbach, 2004). Twelve foods were analysed, of
which four were spiked with the reference gliadin developed by the WGPAT
Detecting wheat gluten in food 257
binding substantially, suggesting that the binding was in the region QQQPFP.
This region is present in α- and ω-gliadin. The truncated motif is present in
avenin (QQQPF) and the more truncated motifs, QQQP and QQPFP, are
present in γ-gliadin, hordein and secalins. The reactivity to ω-gliadin is
lower than expected given that it is present in this fraction. The authors
suggest that this might be due to a lower reactivity of the capture antibody to
ω-gliadins.
Wheat starches, gluten-free ingredients and gluten-free products as well
as spiked samples were analyzed. The initial extract was made by mixing
50 mg product with 1 ml 40% ethanol for one hour at room temperature,
after preliminary defatting twice with four volumes in n-butanol. In a separate
experiment, cooked crust and crumb of a loaf prepared from flour spiked
with 1% var. Rektor gliadin (spiked before the stage of dough formation)
and which had been baked at 230 °C for 10 minutes were analyzed. This loaf
was extracted at 60 °C under nitrogen atmosphere for one hour with a reducing
buffer containing 50% propane-1-ol, 1% mercaptoethanol, 0.08 mol/l Tris-
HCl (pH 7.5) and 2 mol/l urea. In this case, the calibration curve was made
up in the reducing buffer diluted 100-fold. Prolamins from coeliac non-toxic
rice, maize, millet and sorghum as well as presumably gluten-free ingredients
of miscellaneous origin did not cross-react in the assay. The assay could
detect gluten in cooked foods, although at reduced sensitivity, the recovery
being about 17%. Use of reducing agents was not successful, presumably
because they denatured the capture antibodies.
This method might offer good prospects as long as non-reducing conditions
are used. The motif QQQPFP is, however, also present in LMW glutenins
(which become solubilized under reducing conditions) and this raises the
question of what should be taken as a reference material: gliadin or whole
gluten. However, at present this issue is not relevant; it appears that the
antibodies do not tolerate the reducing agent mercaptoethanol at a concentration
of 0.01% and the more efficient extraction is traded off by this phenomenon.
A puzzling phenomenon is the difference between the antibody used in
this assay (against QQQPSP) and the R5 used by Sorell et al. (1998) and
Valdés et al. (2003) (against QQPSP) with respect to the difference in reactivity
to γ- and ω-gliadin and cereals like rye and barley. What a difference a Q
makes! A variety of commercially available gluten-free foods were analyzed
and small quantities of gluten were detected in some products.
Of course it would be feasible to design antibody-based assays against
other peptides as well. Fraser et al. (2003) investigated the toxicity of a
sequence containing residues 56–75 of α-gliadin, which proved to be toxic
in vivo. This peptide would consequently also be a candidate to design an
ELISA around. A problem not yet solved is: how many toxic motifs will be
found in the future and how should they be weighed in the final result to get
a realistic representation of toxicity?
Detecting wheat gluten in food 259
Fig. 14.1 Allelic variance between cereal cultivars. (From left to right): lane 1–5
wheat, 6–8 rye, 9–11 barley, 12, 13 wheat gluten reference. SDS-PAGE (gradient gel
and immunochemical staining with a polyclonal anti-wheat gluten antibody).
and 150 ppm gluten). The technique is very efficient at the qualitative
level, i.e. it can be used to confirm the presence of gluten in a gluten-free
product with a high degree of confidence if the band pattern obtained with
the suspected sample matches that of a standard. Due to this cultivar
dependence an exact match is, however, very difficult to achieve (see
Fig. 14.1).
Format
The composition of the food is very relevant to the format of the assay. A
capture ELISA in sandwich format can be very specific, but it may miss a
small size peptide containing just one epitope. Designing antibodies against
specific peptide motifs and using these in competitive format might circumvent
these shortcomings. Quantitation might be impaired anyway in situations
where stoichiometry is disturbed and if there is no prior knowledge about
size of the peptides. Ideally (but hardly feasible), quantitation of toxic peptides
should be performed with a gliadin standard degraded to the same extent and
in the same manner (sites of hydrolytic cleavage and deamidation) as the
sample. Another option would be to include a predigestion of the sample to
level out differences between them and the reference protein. It remains
questionable whether this is feasible. A patent has been applied for using this
approach (Denery-Papini et al., 2001).
Specificity of antibodies
Increasingly, antibodies are mapped on the molecular level. However,
unexpected reactions may occur, dependent on binding on e.g. plastic surfaces
(Chirdo et al., 1995), temperature (Brett et al., 2002) or extraction method.
We have repeatedly found that antibodies which behaved quite specifically
in a sandwich or competitive assay produced unspecific reactions (a positive
response to maize, sorghum, millet and rice) when used in Western blotting.
Apparently cross-reacting protein fractions which are not extracted by alcoholic
solutions were extracted when using SDS buffer and a reducing environment.
Surprisingly, the antibody, which had been prepared with an ethanol-extracted
gliadin as immunogen and which had been absorbed to remove all unwanted
specificities (and which should thus be specific for gliadin), behaved
unspecifically when brought in contact with proteins extracted differently.
14.4.2 Maldi-TOF
A mass-spectrometric method was explored by Camafeita et al. (1997, 1998)
and Camafeita and Méndez (1998). In this method, the proteins of a food are
extracted and measured on a MALDI-TOF mass spectrometry (MS) system.
264 Detecting allergens in food
Assays that would detect the presence or absence of such peptide motifs
would thus be accurate indicators of safety of the food for consumption by
coeliac disease patients. A novel method to detect such gluten peptides in
food has been developed at the Department of Immunohaematology and
Blood Transfusion of the Leiden University Medical Centre (LUMC), Leiden,
The Netherlands (Spaenij-Dekking et al., 2004). Monoclonal antibodies were
generated that are specific for T-cell stimulatory epitopes in α-gliadin, γ-
gliadin or LMW glutenin molecules. These mABs are used in competition
assays in which peptides present in the sample to be measured compete for
binding to the peptide-specific mAB with a biotinylated indicator peptide.
These assays are highly specific and detect up to 50 ng gluten per ml of the
European gliadin standard. This corresponds to detection of 0.5 ppm in
extracted gluten preparations. The advantages of the new method are multifold:
(i) the assay measures specifically the presence of T-cell stimulatory epitopes
known to be involved in the development of CD; (ii) as a consequence of the
format, this type of assay requires only one mAB per peptide, allowing the
detection of small gluten peptides corresponding to the size of T-cell stimulatory
epitopes – this is impossible with the currently available sandwich ELISA
systems; (iii) the assay detects gliadin and glutenin molecules simultaneously,
allowing a more fair judgement of the potential toxicity of the foods tested
for coeliac disease patients. This type of assay offers good possibility to
screen novel cereal sources, like tef (Eragrostis tef), or other cereals with
disputed coeliac toxicity. However, given the almost idiotypic reaction of
coeliacs, an antibody mimicking the reactivity of a panel of T-cells might be
necessary.
Without any doubt, another trend is the development of rapid screening
tests, by which a production lot can be screened rapidly for gluten contamination
without requiring access to laboratory facilities. In verifying the performance
of hazard analysis of critical control point (HACCP) systems an analysis of
the end-product is the ultimate method to check whether all critical points
during production are under control. At the time of writing, dipstick assays
are commercially available from Tepnel (Deeside, UK), R-Biopharm
(Darmstadt, Germany) and Operon (Cuarte de Huerva, Spain) based on the
immunochemical capture of coloured microparticles on a blotting medium;
however, only limited data about their performance in practice is available
yet. Drawbacks for home use include the need to extract the protein from a
(most often) solid food, which might be cumbersome and prone to mistakes;
however, these methods might have good prospects in HACCP programs.
A biochip is being developed in the framework of an EU funded research
project: Quantitation of coeliac disease toxic gluten in foodstuffs using a
chip system with integrated extraction, fluidics and biosensoric detection
(QLK1-CT-2002-02077) http://www.etseq.urv.es/BBG/dinamic/Cd-chef/. This
project, which is coordinated by the Department of Chemical Engineering,
Universitat Rovira i Virgili, Tarragona, Spain, includes the development of
immuno- and apta-sensor generic platforms with optical/electrochemical/
Detecting wheat gluten in food 267
derived from wheat starch) this amendment will be likely to have dramatic
effects on the labelling of many products. Wheat starch-derived products
like glucose syrup may be used as a raw material for the production of many
food ingredients and additives like polyols, ascorbic acid, acetic acid, etc. In
the near future it may thus become mandatory to label food containing these
ingredients as derived from wheat unless an exemption is allowed (wheat-
based glucose syrups and maltodextrins, glucose syrups based on barley and
cereals used in distillates for spirits are (by directive 2005/26/EC) provisionally
excluded from this mandatory labelling, because, most probably their gluten
content is very low). In the US and other countries, newer labeling laws
require, or will soon require, this type of labeling.
Regarding the gliadin standard: the reference material (IRRM-480)
developed on the initiative of the WGPAT is available from the IRRM in
Geel, Belgium. If one is aiming at producing reproducible results, the use of
a worldwide accepted standard is of utmost importance. The IRRM website
is http://www.irmm.jrc.be/.
14.8 References
Albrecht, E and Toth, A (1995) ‘Investigation of gliadin content of wheat flour by ELISA
method’, Acta Alimentaria, 24, 23–29.
Allmann, M, Candrian, U and Lüthy, J (1992) ‘Detection of wheat contamination in
dietary non-wheat products by PCR’, Lancet, 339, 309.
Allmann, M, Candrian, U, Höfelein and Lüthy, J (1993) ‘Polymerase chain reaction
(PCR) a possible alternative to immunochemical methods assuring safety and quality
of food. Detection of wheat contamination in non-wheat products’, Z Lebens Unters
Forsch, 196, 248–251.
AOAC official methods of analysis: supplement (1995) AOAC official method 991.19,
chap 32:13 [32.31.24].
Arentz-Hansen, H, Fleckenstein, B, Molberg, Ø, Scott, H, Koning, F, Jung, G, Roepstorff,
P, Lundin, KEA and Sollid, L M (2004) ‘The molecular basis for oat intolerance in
patients with celiac disease’, Plos Med 1, Epub, Oct 19.
Ayob, M K, Rittenburg, J, Allen, J C and Smith, C J (1988) ‘Development of a rapid
enzyme-linked immunosorbent assay (ELISA) for gliadin determination in food’,
Food Hydrocolloids, 2, 39–49.
Battais, F, Pineau, F, Popineau, Y, Aparicio, C, Kanny, G, Guerin, L, Moneret-Vautrin, D
A and Denery-Papini, S (2003) ‘Food allergy to wheat: identification of immunoglobulin
E and immunoglobulin G-binding proteins with sequential extracts and purified proteins
from wheat flour’, Clin Exp Allergy, 33, 962–70.
Brett, G M, Mills, E N, Bacon, J, Wellner, N, Husain, R D, Tatham, A S, Shewry, P R and
Morgan, M R (2002) ‘Temperature-dependent binding of monoclonal antibodies to C
hordein’, Biochim Biophys Acta, 1594, 17–26.
Camafeita, E and Méndez, E (1998) ‘Screening of gluten avenins in foods by matrix-
assisted laser desorption/ionization time-of-flight mass spectrometry’, J Mass Spectrom,
33, 1023–1028.
Camafeita, E, Alfonso, P, Mothes, T and Méndez, E (1997) ‘Matrix-assisted laser desorption/
ionization time-of-flight mass spectrometric micro-analysis: the first non-immunological
alternative attempt to quantify gluten gliadins in food samples’, J Mass Spectrom, 32,
940–947.
Detecting wheat gluten in food 269
Koning, F (2004) ‘A novel and sensitive method for the detection of T cell stimulatory
epitopes of alpha/beta- and gamma-gliadin’, Gut, 53, 1267–1273.
Stern, M, Ciclitira, P J, Van Eckert, R, Feighery, C, Janssen, F W, Méndez, E, Mothes, T,
Troncone, R and Wieser, H (2001) ‘Analysis and clinical effects of gluten in coeliac
disease’, Eur J Gastroenterol, 13, 741–747.
Szamos, J, Aubrecht, E and Gelencsér, É (1998) ‘Detection of wheat by adapted polymerase
chain reaction (PCR methodology)’, Acta Alimenta, 27, 87–95.
Troncone, R, Vitale, M, Donatiello, A, Farris, E, Rossi, G and Auricchio, S (1986) ‘A
sandwich enzyme immunoassay for wheat gliadin’, J Immunol Methods, 92, 21–23.
Valdes, I, Garcia, E, Llorente, M and Méndez, E (2003) ‘Innovative approach to low-
level gluten determination in foods using a novel sandwich enzyme-linked
immunosorbent assay protocol’, Eur J Gastroenterol Hepatol, May, 15(5), 465–74.
Erratum in: Eur J Gastroenterol Hepatol, Jul, 15, 839.
Van de Wal, Y, Kooy, Y M, Van Veelen, P, Vader, W, August, S A, Drijfhout, J W, Peña,
S A and Koning, F (1999) ‘Glutenin is involved in the gluten-driven mucosal T cell
response’, Eur J Immunol, 29, 3133–3139.
Van Eckert, R (2002) ‘The PWG gliadin, a new reference material’, Proceedings of the
16th Meeting of the Working Group on Prolamin Analysis and Toxicity, M Stern (ed.),
Zwickau, Germany, Verlag Wissenschaftliche Scripten, 25–27.
Van Eckert, R, Scharf, M, Wald, T and Pfannhauser, W (1998) ‘Determination of proteins
with ELISA-Methods: Doubtful quantitative results?’ Proceedings of the 12th Meeting
of the Working Group on Prolamin Analysis and Toxicity, M Stern (ed.), Zwickau,
Germany, Verlag Wissenschaftliche Scripten, 35–40.
Van Eckert, R, Berghofer, E, Ciclitira, P J, Chirdo, F S, Denery-Papini, S, Ellis, H J,
Ferranti, P, Goodwin, P, Immer, U, Mamone, G, Méndez, E, Mothes, T, Novalin, S,
Osman, A, Rumbo, M, Stern, M, Thorell, L, Whim, A and Wieser, H (2005) ‘Towards
a new gliadin reference material – isolation and characterisation’, J Cereal Sci, submitted.
Vestergaard, P (2003) ‘Bone loss associated with gastrointestinal disease: prevalence and
pathogenesis’, Eur J Gastroenterol Hepatol, 15, 851–856.
Weegels, P L, De Groot, A M G, Verhoek, J A and Hamer, R J (1994a) ‘Effects on gluten
of heating at different moisture contents. II Changes in physico-chemical properties
and secundary structure’, J Cereal Sci, 19, 39–47.
Weegels, P L, De Groot, A M G, Verhoek, J A and Hamer, R J (1994b) ‘Effects on gluten
of heating at different moisture contents. I Changes in functional properties’, J Cereal
Sci, 19, 31–38.
Wieser, H (1998) ‘Investigations on the extractability of gluten proteins from wheat
bread in comparison with flour’, Z Lebens Unters Forsch, 207, 128–132.
Wieser, H, Belitz, H-D, Ashkenazi, A and Idar, D (1983) ‘Isolation of coeliac active
peptide fractions from gliadin’, Z Lebens Unters Forsch, 176, 85–94.
Wieser, H, Seilmeier, W and Belitz, H-D (1994) ‘Quantitative Determination of Gliadin
Subgroups from different Wheat cultivars’, J Cereal Sci, 19, 149–155.
Windemann, H, Fritschy, F and Baumgartner, E (1982) ‘Enzyme-linked immunosorbent
assay for wheat a-gliadin and whole gliadin’, Biochim Biophys Acta, 709, 110–121.
15
15.1 Introduction
Detection of peanuts, tree nuts, wheat, milk and egg as members of the ‘Big
Eight’ allergenic foods is described in the preceding chapters. In this chapter
we describe the tests available for the remaining allergens: soy, crustaceans
and fish. Compared to the other food allergens, detection of the present
group of allergens is relatively unexplored, due to several reasons related to
these particular foods. Soy is a food stuff that is used in a vast number of
applications, undergoing a wide variety of processing steps, with consequences
for the detection of allergenic residues. Crustaceans and fish count a large
number of species, and for detection purposes it is necessary to choose
whether to test for one species, for a subset or for the entire group.
Considerations for such choices are based on the allergic patient’s
characteristics: do they react to one of the species, or even one allergen, or
to more species and more allergens? From the food industry perspective, the
issue is to detect fish and/or crustacean residue, regardless of the species and
the nature of the allergens. The sections below will cover the published and
commercially available methods for soy, fish and crustaceans, and will discuss
the advantages and disadvantages of the chosen approaches.
15.2 Soy
Soy is a food ingredient with both technofunctionality and biofunctionality.
The beneficial effects on health of soy isoflavones have been recently reviewed,1
and food products containing 25 g of soy per intake may claim to be healthy
274 Detecting allergens in food
with soy may be useful. Suitable markers have been chosen for their stability
and ease of assay under the processing conditions used. Examples of markers
that have been used for assay development include insoluble polysaccharides,
oligosaccharides, protein-bound sugars, free amino acids, free peptides, phytate,
saponins, sterols and metals.15 Another approach has used histological
techniques. A microscopic examination of calcium oxalate crystals and plant
cell components indicate the presence of soy. Staining for cell wall
polysaccharides could also be useful in this respect.15 The detection levels of
these methods are too high for quantification of traces of soy for allergenic
concerns.
Meyer et al.19 evaluated a DNA-based PCR method for detecting soy
residue in food products, and achieved a detection limit of 70 ppm. More
recent public discussion on genetically modified organisms (GMO) and foods
originating from such crops led to the development of assays for GMO soy,
with a reasonable sensitivity (for GMO concerns; 0.9%, EU legislation, for
more information see http://europa.eu.int/comm/food/fs/gmo/gmo_
ongoinit_en.html). Methods based on DNA, most often polymerase chain
reaction (PCR) tests, are applied to distinguish between wild-type and GMO
soy. As a test control, a gene present in wild-type soy was detected as well.
Van Duijn et al.,20 for example, measured the soy lectin gene as a positive
control. This test has also been used to detect soy in processed food with a
reported detection limit of 0.01% (100 ppm).
The sensitivities of the described indirect methods are between 0.01%
and 1% (100–10 000 pm). These are appropriate for detecting soy added as
a functional ingredient, but not for detecting traces originating from cross-
contamination. Besides this practical disadvantage, there is a theoretical
concern. In attempting to protect soy-allergic consumers, the allergic
component, i.e. protein, should be measured rather than a marker. This is of
key importance because industrial soy processing results in a variety of soy
protein products that may have lost other (marker) components during
processing.
Antibody-based tests
Enzyme-linked immunosorbent assay (ELISA) is a powerful analysis tool
for detection of specific proteins. It has the advantage of simultaneously
testing a larger series of samples at a high level of sensitivity. A large number
of investigators have used ELISA methods for the detection of soy protein in
meat products. Immunochemical methods are generally limited to the qualitative
screening of raw or mildly processed products, since protein denaturation
often alters the antigen–antibody interaction.15 This has been studied by
different authors15,21–32 and the most important results are discussed below.
Antibodies against soy can be raised in laboratory animals, usually rabbits,
mice, goats, sheep or donkeys, as described in Chapter 3. Individual animals
may react differently to immunization protocols, and it is difficult to predict
which species is the best host for raising antibodies against soy. In addition,
Detecting soy, fish and crustaceans in food 277
there is another issue that influences the success of raising antibodies against
soy. Soybean protein is a common substance in animal food, so the antigenic
responses obtained after immunization are not expected to be high. This may
lead to antibodies with low affinity and, consequently, ELISAs constructed
with such antibodies have poor sensitivities. This may be overcome by using
animals kept on soy-free diets.24 Aiming for a sensitive method to detect soy
protein, it seems to be logical to choose glycinin or β-conglycinin as antigenic
protein since these are the major soy proteins and they are abundantly present
in soy. However, using a single-allergen method creates the risk of false
negative test results in the case that the analyte is not present or does not
react while other allergens may still be present.
protein. This was also found to occur at pH 7.6 at different ionic strengths.21,33
The epitope of the antibody used by these researchers corresponds to the
residues 78–84 in the acidic extension present in the α′ subunit of β-
conglycinin,29 and seems to be continuous in nature.31 A linear epitope, in
contrast to a conformational epitope, is expected to be more heat stable and
may become more exposed after denaturation. This may explain the increased
detectability of heated β-conglycinin.
required dilution factor was 20. Recently a method with a high sensitivity
(around 1 ppm) was described.9 This method uses an easy extraction protocol,
a one-step extraction at pH 12. Table 15.1 shows that this extraction has high
recoveries for a variety of different soy ingredients as compared to extraction
under native condition or extraction with urea and reducing agents. A mixture
of soy protein isolate, soy protein concentrate and texturized soy protein was
extracted at pH 12, and the extract used for raising antibodies in goats and
rabbits. A classical protocol for immunization was used and, prior to that, the
animals were kept on a soy-free diet for at least three months. An inhibition
Table 15.1 Extractability of processed soy ingredients using three different methods
2500
2000
1500
mAbs
1000
500
0
0.0001 0.001 0.01 0.1 1 10 100 1000 10 000
Soy flour concentration (μg protein/ml)
Fig. 15.1 Standard curve for soy protein ELISA. (ELISA plates were coated with pH
12-extracted soy and incubated with mixtures of rabbit antibody against pH 12-
extracted soy with calibrator (pH 12-extract of lightly toasted soy flour).
ELISA using rabbit antibodies was constructed and the detection limit is
0.02 μg/ml (Figure 15.1) which corresponds to 0.4 ppm for solid food samples.
Table 15.2 shows that the recovery of this test is high (mean 73%) and that
the variation between individual samples is only moderate (35%). These
characteristics are promising as they allow soy detection with sensitivity
15.3 Crustaceans
Shrimp is the most commonly consumed crustacean shellfish and causes
severe allergic reactions, but other members of the Crustacean family such
as lobster, crab and crawfish can also elicit severe reactions.37,38,39,40 Estimates
of the number of shrimp-allergic individuals range from 0.6–2.8% of the
population. In the US, allergy to crustacean shellfish is more prevalent than
peanut allergy.
282 Detecting allergens in food
15.4 Fish
Fish allergy is relatively common, especially in geographic regions where
fish is an important part of the diet. Fatal reactions have occurred as a result
of allergic reactions to fish.39,52 Numerous studies have attempted to estimate
the prevalence of fish allergy, usually ranging from 0.3–0.5%.38,53,54 Although
the exact prevalence of fish allergy is not well established, it is listed among
the most common of all food allergens.55 Fish can also cause occupational
reactions. While asthma is the most prominent symptom, anaphylaxis has
been reported, and physical contact with seafood can also elicit symptoms in
allergic individuals.56,57
284 Detecting allergens in food
Sal s 1 is described as having two bands, at 12 and 14 kD, and therefore has
at least two isotypic variants. The allergens from other fish species are also
parvalbumins including horse mackerel (Tra j 1) and bigeye tuna (Thu o
1).69–71 While tuna-allergic individuals have been noted to have IgE-binding
to Gad c 1, one study noted that raw tuna extracts seemed to lack IgE-
binding bands in the parvalbumin size range that were present in extracts of
catfish, cod and snapper.65 However, Park et al.72 described IgE-binding to
tuna proteins at 12–13 kD using sera from fish-allergic persons and IgE-
binding bands at 12–13 kD in mackerel, pollock and cod. Also, unique
binding to a band at 19 kD from saury and 37 kD from tuna were observed.
In one study, canned tuna did not present problems for five fish-allergic
children;73 canning may decrease the allergenicity of cod. However, amino
acid sequencing has shown that the 12–13 kD band from tuna does not have
the same sequence as does cod parvalbumin; this may explain why tuna did
not cross-react extensively with other species in this study.
Bernhisel-Broadbent et al.74 found that in 11 subjects demonstrating in
vitro allergy to fish, positive oral challenge only occurred to one species in
seven subjects, to two species in one subject, and to three in two others. This
observation has been also seen in other studies.62,75,76 However, some studies
have found that subjects clinically react to multiple fish.65,77 A recent survey
found that the rate of reactions to multiple fish among those with fish allergy
was 67%.39
15.6 References
1. Fitzpatrick, L A (2003) ‘Soy isoflavones: hope or hype?’ Maturitas, 44, S21–S29.
2. Hoogenkamp, H, Vegetable Protein, 3rd edn, St Louis, MO, Protein Technologies
International.
3. Hourihane, J O, Kilburn, S A, Nordlee, J A, Hefle, S L, Taylor, S L and Warner, J O
(1997) ‘An evaluation of the sensitivity of subjects with peanut allergy to very low
doses of peanut protein: a randomized, double-blind, placebo-controlled food challenge
study.’ J Allergy Clin Immunol, 100, 596–600.
4. Wensing, M, Penninks, A H, Hefle, S L, Koppelman, S J, Bruijnzeel-Koomen, C A
Detecting soy, fish and crustaceans in food 287
43. Sampson, H A, Mendelson, L and Rosen, J P (1992) ‘Fatal and near-fatal anaphylactic
reactions to food in children and adolescents.’ N Engl J Med, 327, 380–384.
44. Cartier, A, Malo, J.-L, Ghezzo, H, McCants, M and Lehrer, S (1986) ‘IgE sensitization
in snow crab-processing workers.’ J Allergy Clin Immunol, 78, 344–348.
45. Malo, J-L and Cartier A. ‘Occupational reactions in the seafood industry.’ Clin Rev
Allergy, 11, 223–240.
46. Desjardins, A, Malo, J L, L’Archeveque, J, Cartier, A, McCants, M and Lehrer, S B
(1995) ‘Occupational IgE-mediated sensitization and asthma caused by clam and
shrimp.’ J Allergy Clin Immunol 96, 608–617.
47. Goetz, D W and Whisman, B A (2000) ‘Occupational asthma in a seafood restaurant
worker: cross-reactivity of shrimp and scallops.’ Ann Allergy Asthma Immunol, 85,
461–466.
48. Jeoung, B J, Reese, G, Hauck, P, Oliver, J B, Daul, C B and Lehrer, S B (1997)
‘Quantification of the major brown shrimp allergen Pen a 1 (tropomyosin) by a
monoclonal antibody-based sandwich ELISA.’ J Allergy Clin Immunol, 100, 229–
234.
49. Ben Rejeb, S, Davies, D, Querry, J, Cleroux, C, Langlois, D and Delahaut, P (2002)
‘Enzyme immunoassay for the detection of crustacean proteins in food.’ AOAC
Annual Meeting Abstracts, Los Angeles, CA, Abstract C-124, p. 102.
50. Lu, Y, Oshima, T, Ushio, H and Shiomi, K (2004) ‘Preparation and characterization
of monoclonal antibody against abalone allergen tropomyosin.’ Hybrid Hybridomics,
23, 357–361.
51. Verrez-Bagnis, V and Escriche-Roberto, I (1993) ‘The performance of ELISA and
dot-blot methods for the detection of crab flesh in heated and sterilized surimi-based
products.’ J Sci Food Agric, 63, 445–449.
52. Bock, S A, Muñoz-Furlong, A and Sampson, H A (2001) ‘Fatalities due to anaphylactic
reactions.’ J Allergy Clin Immunol, 107,191–93.
53. Bjornsson, E, Janson, C, Plaschke, P, Norrman, E and Sjoberg, O (1996) ‘Prevalence
of sensitization to food allergens in adult Swedes.’ Ann Allergy Asthma Immunol, 77,
327–332.
54. de Martino, M, Peruzzi, M, de Luca, M, Amato, A G, Galli, L, Lega, L, Azzari, C
and Vierucci, A (1993) ‘Fish allergy in children.’ Ann Allergy, 71, 159–165.
55. Food and Agriculture Organization of the United Nations. (1995) Food allergies,
Report of the Technical Consultation of the Food and Agriculture Organization of
the United Nations, FAO, Rome, November 13–14.
56. Beck, H I and Nissen, B K (1983) ‘Contact urticaria to commercial fish in atopic
persons.’ Acta Derm Venereol (Stockh), 63, 257–260.
57. Cronin, E (1987) ‘Dermatitis of the hands in caterers.’ Cont Derm 17, 265–269.
58. Elsayed, S and Bennich, H (1975) ‘The primary structure of allergen M from cod.’
Scand J Immunol, 4, 203–208.
59. Lehky, P, Blum, H, Stein, E and Fischer, E (1974) ‘Isolation and characterization of
parvalbumins from skeletal muscle of higher vertebrates.’ J Biol Chem, 249, 4332–
4334.
60. Aas, K and Elsayed, S (1975) ‘Physio-chemical properties and specific activity of a
purified allergen (codfish).’ Dev Biol Standard, 29, 90–98.
61. Elsayed, S and Aas, K (1971) ‘Characterization of a major allergen (cod). Observations
on effect of denaturation on the allergenic activity.’ J Allergy, 47, 283–291.
62. Hansen, T K, Bindslev-Jensen, C, Skov, P S and Poulsen, L K (1997) ‘Codfish
allergy in adults: IgE cross-reactivity among fish species.’ Ann Allergy Asthma Immunol,
78, 187–194.
63. Galland, A V, Dory, D, Pons, L, Chopin, C, Rabesona, H, Gueant, J L and Fleurence,
J (1998) ‘Purification of 41 kDa cod-allergenic protein.’ J Chromatogr B Biomed Sci
Appl, 706, 63–71.
64. Dory, D, Chopin, C, Aimone-Gastin, I, Gueant, J L, Guerin, L, Sainte-Laudy, J,
290 Detecting allergens in food
16.1 Introduction
The development and production of hypoallergenic infant formulas (HIF)
present some unique food allergen control challenges. Almost all infants
using HIF have already developed strong allergies to cows’ milk and soy
proteins. Some of these infants will react violently to extremely small amounts
of allergen.1 Formulas made for this population represent a severe test of
allergen content control in manufactured foods.
HIF are usually made using extensively hydrolyzed proteins selected based
on their almost completely diminished immunologic reactivities.2 Current
HIF use enzymatically hydrolyzed cow milk casein or whey proteins.
Manufacturing hydrolysates for HIF is a challenging biochemical task. As
the amount of hydrolysis increases, the functional properties, organoleptic
characteristics, and biochemical stability of the hydrolysate deteriorate. During
hydrolysate development these factors must be balanced while achieving the
required reduction in immunologic reactivity. Once a satisfactory hydrolysis
procedure is set, it must be validated and controlled, and each hydrolysate
ingredient batch must be tested against strict allergen content specifications.
HIF are often produced in facilities that also manufacture other products
containing intact or partially hydrolyzed cows’ milk and/or soy protein allergens,
often using at least some shared equipment. In this environment, HIF allergen
quality assurance requires careful control of ingredients, effective and consistent
equipment cleaning procedures, processing and packaging controls, and valid
testing methods to verify product identity and quality. These methods and
procedures must be executed in an appropriately documented total quality
environment that includes allergen awareness training for plant personnel.
294 Detecting allergens in food
100
Peanut
90 Hazelnut
80 Egg
70 Soy
% Responders
Milk
60
50
40
30
20
10
0
0.01 0.1 1 10 100 1000 10 000
Allergen dose (mg Protein)
Fig. 16.1 Food allergen reaction thresholds (by DBPCFC). Ingested allergen dose
(mg protein) vs % allergic responses in challenged patients. Adapted from reference 7.
Allergen quality assurance for hypoallergenic formula 297
antigen that may contain only one or two antibody-binding epitopes. Even
these small fragments can trigger allergic reactions in some patients. This
makes assay formats based on antigen capture or ‘sandwich’ approaches
unusable. Measuring hydrolysate allergen content is made easier because
these ingredients are usually highly soluble powders that can be dissolved at
relatively high concentrations to achieve desired method sensitivity. The
assay may also be carried out in buffer systems that are optimized for method
performance. Data are best reported in units of μg immunologically active
antigen/g total protein. Hypoallergenic hydrolysates usually range from 20–
100 μg immunologically active antigen/g protein. Assuming that these
ingredients are tested at a concentration of 10 mg protein/mL, the ELISA
method should be sensitive in the range of < 200 ng/mL.
Testing for contaminants from non-protein ingredients, carry-over from
production equipment, and the manufacturing environment can be accomplished
using sandwich-type assays since the contaminants will usually be intact
proteins from other ingredients and products. These capture assays are highly
sensitive and are generally less time-consuming. Capture assays also operate
with less interference from sample matrix anomalies and can be validated for
use with a number of liquid and solid product forms. Currently, a number of
commercial ELISA kits are offered to measure allergenic food residues
(Neogen, Lansing, MI; ELISA Systems Ltd, Brisbane, Australia; R-Biopharm,
Darmstadt, Germany; Tepnel Life Sciences PLC, Deeside, UK).
Samples for these tests include product contact surfaces and environmental
samples collected by swabbing techniques. The swabbing methods are similar
to those used to collect samples for microbiological testing. A standard
surface area is swabbed, then the swab is placed in a set volume of solvent
(usually a neutral buffer with dilute detergent) and the dissolved allergen is
measured. A second sample type is rinse water following a clean-in-place
(CIP) cycle used to clean closed liquid processing systems. Care must be
exercised in collecting these samples. The samples must be a representative
rinse water sample and not a diluted cleaning fluid. Chemicals in cleaning
fluids in many cases will interfere with the assay immunochemistry. The
preferred protocol is to perform the CIP cycle, then flush the system with a
volume of clean water similar to the normal volume of liquid product that is
used to flush the system prior to initiating product collection during normal
production. Sensitivity requirements for assays used in this application are
difficult to assess since there is usually not a clear relationship between
antigen levels found on product contact surfaces or in rinse water samples
and antigen content of the initial amounts of formula product produced at
start-up. This will be discussed in more detail in Section 16.4.
Measuring residual allergen in finished hypoallergenic products has
immunochemical and sensitivity requirements similar to hydrolysate
characterization methods. Capture format assays are not useful. The tests are
also made more difficult by interference from matrix effects associated with
the emulsified formula. Similar problems in non-hypoallergenic food
Allergen quality assurance for hypoallergenic formula 299
laboratory animals seem to be the best choice. These antisera can be obtained
in relatively large amounts using standardized food allergen samples as
immunogens. This yields reproducible potency and specificity, especially
when more than 10 animals are immunized and the processed and qualified
antisera are pooled. Animal antisera generated in this way are preferable to
food allergen-specific human IgE pools because they can be reproducibly
generated, have a broader and more consistent specificity profile, and are
cheaper and more widely available. The disadvantage of using animal antisera
is that allergen epitopes recognized by allergic humans are not necessarily
the same as immunogen epitopes recognized by hyperimmunized animals.
Food allergy in humans tends to have limited and individually variable reactivity
patterns to the complex mixture of proteins contained in a food allergen.
Establishing a pool of human IgE antisera that reacts strongly with all proteins
in a selected food allergen (and only those proteins) has not been feasible to
this point. Use of IgG antibodies from animal antisera means that the ELISA
is technically measuring food antigens, not food allergens. However, while
antigens may or may not be allergens, all allergens must be antigens. Using
animal antisera to measure food antigens may overestimate food allergen
content, but this theoretical error is on the side of product safety. Operationally,
the use of animal antisera has not been a problem in interpreting food allergen
information in the hypoallergenic formula industry for more than 15 years.
Having selected hyperimmunized animals as the source of antibody for
food antigen analysis, interest then turns to optimizing antibody potency and
specificity. Both are controlled by the hyperimmunization protocol.9 Animal
immunizations should be given at relatively high antigen doses (5 mg protein/
immunization in 4 mL of immunogen), in a potent oil-based adjuvant (Complete
Freund’s Adjuvant, CFA) processed mechanically to form a stable emulsion
(two volumes CFA: one volume antigen solution). Animals are given a booster
immunization at three weeks (5 mg protein, one volume CFA: one volume
antigen solution) with additional boosters at four to six week intervals
(substituting Incomplete Freund’s Adjuvant for CFA). Antisera collection is
started at four weeks and can continue for several months. Typically, at least
10 animals are immunized. This immunization protocol assures antisera potency
and can generate antibody titers greater than 10 million.13 Antigen-specific
antibody titers are obtained from individual animals at all time points and
evaluated for sample pooling. After the desired serum pool is made and
mixed thoroughly, the pool is aliquoted into cryo storage vials and stored at
< –70 °C. Antibody activity stability under these conditions is > 10 years.
Upon thawing, the cryo vial contents are realiquoted into smaller freezer
tubes that are used as working sources. These are stored frozen (~ –20 °C)
in a non-frost-free freezer and are stable for at least 12 months.
Table 16.2 Relative antisera and assay specificity (from ELISA data)
Sample integrity
Sample integrity is critical in food allergen testing, especially with the high
sensitivity of the test methods. Sample contamination must be avoided. Special
care must be used in sample collection where contaminated collection
equipment and poorly cleaned equipment sampling ports can cause problems.
Another concern is that very low but significant levels of antigen can be lost
from sample solutions if the antigenic proteins adsorb to the surface of the
304 Detecting allergens in food
sample container. For rinse water samples this can be avoided by using EPA
(US Environmental Protection Agency)-approved glass vials designed for
water testing. Place 5 mL of a 10X concentrated buffer containing Tween®
20 (ICI Americas Inc, Wilmington, DE) detergent into the vial, then add 45
mL of the rinse water sample. The resulting solution is a 10% dilution of the
sample that is properly buffered while any protein is kept in solution by the
dilute detergent. Sample integrity issues should be resolved on a case-specific
basis during method validation.
Data handling
The final element of analytical quality is data manipulation and reporting. As
in all cases, the automated data collection and calculation hardware and
software must be justified and validated. Standard curves will be used and
the fit parameters for standard curve acceptance must be established. Acceptable
control sample performance parameters will be established and a data quality
acceptance tree can be constructed (Figure 16.2). Consistent data reporting
in meaningful units of measurement is also important. Since agreed food
allergen standards do not exist, antigen content data will not be traceable to
officially designated standard materials. The chosen assay standards must
therefore be internally consistent to establish data quality. Linking data collected
using internal standards with the clinical performance of formula in DBPCFC
trials establishes the utility of the assay data as a product quality control tool.
Optimal reporting units for hypoallergenic formula quality assurance testing
are situational: for hydrolyzed protein ingredients and hypoallergenic formula,
Allergen quality assurance for hypoallergenic formula 305
Yes: Go to step 2.
16.4 Applications
16.4.1 Ingredient identification
Quality assurance for hypoallergenic formula begins with selection of protein
hydrolysates with the capacity to achieve hypoallergenic clinical performance.
Whether these ingredients are self-manufactured or purchased from a vendor,
a careful analysis of the hydrolysate manufacturing process is required. The
goal of this effort is to reproducibly manufacture a hydrolysate with an
extremely consistent and well-controlled degree of hydrolysis that is free of
contaminants. Hypoallergenic utility of the hydrolysate is best predicted
using animal models of immunogenicity1, 13 and established in blinded food
challenge clinical studies with food-allergic patients. Before this research is
started, it is critical that the process for manufacturing the hydrolysate be
consistent and fixed. This is not a straightforward task. Variables include the
nature and consistency of the intact protein substrate, the consistency and
composition of the enzyme and/or chemical systems used for hydrolysis, the
reproducibility and control of the hydrolysis process (time, temperature profiles,
concentrations of reactants over time, pH and ionic character control),
microbiological control, reproducibility of post-hydrolysis purification, drying
conditions, packaging, storage conditions, and transportation. In this process
the ability to measure very low levels of all allergenic protein systems present
in the hydrolysate manufacturing facility is important. The inhibition format
ELISA is clearly the best method available for this purpose. The end result
of this research is a rigorously defined and validated set of ingredients,
procedures, and specifications, along with a quality system that will ensure
Allergen quality assurance for hypoallergenic formula 307
their execution, which produces a consistent hydrolysate with very low residual
antigen content that is free of contamination.
The use of single protein-dedicated or ‘functionally dedicated’ facilities
for hypoallergenic hydrolysate production is most helpful and allows monitoring
of only one protein allergen source to establish adequate cleaning. If additional
proteins must be present in the production environment, additional allergen
testing will be necessary. Environmental controls deserve special attention
when configuring these facilities. Special care must be taken to control allergen-
containing dust if the protein substrate or any of the hydrolysis components
are powdered products (as is most often the case). Creative air handling and
filtration can be of great value in controlling airborne allergens. Protein dust
may be vacuumed or washed clean but should never be blown with compressed
air. The analytical goal is to perform environmental testing to identify and
eliminate allergen hot spots, then correlate the cleaned environment with
production of hydrolysate batches that are free of contamination. Controlled
repetition of the cleaning process with appropriate ELISA monitoring of
antigen content of the environment and hydrolysate produced after cleaning
will establish validity of the cleaning process for both environment and
production equipment. These data will also validate the allergen integrity of
the production processes and their controls.
to raise awareness of the serious nature of food allergies in all who contribute
to formula quality. It is vital to gain commitment from operators, analysts,
and managers to proactive implementation of food allergen safety programs.
Training should foster continuous improvement in procedures and equipment
to ensure the food allergy safety of products. There is no substitute for
informed, committed, and empowered employees in food allergen control.
Generally, food allergen awareness training should include the what, why,
and how of food allergen control. This will begin with a description of food
allergies, what causes the allergic reactions, and the serious consequences
including death that can result from uncontrolled allergic reactions to food.
The regulatory environment should also be addressed, describing the increased
emphasis on food allergen regulation and FDA inspection, and the growing
food allergen product recall pattern. The potentially life-threatening
consequences for consumers should be re-emphasized and product liability
risks should be covered. Training should raise awareness in the context of
the regulations and establish personal commitment to proactively managing
food allergens in the manufacturing environment. With that accomplished,
training on allergen management can begin. This will include training on
plant-specific allergen control procedures for ingredient receipt, acceptance
testing and storage, equipment and environment cleaning procedures, allergen
testing and data interpretation, allergen control in formulation and processing,
and the importance of labeling information and packaging. Finally, the quality
systems that ensure compliance with food allergen management procedures
should be described and the trainees should be invited to suggest further
improvements as opportunities arise.
Training for operators and production supervisors is usually most meaningful
when it is custom designed for individual production environments. Training
for middle and upper managers should be more global and take advantage of
combined information from a variety of food industries. The University of
Nebraska’s Food Allergy Research and Resource Program (Lincoln, NE,
USA; www.farrp.org) provides an excellent short course on food allergen
management in industry and is recommended for this group.
has reduced but not eliminated antigen. In trial 3, modifying the CIP cleaning
fluids has effectively cleaned the heat exchanger while acceptable cleaning
of the valve requires disassembly and hand cleaning. The can filler is also
effectively cleaned using a warm water rinse. This scenario, while theoretical,
is based on actual cleaning trial data and is presented to illustrate data
interpretation. Once the cleaning processes are optimized they should be
fully documented and replicated to yield an allergen cleaning validation
package and standard operating procedures (SOPs) for allergen cleaning.
lines, sampling and cleanup tools might all be considered for dedication.
Some disadvantages of equipment dedication in a mixed product manufacturing
setting are maintaining operator training to use only the right equipment and
keeping accurate records of compliance. Clearly, dedicated equipment in a
mixed manufacturing environment does not ease the requirements to execute
validated equipment and environmental cleaning.
Assuming a mixed manufacturing or partially dedicated facility, the first
step in manufacturing control is to optimize product production scheduling.
This is a simple but powerful tool that takes advantage of a known hierarchy
in product allergenic reactivity: hypoallergenic products are least allergenically
active and are made first following the required cleaning, soy-based products
have relatively low allergenic reactivity7 and are made next, cows’ milk-
based products are most allergenic and are manufactured last in the cycle. It
is also helpful to schedule longer run times, producing a number of formula
batches and thereby minimizing the costs and down time associated with
cleaning between product change-overs.
In-process testing of the formula for antigen content is not required but
may be useful, especially when starting new production lines or incorporating
new production equipment. The advantage of in-process testing is that it
allows savings of sterilization and packaging costs of contaminated batches
that must be discarded, and it provides information useful in investigating
possible sources of contamination.
not include many key allergens, and there is a general lack of standardization
and validation. There are no widely accepted standards for allergens, antigens,
or antisera. Also lacking are standard assay protocols, sample collection and
preparation procedures, data calculation and reporting tools, and assay
validation data for food quality assurance applications. Substantial progress
is being made to fill these gaps and new options, better understanding, and
new partners will change the future.
A postscript regarding allergen quality assurance of ‘non-allergenic’
‘elemental’ diets: Recall that by definition, a ‘clinically hypoallergenic’ formula
can have an allergic reaction failure rate of up to 10%. Patients who react to
hypoallergenic formula must use diets that are devoid of immunological
reactivity. The protein components of these formulas are based on purified
amino acids and the products must be completely free from food protein
contamination. Amino acid-based diets were initially developed to manage
digestive difficulties. In these applications small amounts of intact protein
contamination did not have a significant negative metabolic effect on the
diet’s performance. However, when the diets are used to control extremely
severe food allergies, additional allergen quality assurance requirements must
be observed. HIF testing technology can be used with some slight modifications:
first, product contamination will only be with intact proteins from the production
environment, therefore commercially available capture-format ELISAs can
be used; second, testing will need to include all protein sources present in the
manufacturing facility; finally, while amino acid-based diets can be shown
to be clinically hypoallergenic, there are (can be) no testable clinical
performance standards for ‘nonallergenic’ formulas. Operationally, the modified
quality assurance programs have enabled us to successfully manage the
allergen quality of amino acid-based diets used in highly allergic patients for
more than six years.
16.6 References
1. Amonette, M S, Schwartz, R H, Mattson, L, Peers, L B and Eldredge, D M (1991)
‘Double-blind, placebo-controlled food challenges (DBPCFC) demonstrating acute
IgE-mediated allergic reactions to good start, ultrafiltered good start, alfare, nutramigen,
and alimentum in a seven-year-old’, Pediatr Asth Allergy Immunol, 5, 245–251.
2. Cordle, C T (1994) ‘Control of food allergies using protein hydrolysates’, Food
Techno, 48, 72–76.
3. American Academy of Pediatrics, Committee on Nutrition, ‘Hypoallergenic infant
formula’, Pediatrics, 2000, 106, 346–349.
4. Host, A, Koletzko, B, Dreborg, S, Muraro, A, Wahn, U, Aggett, P, Bresson, J L,
Hernell, O, Lafeber, H, Michaelsen, K F, Micheli, J L, Rigo, J, Weaver, L, Heymans,
H, Strobel, S and Vandenplas, Y (1999) ‘Dietary products used in infants for treatment
and prevention of food allergy. Joint Statement of the European Society for Paediatric
Allergology and Clinical Immunology (ESPACI) Committee on Hypoallergenic
Formulas and the European Society for Paediatric Gastroenterology, Hepatology
and Nutrition (ESPGHAN) Committee on Nutrition’, Arch Dis Child, 81, 80–84.
314 Detecting allergens in food
17.1 Introduction
Food allergy has been long recognised as a clinical phenomenon, with numerous
reports in the 20th century medical literature.1,2,3 However, while it was
known that patients could suffer extremely severe and sometimes fatal reactions
following ingestion of minute amounts of the offending food, food allergy
was perceived as a problem for the individual sufferers. Since the late 1980s,
however, this perception has changed, and food allergy is now recognised as
an important public health problem. A major factor in this increased concern
is probably the rise in the prevalence of atopic disease,4 of which it can be
considered a manifestation. The prevalence and incidence of food allergy
and the number of severe reactions5 may be increasing, although the lack of
sound baseline epidemiological data precludes firm conclusions. The new
perception of food allergy has been accompanied by the recognition that the
solution to the problem lies with collaboration between all the stakeholders,
including patients and those who look after them, clinicians, public authorities
and the food industry.
The ultimate aim for all stakeholders is to avoid food allergy sufferers
reacting to the allergens to which they are sensitised. This can be achieved
in two ways. One is to ensure accurate allergen declaration through labelling,
the other is to ensure that where a specific allergen is not declared, the
product does not contain it in an amount that would pose a risk and food
allergy sufferers can assume it is safe for them. Both these requirements can
only be fulfilled by detailed knowledge of the composition of products. Food
manufacturing processes are extremely complex. This complexity derives
from several factors including material sourcing, processing, efficient use of
316 Detecting allergens in food
For instance, Unilever has a policy for dealing with allergens which states
that it shall declare the presence in its products of any allergen which is a
common cause of allergic reactions. At a minimum, any allergen required by
local regulations will be declared. However, beyond that, the allergenic risk
from foods not commonly known to be allergenic may be assessed if clinical
or epidemiological data indicate the need. If then classed as a common cause
of allergic reactions, this food component would be declared on labels and
included in hazard analysis of critical control points (HACCP) plans. Unilever
also undertakes to inform any consumer on request about the presence of
uncommon allergens in specific products.
Allergen management guidelines need to ensure that allergens are correctly
and intelligibly declared in products, but also to make sure that allergen is
not present inadvertently at levels likely to cause adverse effects on health.
Such guidelines specifically need to address all stages in the product life-
cycle, from its design, through the sourcing of ingredients, to manufacture,
labelling and distribution. Specifically, they need to deal with:
• Innovation. Is the use of the allergenic ingredient necessary for the
functionality of the product or could an equivalent non-allergenic ingredient
serve as well?
• Supply chain. Control of allergens in the supply chain requires a close
relationship between suppliers and manufacturers so that the needs and
requirements of the latter can be met. Typically, the starting point of the
supplier assessment will be a questionnaire about allergens handled and
precautions in place to avoid cross-contact, including the existence of a
HACCP plan. This is backed up by periodic audits of the suppliers’ facilities.
Additionally, suppliers are required to seek agreement to any change in
the formulation of the ingredient they supply.
• Manufacturing protocols. Main considerations are the inclusion of common
allergens in HACCP plans, production scheduling to minimise cross-contact,
validated cleaning procedures and clear labelling and separation of
specific allergenic ingredients within the factory. Procedures need to cover
rework, where sound product is not packaged but ‘recycled’. Staff training
to understand the importance of allergen control procedures is vital and
improves support for what can be additional procedures in the production
process. Finally, the same degree of attention is needed whether the
company’s own manufacturing facility is concerned or that of co-packers.
• Packaging, promotion and advertising. Packaging carries the label and
therefore the allergen information. Care is required to ensure that information
remains with the product until it reaches the consumer. Other considerations
include warnings if the formulation has changed to include an allergenic
ingredient previously not present.
• Retailers. Generally, the manufacturer’s allergen information will be
sufficient. However, situations such as in-store promotions require care to
ensure that the consumer is fully informed. Sound product, which fails to
meet all standards for general sale, may be repackaged and sold in specialised
320 Detecting allergens in food
trying to monitor the effect of processing on such proteins, but may give
highly misleading results if used in an assay intended for other purposes,
such as the estimation of the extent of cross-contact or establishing whether
a product contains more allergenic material than a set limit. The key
consideration with respect to assay development should therefore be the
purpose of the assay. The allergenic protein, although the obvious candidate,
may not always be the optimal choice. However, detecting the protein is
probably the most common approach and it is therefore appropriate to discuss
the options available.
In developing an assay based on protein detection, two main choices of
methodology exist: monoclonal and polyclonal antibody technology. Both
have advantages and drawbacks. As monoclonal antibodies recognise single
epitopes on proteins, this technology usually results in a highly specific
assay, with relatively low incidence of cross-reactivity, even with closely
related proteins, provided the antibodies have been correctly screened.
Theoretically there is also an endless supply of antibody with exactly the
same performance characteristics as the original antibody. However, the narrow
specificity of the monoclonal antibody can also be its Achilles’ heel, as
detection of the protein of interest will only take place if the antibody binding
site remains intact and accessible in the various food matrices in which the
protein may be present. Polyclonal antibody technology relies on the production
of a range of antibodies, either to a single protein of interest, or to all the
proteins within the food, depending on the preparation which is used. This
provides a detection system which is less likely to fail completely to identify
the presence of proteins of interest, although quantitation may remain
problematical if processing alters the relative proportions of the different
immunochemically active proteins in a food. However, the main drawbacks
of polyclonal technology lie in the need for extensive purification procedures
that may need to be applied to the protein(s) of interest, as well as to the
resulting antiserum to ensure specificity and absence of unwanted cross-
reactivity, together with the need to develop procedures to ensure batch to
batch reproducibility.
Once the technology itself has been selected, there remain several
possibilities in developing immunoassays based on protein. As discussed,
monoclonal technology results in a highly specific detection system, but it
can nevertheless be broadened by using a combination of detection antibodies
against different epitopes of the same protein, and/or different proteins.
However, beyond a few proteins, this becomes complex to optimise. Polyclonal
technology leaves open the choice of the material against which the antiserum
can be raised. Thus it can be as general as a protein extract of the whole food,
or as specific as a highly purified protein.
However, detection of the protein, or proteins, may not be necessary or
even be the method of choice in all instances where detection of allergenic
residues is sought. Instead, a marker molecule, for which a robust and sensitive
analytical method exists and which is always found in a known ratio to the
Issues in detecting on equipment and in processed food 323
Matrix interference
As well as interfering with the recovery of the analyte(s) of interest, the food
matrix, or some of its components, may actually interfere with the subsequent
assay, if those components are co-extracted in sufficient amounts. For instance,
we have found that on occasions, solutions with very high sugar content
(although within the range used in several foods) reduced considerably the
recovery of β-lactoglobulin (unpublished results). Other materials commonly
used in foods, such as colours, could obviously also interfere with the
performance of assays based on colorimetric endpoints, depending on their
fate during extraction.
patients and their support groups for the means of monitoring foods for the
presence of cross-contact allergens may lead to development of some rapid
assays. The likely influence of each of these trends will be examined separately.
The legal framework with respect to food allergens is developing fast,
with Switzerland, Japan, Australia/New Zealand, the United States and the
European Union bringing in legislation specifying which allergenic foods
must be declared. The lists are usually based on the list of allergens in the
Codex General Standard on Labelling, but extend it to cover allergens of
regional importance such as celery and mustard in the European Union. This
legislation can be anticipated to drive food manufacturers to use test kits
much more extensively to demonstrate for legal purposes that their allergen
risk management procedures are effective. Although allergen testing has not
proved to be the primary mechanism of enforcement, in some legislatures
where it has a longer history such as the USA, enforcement authorities will
undoubtedly seek to use them to support other evidence. Except for Switzerland,
current legislation does not address the issue of allergen presence through
cross-contact, and action levels have not yet been set. However, as allergen
test kits become used to a greater extent, pressure is likely to grow, particularly
from manufacturers, for defined action levels, below which the presence of
the allergen would not constitute an infringement of the law. If action levels
are not set by the agencies or the legislators, they will likely be defined by
case law, which is probably not an ideal mechanism for this type of issue.
Determination of NOAELs and their use will provide manufacturers with
defined targets for their allergen management policies, in terms of what
amounts constitute a risk to what proportion of food allergy sufferers. They
will also provide manufacturers with information for improved control of
allergen hazards. Such control will, however, require that they know what
level of allergenic residues is present in their products. Measurement of
allergenic residues at appropriate points during the manufacturing process
will be one way to obtain this information and could therefore increase
considerably from its current relatively limited use.
Monitoring the allergenicity of certain foods or food products is another
area where detection of allergenic residues could play an increasing role, as
manufacturers seek to provide foods with reduced allergenicity. However,
this area is probably likely to have a lower impact than the previous two, as
looking for residual allergenicity by protein quantification is only one of
several steps in defining reduced allergenicity.
Food allergy significantly impairs quality of life for sufferers.23,24 Greater
control over their condition by food allergy sufferers would undoubtedly
help restore some of this quality. Demand for means to do so could spur an
extension of the measurement of allergenic residues to this totally new area.
This prospect is probably still quite distant, inasmuch as it requires methods
which are simple to use and robust. A critical question will be the extent of
the test manufacturer’s legal liability in the event of an allergen not being
detected and producing a reaction in a sufferer.
328 Detecting allergens in food
17.6 References
1. Prausnitz, C and Küstner, H (1921) ‘Studien über die Ueberempfindlichkeit’, Centralbl
Bakteriol Abt Orig, 86, 160–169.
2. Loveless, M H (1950) ‘Milk allergy: a survey of its incidence: experience with
masked ingestion test’, J Allergy, 21, 489.
3. Tuft, L and Blumstein, G I (1942) ‘Studies in food allergy. Sensitization to fresh
fruits: clinical and experimental observations’, J Allergy, 13, 574–581.
4. Lewis, S, Butland, B, Strachan, D, Bynner, J, Richards, D, Butler, N and Britton, J
(1996) ‘Study of the aetiology of wheezing illness at age 16 in two national British
birth cohorts’, Thorax, 51, 670–676
5. Sheikh, A and Alves, B (2000) ‘Hospital admissions for acute anaphylaxis: time
trend study’, Br Med J. 320, 1441.
6. Bruijnzeel-Coomen, C, Ortolani, C, Aas, K, Bindslev-Jensen, C, Bjorksten, B, Moneret-
Vautrin, D, Wuthrich, B (1995) ‘EAACI position paper. Adverse reactions to food’
Allergy, 50, 623–635.
7. American Academy of Pediatrics (2000) ‘Hypoallergenic infant formulas’, Pediatrics,
106, 346–349.
8. Bindslev-Jensen, C, Briggs, D and Osterballe, M (2002) ‘Can we determine a threshold
level for allergenic foods by statistical analysis of published data in the literature?’
Allergy, 57, 741–746.
9. Steensma, D P (2003) ‘The kiss of death: a severe allergic reaction to a shellfish
induced by a good-night kiss’, Mayo-Clin-Proc, 78, 221–222.
10. Said, M and Weiner, J M (2004) ‘May contain traces of…’: hidden food allergens in
Australia’, MJA, 181, 183–184.
11. Kosa, K M, Cates, S C, Post, R C and Canavan, J (2004) ‘Consumers’ attitudes
toward labelling food products with possible allergens’, Food Prot Trends, 24, 605–
611.
12. de Jong, E C, Van Zijverden, M, Spanhaak, S, Koppelman, S J, Pellegrom, H and
Penninks, A H (1998) ‘Identification and partial characterization of multiple major
allergens in peanut proteins’, Clin Exp Allergy, 28, 743–51.
13. Lewis, S A, Warner, J O and Hourihane, J (2003) ‘Promiscuity of IgE binding to
peanut allergens correlates with clinical reactivity to whole peanut during double-
blind placebo controlled challenge’, Poster 136, presented at the XXIInd Congress
of the European Academy of Allergy and Clinical Immunology, Paris, June 7–11.
14. Knippels, L M J, Penninks, A H and Bannon G A (2003) ‘The sensitising potential
of peanut proteins in four different mice strains’, Poster 681, presented at the XXIInd
Congress of the European Academy of Allergy and Clinical Immunology, Paris,
June 7–11.
15. Taylor, S L, Hefle, S L, Bindslev-Jensen, C, Bock, S A, Burks, A W, Christie, L, Hill,
D J, Host, A, Hourihane, J O, Lack, G, Metcalfe, D D, Moneret-Vautrin, D A, Vadas,
P A, Rancé, F, Skrypec, D J, Trauman, T A, Malmheden, Yman, I and Zeiger, R S
(2002) ‘Factors affecting the determination of threshold doses for allergenic foods:
How much is too much?’ J Allergy Clin Immunol, 109, 24–30.
16. Crevel, R W R, Kerhoff, M A T and Koning, M M G (2000) ‘Allergenicity of refined
vegetable oils’; Food Chem Toxico, 38, 385–393.
17. Morisset, M, Moneret-Vautrin, D A, Kanny, G, Guénard L, Beaudouin E, Flabbée,
J and Hatahet, R (2003) ‘Thresholds of clinical reactivity to milk, egg, peanut and
sesame in Immunoglobulin E-dependent allergies: evaluation by double-blind or
single-blind placebo-controlled oral challenges’, Clin Exp Allergy, 33, 1046–1051.
18. Bradford, M M (1976) ‘Rapid and sensitive method for quantification of microgram
quantities of protein utilizing principle of protein-dye binding’, Anal Biochem, 72,
248–254.
19. Smith, P K, Krohn, R I, Hermansson, G T, Mallia A K, Gartner, F H, Provenzano,
Issues in detecting on equipment and in processed food 329
18.1 Introduction
More than 40 years have passed since the first publication of the
radioimmunoassay technique by Yalow and Berson.1 Despite pessimistic
prophecies about its efficacy, it has survived, developed, and recently been
augmented by other diagnostic tests to become a powerful tool to investigate
very small amounts of substances in the nanogram to picogram range.
Developed originally to analyze hormones, the method is nowadays used for
a wide range of applications, for example, in the area of clinical diagnosis,
food and feed diagnostics and to test for environmental pollutants.
The effectiveness of any immunoassay depends directly on the quality of
the antigen used as a target and the quality of the antibody used for capture
and detection but in addition to this, the performance of the assay itself is
important. Allergens are proteins, and therefore immunoassays are appropriate
tools to measure them. Often more than one allergenic protein exists in a
complex matrix amongst many other proteins. The nature of food can vary
over an extremely wide range and sample preparations are often very difficult
to work with. Many food products are affected by heating, which can alter
the allergenic and non-allergenic proteins. So there are two components
which can influence the assay performance; one comes from the extraction
of the allergen and the other from the assays themselves.
Factors affecting the effectiveness of allergen detection 331
2.5 100
Sandwich ELISA Competitive ELISA
90
2 80
70
1.5 60
OD
OD
50
1 40
30
0.5 20
10
0 0
0 2 6 18 54 10 30 90 270 810
egg protein (ppm) β-Lactoglobulin (ppb)
Test performance
Because it is desirable that results should be available as fast as possible, the
tendency is to shorten the assay performance time more and more. Speed,
however, comes at a price. Antigen–antibody reactions are subject to the
laws of chemical equilibrium. If the reaction is interrupted before the steady
state is reached, the assay becomes subject to greater variability. Usually it
is required that assays should not take longer than a few minutes, at most one
to three hours. To obtain consistent results in commercial kits, the manufacturer-
recommended temperature and incubation times have to be strictly adhered
to. During each moment of incubation, the result is changing. Times and
temperatures are optimized in such a way that the recovery of the analyte
should be nearly 100%. The manufacturer has to check the robustness of the
assay concerning variation of time and temperature (in most cases a variation
of about 5% is tolerated) to guarantee the correctness of results within certain
limits.
It is necessary to understand the basic principle of the type of test being
used. In competitive assays, the reaction is initiated by adding the antibody
to the wells, which allows for slow and deliberate pipetting of samples and
standards. In the sandwich-type format, the reaction is more immediate, and
pipetting must take place without delay, since the capture antibody on the
plate binds the allergenic residues on initial contact. In the latter type of
assay, an analyst has a little bit more time if a reaction step covers about 30
minutes or more. But if the assay time takes only a few minutes (5–10) the
technician has a challenge to pipette quickly. Furthermore, for commercial
kits, it is clear that more assay wells can be processed if the assay takes a
longer time. Hence, it is important that the same velocity of pipetting is done
throughout the entire test procedure.
It is clear that the maximum number of possible samples which may be
investigated depends on the reaction time as well. An assay with a total of
five minutes incubation time will be designed with only a few standards,
with the standards and samples running as single determinations and the
number of samples not greater than 10 so that the technician is able to pipette
them within a minute. Otherwise the delay between the first and the last
pipetting is too long. If an incubation time of about 30 minutes or more is
included, it is possible to prepare half a plate or more, provided that the
pipetting is finished in less than five minutes.
For commercial kits, all of the included test components have to be handled
carefully according to the manufacturer instructions. Before using, kit
components should reach room temperature and they need to be mixed
thoroughly without creating foam within the solutions.
One of the most important points for either in-house methods or commercial
Factors affecting the effectiveness of allergen detection 333
0 3.067 0.075
10 3.067 0.594
20 3.240 1.118
40 3.064 1.931
80 2.882 2.651
334 Detecting allergens in food
For both commercial methods and in-house methods, the washing of the
ELISA plate between the reaction steps counts as an additional factor which
may influence the results. It is important to use the right washing buffer, the
right amount of buffer and the right number of washing steps. It is also
important to guarantee that no cross-contamination from well to well occurs
during washing. It is better to use more washing steps than too few. The plate
should be inverted strongly against very absorbent paper two or three times
after each washing step. It is necessary to ensure that the wells are clearly
empty between the washing steps and before each new reagent addition
begins, but the wells should not be allowed to dry. Improper washing often
leads to a higher background level, which results in higher absorbance values
in the blank control and, in both types of ELISA formats, gives rise to a
poorer differentiation between the lower standards. Often, a high coefficient
of variation (CV) indicates improper washing. Table 18.3 shows high
absorbance at the low standards in a sandwich-type commercial method. The
very high CV values highlighted are due to incomplete washing.
Technicians should make sure that pipettes are calibrated and take care to
pipet without splashing into neighbouring wells. In addition, the use of a
calibrated ELISA reader is absolutely necessary and should be maintained.
Moreover, some chromogens/substrate solutions should be kept in the dark
and for these, of course, the color reaction should be run in the dark as well.
Parallelism between standard and analyte is essential to producing an accurate
Table 18.3 RIDASCREEN® Gliadin ELISA: very high OD in the lower area of curve
connected with high CV due to incomplete washing
0 0.639 13.1
5 0.988 20.1
10 1.247 1.5
20 1.832 1.0
40 2.126 6.2
80 2.682 7.1
Factors affecting the effectiveness of allergen detection 335
Test characteristics
Whether using a commercial ELISA kit or an in-house method, the ELISA
336 Detecting allergens in food
must be robust and reliably employ a procedure that ideally has a minimum
number of manipulations.
For ELISA methods:
• For commercial kits, the standard curve should be nearly identical to the
one documented by the manufacturer. Most manufacturers deliver a quality
control data sheet for comparison.
• For in-house methods, internal quality control methods should be used to
make sure that standard curves run similarly on different days, with different
technicians, etc.
• Sandwich ELISAs should start with very low OD at the zero standard, and
maximal OD should be similar to that reported by the manufacturer; in the
case of an in-house method, again, internal quality control should be done
to ensure that the assay is running identically from day to day.
• Competitive ELISA curves should begin in a steep manner between the
first and second standard to guarantee accurate determination of samples
in the low range of the curve.
• Benchmarks of 80%, 50%, and 20% values can be used to characterize
the curve as well. These are the points of intersection of the curve at 80/
50/20% binding of the analyte and can be used to describe the reproducibility
of the standard curve. For commercial kits, most manufacturers indicate
the 50% level in the kit insert, which is that concentration reached at
50% binding of the analyte.
• It is advisable to establish quality control data, which should indicate the
80/50/20% levels (or whatever benchmark in-house methods users define)
from run to run as well as the maximum and minimum OD to evaluate the
method performance. Over time the analyst has the possibility to compare
the data from run to run and this will make it easier to judge a run that
experienced problems.
• Of additional importance is the CV of samples and standards, which is
calculated by dividing the standard deviation by the mean of replicate
results and multiplying by 100. A CV of greater than 10% within the assay
can indicates insufficient washing or pipetting. The CV calculated within
one run refers to the intra-assay reproducibility, while the CV calculated
between runs refers to the inter-assay reproducibility. Both terms help to
evaluate the assay performance.
• To evaluate the accuracy of test runs, known negative and positive controls
should be included within each run. The results can be included additionally
in the quality control data. This also helps to promptly recognize outlying data.
• To control the reproducibility of a run, the suggestion would be to run one
to three recovery samples with known contents of analyte (one negative
and two positive). Intra-assay and inter-assay reproducibilities as well as
the recoveries (measured mean value of the sample divided by the theoretical
value multiplied by 100) can then be calculated.
All of these parameters help to identify outlying data and enhance the accuracy
and reliability of the results obtained.
Factors affecting the effectiveness of allergen detection 337
18.3 Troubleshooting
This part of chapter is dedicated to troubleshooting in case something goes
wrong with an assay.
Factors affecting the effectiveness of allergen detection 341
Inter-assay problems
In a test, the ODs between runs of the same lot or different lots show a large
range of variance. Pipetting errors or reagent deterioration could be the
reason. Further, the stability of sample extracts should be tested. Also, the
reproducibility of the extraction itself should be inspected by a reference
sample (known concentration) by repeating the extraction. The homogeneity
of the sample should be examined. The analyst should increase the sample
size and should repeat the extraction again.
18.4.2 Dipsticks
Dipstick formats have been developed for allergenic residues – the antibodies
or antigens are immobilized to a microporous surface on a strip. The assay
can be easily performed by transferring the stick into a sample after extraction
from food. For a complete discussion of dipsticks/lateral flow devices, see
Chapter 10 of this book.
Factors affecting the effectiveness of allergen detection 345
18.5 Summary
The success of any immunoassay depends directly on the main components
(antigens, antibodies), but the performance of the assay is also important in
obtaining a reliable result.
Two kinds of errors can be found – a systematic (in the form of bias) and
a random error (reflected in poor reproducibility or ‘imprecision’). Bias and
imprecision are two statistical parameters which describe each assay. Some
suggestions to minimize random errors of an assay are made in this chapter.
The performance of a test influences the precision (intra-/interassay),
reproducibility, recovery, sensitivity and reliability of the system. It is neccessary
to understand the basic principle of the assay type one is using. The assay
conditions and the handling on the one hand and the analyte extraction
procedure on the other are important parameters influencing the result. Before
starting, the user should be clear about the assay format, the time requirements,
the number of possible samples with one run, and the standardizing/calibration
of the assay. Further, the careful handling of test components and mistakes
that can occur (adsorption, contamination, degradation) and the factors
influencing the procedure (time, temperature, washing steps, starting point
of the reaction) are covered. Today’s immunoassays are very sensitive. It is
clear that care must be taken to avoid cross-contamination in the laboratory
and some examples are given concerning possible origins of contamination.
The evaluation of results should come from a standard curve which fits the
measured values in the best way; often a cubic spline method is needed.
Extrapolations above and below the standard range of the curve should not
be made. Hints are given for an internal quality control regime, which should
be introduced into each laboratory that deals with immunoassays, to secure
the results.
The sensitivity and selectivity of the immunoassay has to be combined
with a selective and effective extraction procedure of the analyte. Factors of
interest are the incubation time and temperature during the extraction, but
most important is the suitability of the extraction buffer. Optimization is
absolutely neccessary. Tips are given on how to spike different kinds of
matrices effectively to control the assay and the extraction procedure. Finally,
recommendations are made concerning troubleshooting in the case of problems
in obtaining good analytical results.
18.6 References
1. Yalow, R S and Berson, S A (1960) ‘Immunoassay of endogenous plasma insulin in
man’, J Clin Invest, 39, 1157–1161.
2. Lopez, E, Reyes, E, Hernandez, M, Llorente, M, Mendez, E and Wieser, H (2000) ‘A
quantitative gluten “cocktail extraction” procedure for heat-processed foods’,
Proceedings of the 15th meeting of the Working Group on Prolamin Analysis and
Toxicity, Nov. Meran, 129–138.
Factors affecting the effectiveness of allergen detection 347
19.1 Introduction
Reliable detection and quantification methods for food allergens are necessary
in order to improve consumer protection and to ensure compliance with food
labelling regulations. However, the analysis of allergenic components in
food products can be very difficult, because their chemical identity is often
not sufficiently known and it is suggested that they are frequently only
present in trace amounts. Another question yet to be answered is the required
detection power and selectivity of corresponding analytical methods, as
scientifically sound threshold levels have not yet been established, e.g. by
human oral challenge studies. Threshold levels for specific allergic reactions
determined until now by double-blind placebo-controlled food challenges
(DBPCFC) range between less than 1 mg and more than 1 g of allergenic
protein depending on the food concerned and the selected individuals (Taylor
et al., 2002). There seems to be a general assumption that the detection limits
for different food products need to be around 10 ppm [mg allergen (protein)/
kg food] or lower, depending on the particular food (Koppelman et al., 1996,
Poms and Anklam, 2004). But one should keep in mind that mass is usually
not an appropriate quantification and measurement parameter for analytes
with varying chemical/biological activity such as proteins.
Currently, there are several analytical approaches for the detection of
potential allergens in food products (reviewed in Poms et al., 2004). The
methods employed are targeting either the allergen (protein) itself or a marker
that indicates the presence of the offending food. At present, detecting the
allergen per se is not always feasible, as the crucial chemical properties may
not be well characterized and understood or the detection limits of the methods
Reference materials and method validation in allergen detection 349
used are insufficient. Typically, specific proteins or DNA fragments are targeted
as markers for the presence of potentially allergenic food products or
ingredients. Protein-based methods usually involve immunochemical detection
protocols such as the radio-allergosorbent test (RAST), enzyme-allergosorbent
test (EAST), enzyme-linked immunosorbent assay (ELISA) and
immunoblotting (discussed in other chapters of this book), whereas only the
ELISA technique is presently used in routine food analysis. Methods operating
on the DNA level are usually based on an amplification of a specific DNA
fragment by the polymerase chain reaction (PCR) (see Chapter 7). Currently
the employment of DNA-based methods for the detection of allergens in
food products is a matter of controversy, since proteins are the causative
agents in allergy and PCR results cannot be linked to any allergen/protein
content. However, the choice of method is still mainly dependent on the food
concerned (availability of specific antibodies/DNA-primers and the achievable
detection limit) and on the effects of food processing during production.
Therefore, protein-based and DNA-based methods each have their own
characteristic merits and drawbacks concerning their applicability in the
detection and quantification of allergens in various food products (Poms and
Anklam, 2004).
Problem definition
Sampling
Processing, conservation
Quality assurance
Analyte identification
Quantitative measurement
Evaluation
Assessment
Feasibility study
Planning Material selection (collection, processing,
characterisation)
19.8 References
BIPM-IEC-IFCC-ISO-IUPAC-IUPAP-OIML (1993) International Vocabulary of Basic
and General Terms in Metrology (VIM), 2nd edn, Geneva, ISO.
Emons, H, Linsinger, T P J and Gawlik, B M (2004) ‘Reference materials: terminology
and use. Can’t one see the forest for the trees? Trends Anal Chem, 23, 442–449.
Hischenhuber, C, (2002) ‘Analytical methods for the detection of hidden allergens – their
use and limitations’, Minutes and Proceedings of an International Workshop on Food
Allergy: Chemical and Technological Aspects (Ispra, Italy, 2001), EUR 20241 EN
2002.
Hurst, W J, Krout, E R and Burks, W R (2002) ‘A comparison of commercially available
peanut ELISA test kits on the analysis of samples of dark and milk chocolate’,
Immunoassay Immunoch, 23, 451–459.
356 Detecting allergens in food
20.1 Introduction
A company selling food products in the US is vulnerable to potential regulatory
and product liability when the food product causes injury due to the presence
of an undeclared allergen. There is potential regulatory liability because the
US Food and Drug Administration (FDA) has long taken the position that
foods should be recalled when they are found to contain an undeclared major
allergen. In addition, the Food Allergen Labeling and Consumer Protection
Act, signed into law in August 2004, deems a food product misbranded if the
label fails to declare a major allergen by a common English name such as
‘milk,’ ‘egg,’ or ‘peanut.’ Once a company discovers that one of its products
contains an undeclared major allergen, either through its own due diligence,
in response to a consumer complaint, or in response to an FDA inquiry, the
company generally will recall the offending product. Given the severity of
reaction that can occur when a food-allergic consumer is exposed to a major
allergen, companies generally must issue press releases announcing the recall.
A company can easily spend hundreds of thousands of dollars recalling
products with an undeclared allergen.
The sale of a food product containing an undeclared allergen also presents
potential product liability. Americans have long been known for their fascination
with lawsuits and their propensity to resolve disputes through the court
system. The media carries numerous stories of consumers who have been
harmed from food and are seeking huge damages from restaurants or food
manufacturers. Indeed, customers have sued food restaurants for serving hot
coffee that caused a serious burn and for serving food that allegedly contributed
to obesity. There have also been hundreds of cases that have been brought
358 Detecting allergens in food
against the food industry for selling foods that contained pathogens such as
Salmonella or E. coli that have caused consumer injury. Numerous cases
have also been filed against the food industry for selling a food that contained
an undeclared major allergen. While the vast majority of cases are settled out
of court, the few reported cases available indicate that the potential liability
for selling a food with an undeclared allergen can range from a few thousand
to over a million dollars.
The food industry has invested considerable time and resources since the
1990s in attempting to reduce the presence of undeclared allergens in the
food supply. Other chapters in this publication focus on the various ways in
which undeclared allergens can become integrated into the food supply and
the manufacturing controls that can be implemented to reduce them. Indeed,
many companies are now using allergen test kits to monitor for the presence
of undeclared allergens that could be incorporated into foods by use of
shared equipment. These test kits can be incorporated effectively into a
company’s allergen control practices by identifying the presence of an
undeclared allergen that otherwise is not visible to the naked eye. Companies
must realize that the results from the test kit may one day be available to
regulators. If the company becomes involved in a product liability action
resulting from an alleged injury due to an undeclared allergen, the allergen
test results would also need to be made available to lawyers that are
bringing the action against the company. Although a powerful tool for
incorporation into an allergen control program, companies must give careful
consideration to how they will incorporate test kits into their allergen control
program.
that are used during the manufacturing process but then later removed), and
substances migrating to food from equipment and packaging materials.
The Food Allergen Labeling and Consumer Protection Act establishes
new requirements for the labeling of major allergens. The President signed
the law in August 2004 and its provisions become effective for products
labeled after January 1, 2006.6 At the time of this article, FDA has not yet
provided any guidance on how it intends to interpret this new law. The new
law amends the FFDCA in two significant ways. First, it defines the term
‘major food allergen’ and second, it deems a food misbranded unless the
major food allergens are declared by a ‘common English name’ that is easily
recognizable by consumers. In large part, the legislation is limited to what
are commonly called the ‘Big 8 allergens.’ ‘Major food allergen’ is defined
as ‘milk, egg, fish (e.g. bass, flounder, or cod), crustacean shellfish (e.g.
crab, lobster, or shrimp), tree nuts (e.g. almonds, pecans, or walnuts), wheat,
peanuts, and soybeans’ and food ingredients that contain proteins derived
from the major food allergens.7 These proteins nevertheless may be excluded
from this definition if they fall under one of two exceptions.
First, highly refined oils, and ingredients derived from these oils, are not
a ‘major food allergen.8 These “highly refined oils” are refined, bleached,
deodorized oils. In its report, the Committee notes that the legislation would
not change the common or usual name of these highly refined oils. Thus,
oils, such as peanut oil, would still be labeled as such in ingredient statements.
Second, a manufacturer may obtain an exemption for ingredients derived
from major allergens by submitting a premarket notification or petition
demonstrating that such ingredient should not be considered a major allergen.9
The premarket notification procedure would be available when (i) scientific
evidence establishes that the food ingredient does not contain allergenic
protein or (ii) FDA has made a determination under a Section 409 premarket
approval or premarket notification program that the ingredient does not cause
an allergic response that poses a risk to human health.10 The Committee
explains that the Generally Recognized as Safe (GRAS) notification process
is not included as part of this exception.
It is difficult to predict how the agency will implement the allergen law,
particularly when it comes to its application to processing aids and other
incidental additives that are derived from major allergens. Ingredients such
as soy lecithin, fish gelatin, and wheat starch have been used for decades as
processing aids and have been exempt from ingredient labeling requirements.
While these ingredients may have detectable levels of protein, they are
frequently used at very low levels in finished foods. In many instances the
level of protein in the finished food contributed by the use of an ingredient
such as soy lecithin can be well below 1 part per million.
It would be reasonable for FDA to take the position that allergenic proteins
present at de minimis, or very low levels, essentially are not present in
finished foods and should not be subject to the allergen labeling requirements.
Indeed, the courts have applied a de minimis concept with regard to the
360 Detecting allergens in food
regulation of food additives that may be present at very low levels in foods.11
At the time of this article, FDA has not offered guidance on whether it would
apply a de minimis analysis and exempt from the allergen labeling requirements
those food ingredients derived from major allergens that contribute insignificant
levels of allergenic protein in finished foods.
Moreover, even if FDA does agree that a de minimis analysis is appropriate,
it remains unclear what level of allergenic protein will be considered a de
minimis level. The absence of an established threshold below which allergens
will not cause an allergic reaction complicates the agency’s ability to establish
a de minimis level. There are factors, however, that would support the
establishment of 10 parts per million or a similar level as the de minimis
level. This is the level identified by some experts as being unlikely to trigger
an allergenic reaction in most food-allergic consumers. It also is the level
that has been used informally by many in the food industry for years in
determining whether an allergen should be declared. In addition, 10 parts per
million is reasonably close to the one to five parts per million analytical level
found in some of the commercially available allergen test kits.
The de minimis concept not only is important when determining whether
a food ingredient should be subject to the allergen labeling requirements, but
also could apply when determining if an ingredient is eligible for the notification
or petition process. The notification process is available in those instances
when a manufacturer can demonstrate that an ingredient derived from a
major allergen, although containing protein, does not contain an allergenic
protein. The law does not comment, however, on what is meant by ‘containing
allergenic protein.’ The law does not comment on whether the agency should
consider the limit of detection of the most sensitive analytical method or the
intended use of the ingredient and the level of allergenic protein that would
theoretically be present in the finished food through such use.
It presumably would be reasonable for the agency to focus on the manner
in which the ingredient is used in the finished food and the theoretical levels
of allergenic proteins that would be present through such use. In instances
when the ingredient would contribute a de minimis level of allergenic protein
under its intended conditions of use, it would be reasonable for the agency
to take the position that the ingredient does not contain allergenic protein
and is eligible for the notification process. If the agency instead focuses on
whether the ingredient contains a detectable level of allergenic protein, there
would be very few ingredients that would be eligible for the notification
program. Under such an interpretation, an ingredient such as soy lecithin
that may contain less than 50 parts per million of total protein would be
ineligible for the notification process.
A petition process is established for those ingredients derived from major
food allergens that are ineligible for the notification program.12 An exemption
would be obtained by submitting a petition with data demonstrating that the
ingredient does not cause an allergic response that poses a risk to human
health. The petitioner would bear the burden of proof. The petition would be
US regulation of undeclared allergens in food products 361
deemed denied after 180 days unless the petitioner and FDA mutually agree
to an extension. FDA is required to post to a public site the notifications and
petitions that have been received and any responses or objections thereto.
Once within the definition of ‘major food allergen,’ ingredients would be
subject to the provisions of the law regardless of whether they had previously
been treated as ‘incidental additives.’ If the agency is unwilling to apply a de
minimis analysis or find some other legal means to interpret the allergen law
flexibly so it is possible to obtain an exemption for processing aids that
contribute insignificant levels of allergenic proteins in finished foods, there
will be many food products that will be subject to the allergen labeling
requirements. Indeed, products containing soy lecithin, fish gelatin, wheat
starch, and other common processing aids would suddenly be subject to
allergen labeling. The food-allergic consumer in many instances has been
consuming these products without incident and will suddenly be presented
with a label disclosing the presence of the major allergen in the food.
In addition to defining major food allergen, the new law amended Section
403 of the FFDCA to provide three new misbranding provisions. The first
provides FDA the authority to require by regulation the appropriate labeling
of any spice, flavoring, coloring, or incidental additive that includes, as a
constituent, a food allergen that is not one of the major food allergens. The
second requires colors, flavors, and incidental additives that contain major
allergens to be subject to the labeling requirements for major allergens. The
third requires the eight major food allergens to be labeled on foods that are
not raw agricultural products.
Manufacturers will have the following two options for identifying the
eight major food allergens on the food label:13
1. By placing the statement ‘Contains ’ with the blank filled in with
the name of each major food allergen, at the end of, or immediately
adjacent to, the ingredient statement; or
2. By using a parenthetical following the name of the ingredient that identifies
the name of the major allergen, such as ‘casein (milk).’ The parenthetical
would not be required when the ingredient name uses the name of the
major food allergen (such as ‘milk’ or ‘soy protein’) or when the name
of the major food allergen appears elsewhere in the ingredient statement
(unless the other listing is for a food ingredient that the Secretary has
determined does not cause an allergenic response).
Regardless of the option selected, the major food allergens would need to be
identified by the name of the food source from which the major allergen is
derived, such as milk, egg, wheat, peanut, or soybean. The specific type of
tree nut (e.g., almonds, pecans, or walnuts), crustacean shellfish (e.g., crab,
lobster, or shrimp) or fish (e.g., bass, flounder, or cod) would also need to be
identified.
The agency has for several years had an allergen labeling policy that
applies to the same ‘major allergens’ that are covered by the allergen labeling
362 Detecting allergens in food
FDA has clarified that it considers products that do not comply with this
policy misbranded. FDA reasons ‘the article was misbranded when introduced
into and while in interstate commerce and is misbranded while held for sale
after shipment in interstate commerce, within the meaning of the section
403(i)(2) of the Act, in that it is fabricated from two or more ingredients, and
its label fails to bear the common or usual name of each such ingredient,
namely (specify the undeclared allergenic ingredient).’16
Since 1996, FDA has stated ‘processing aids that contain allergenic
ingredients must be declared in accordance with 21 CFR 101.4(a)(1).’17
Under this FDA policy, a substance that may be present at very low levels
and have no technical or functional effect on the food would be subject to
ingredient labeling if it contains a protein from a major allergen.
US regulation of undeclared allergens in food products 363
claiming that the restaurant had a duty to warn of the presence of carageenan
in its McLean hamburger.30 McDonald’s filed a summary judgment motion
requesting that the case be dismissed, in part, because the company made
available to consumers upon request brochures that identified the carageenan
in the product. The trial court granted the motion without issuing a decision.
The plaintiff appealed and the appellate court reversed the trial court decision
finding issues of ‘material fact upon which reasonable minds could differ.’
While acknowledging that McDonald’s listed ‘carageenan’ as an ingredient
in a flier that it made available to consumers, the appellate court noted that
the flier ‘does not inform the reader that carageenan is derived from seaweed,
nor that persons who are allergic to seaweed may experience an adverse
reaction to that ingredient.’31 The court also noted that the plaintiff purchased
the sandwich at a drive-through window and there was no evidence as to
whether she received a flyer or that it was offered to her at that time. Under
the applicable Ohio statute, a jury reasonably could find, for instance, that a
manufacturer should have known of a risk to even a small number of consumers.
The statute asks
McDonald’s also claimed that there was no evidence indicating that the
carageenan in its hamburger caused the plaintiff’s reaction. The appellate
court rejected this line of argument noting that the plaintiff had alleged that
the product caused the reaction and that she supplemented this evidence with
a note from a treating physician identifying the hamburger as the cause of
her reactions. Because the plaintiff offered evidence ‘tending to prove causation,
the element remains a question for the jury.’33 Noting that the plaintiff had
raised genuine issues of material fact, the appellate court held that it would
be up to a jury to decide whether McDonald’s should be liable for the
plaintiff’s harm.
The McDonald’s case is recognizably troubling because McDonald’s did
in fact identify the presence of carageenan as an ingredient in flyers made
available to consumers and because carageenan is not known or recognized
as an allergen. McDonald’s likely reached an out-of-court settlement after
losing the appeal. Had the case gone to trial, McDonald’s then could have
presented evidence establishing that (i) its flyer constituted adequate warning
and (ii) that its hamburger did not cause the plaintiff’s injury. It would have
been up to the judge or jury, however, to decide these issues of fact.
US regulation of undeclared allergens in food products 369
20.3.3 Damages
The failure to provide adequate warnings to allergens exposes the food and
restaurant industries to potential product liability lawsuits. Plaintiffs that
have suffered injury from consuming the allergen may decide to file lawsuits
seeking damages against the food manufacturer or restaurant. Damage awards
can include both compensatory damages and punitive damages. Compensatory
damages are intended to compensate the consumer for injuries, which may
include medical expenses and the amorphous ‘pain and suffering.’
A food company or restaurant can also be liable for punitive damages if
its failure to warn is judged to be the result of what is known as ‘actual
malice’ or a ‘reckless disregard’ for the safety of customers. Although the
definitions of ‘actual malice’ and ‘reckless disregard’ vary by jurisdiction,
the general concept describes a situation where the defendant was aware of
the dangers posed by his conduct but went ahead anyway. For example,
punitive damages might be imposed where a server had serious doubts about
the truth of his answer when he told a customer a dish did not contain a
particular ingredient, but he chose to ignore those doubts, thereby endangering
the customer. Punitive damages also could be awarded, for example, if a
company becomes aware that it failed to include eggs in the ingredient
statement, but fails to recall the product. Although no cases could be found
when a company has been held to pay punitive damages, these are the types
of activities that in some jurisdictions may expose a company to a punitive
damages award. Importantly, punitive damages could end up being many
times the amount of actual damages sustained.
Other damages that should not be overlooked are the adverse publicity,
the lost business that may result from a lawsuit, and the legal fees associated
with defending the action. It is the potential for adverse publicity that likely
is responsible for the large number of cases that are settled out of court. With
regard to lost business, in some instances the loss of business has caused
companies to go out of business.
to maintain the confidentiality of the test kit results by having the analysis
performed by an in-house or outside lawyer. A company, therefore, should
assume that the allergen analytical results would need to be made available
to a plaintiff that is suing the company for injuries resulting from the alleged
presence of undeclared allergens in the food.
Allergen test kits provide the industry with a useful tool in controlling the
presence of undeclared allergens. Careful thought and consideration, however,
must be given to how the company will incorporate the test kits into its
overall allergen management program.
bar of the potential for a new and potentially lucrative cause of action to be
brought against the food industry. Because test kits are available to the
general public, savvy plaintiff’s lawyers could begin testing products for the
presence of undeclared allergens and then attempt to bring class action lawsuits
against companies marketing products with undeclared allergens. The number
of product liability actions brought against the food industry for undeclared
allergens may very well increase, unless the industry is able to implement
procedures to reduce the release into commerce of products with undeclared
allergens.
20.5 Conclusion
The presence of undeclared allergens in the food supply is proving to be an
issue that subjects the food industry to potential product liability. Restaurants
can reduce their potential liability by educating their waiting staff of the
importance of providing consumers with accurate information about products.
Restaurants also should consider ways in which they can inform the food-
allergic community of the presence of allergens in products where they
would not ordinarily be expected.
Manufacturers of finished food can also take numerous steps to reduce the
potential regulatory and product liability. The labeling of products with
ingredient statements that identify allergens is the best way to minimize
product liability. Although there are no magic formulas that will guarantee
immunity from liability for an allergic reaction or sensitivity to food, there
are numerous steps that can be taken to minimize potential liability.
20.7 References
1. 21 United States Code (U.S.C.) § 331(a); FFDCA 301(a).
2. 21 U.S.C. § 343(i)(1); FFDCA § 403(i)(1).
3. 21 U.S.C. § 343(i)(1); FFDCA § 403(i)(1).
4. 21 U.S.C. § 101.100(a)(3).
5. 21 C.F.R. § 101.100(a)(3).
6. Food Allergen Labeling and Consumer Protection Act, Pub.L. No. 108–282, 118
Stat. 905 (August 2, 2004).
7. 21 U.S.C. § 321(gg); FFDCA § 201(gg).
8. 21 U.S.C. § 321(gg)(2)(A); FFDCA § 201(gg)(2)(A).
374 Detecting allergens in food
Appendix I: Continued
Appendix II: Summary of cases involving ingredients that are not allergens
21.1 Introduction
The European Union promotes the free movement of goods between Member
States through the EC Treaty, which prohibits quantitative restrictions on
imports between them (Article 28). This principle of free movement was
affirmed in the Cassis de Dijon case.1
In the late 1990s, the BSE crisis led to major changes in the regulatory
regime for foodstuffs. The White Paper on Food Safety, presented by the
Commission, stated that ‘assuring that the EU has the highest standards of
food safety is a key policy priority for the Commission,’ and that ‘a radical
new approach is proposed.’ The White Paper recommended the establishment
of an independent European Food Authority to address the need to guarantee
a high level of food safety. A range of other measures aimed at improving
and standardising legislation covering all aspects of food products, from
‘farm to table’, is needed to support this recommendation.
The supporting measures proposed by the Commission are presented in
the Annex, with an indication of priority and likely timing. The ‘Action Plan
on Food Safety’ (Action Number 3) proposes a General Food Law Directive.
The objectives are to establish food safety as the primary objective of EU
food law and to lay down the common principles underlying food legislation,
in particular the scientific basis for legislation, responsibility of producers
and suppliers, traceability along the food chain, efficient controls and effective
enforcement. Action Number 16 concerns a proposal to amend the existing
directive on the labelling, presentation and advertising of foodstuffs. The
objectives of this action are to ensure that the components of compound
ingredients forming less than 25% of the final product are indicated, and to
EU regulation of undeclared allergens in food products 379
21.2.1 Definitions
Labelling shall mean any words, particulars, trade marks, brand name, pictorial
matter or symbol relating to a foodstuff and placed on any packaging, document,
notice, label, ring or collar accompanying or referring to such foodstuff. Pre-
packaged foodstuffs are those where the contents cannot be altered without
opening or changing the packaging.
380 Detecting allergens in food
21.2.4 Name
The name under which a foodstuff is sold shall be the name provided for in
the Community provisions applicable to it or, where there are no Community
provisions, the name provided for in the laws, regulations and administrative
provisions applicable in the Member State in which the product is sold.
Failing this, the name or description of the foodstuff and, if necessary, its use
must be clear enough to let the purchaser know its true nature and distinguish
it from other products.
In general, the list of ingredients shall include all the ingredients of the
foodstuff, in descending order of weight, as recorded at the time of their use
in the manufacture of the foodstuff. Exceptions include added water and
volatile products, ingredients used in concentrated or dehydrated form,
concentrated or dehydrated foods, mixtures of spices or herbs, and ingredients
constituting less than 2% of the finished product (may be listed in a different
order after the other ingredients). Ingredients may be listed by their specific
name, or by category name where applicable (see Annex I of the Directive).
Additives that belong to one of the categories listed in Annex II of the
Directive must be designated by the name of that category, followed by their
specific name or EC number. Flavourings shall be designated in accordance
with the rules laid down in Annex III of the Directive. There are some cases
where ingredients do not need to be listed, for instance single-ingredient
foodstuffs such as fresh fruit and vegetables, carbonated water, fermentation
vinegars, cheese, butter, fermented milk and cream.
the specified period. The date of minimum durability does not apply to all
foodstuffs. In the case of highly perishable foodstuffs, which are likely to
constitute an immediate danger to human health after a short period, the date
of minimum durability is replaced by the ‘use by’ date.
21.2.11 Language
The information on the label should be written in a language easily understood
by the consumer, unless the consumer is informed by other means. Member
States may specify the language(s) used to label products marketed within
their own territory, in accordance with the rules of the Treaty.
21.3.1 Considerations
Allergic or intolerance reactions constitute a danger to the health of the
consumer. The Scientific Committee on Food has stated that food allergies
affect the lives of many people, and reactions range from very mild to potentially
fatal. Common food allergens include cows’ milk, fruits, legumes, especially
peanuts and soybeans, eggs, crustaceans, tree nuts, fish, vegetables (celery
and other foods of the Umbelliferae family), wheat and other cereals. The
most common food allergens are found in a wide variety of processed foods,
EU regulation of undeclared allergens in food products 383
so labelling rules are necessary to ensure that consumers suffering from food
allergies receive appropriate information.
A list of allergenic substances has been drafted, and includes those foodstuffs,
ingredients and other substances recognised as causing hypersensitivity. The
list of allergens may be revised if new allergens are identified, or if technological
developments permit the removal of allergenicity in ingredients and other
substances, in order to avoid unnecessary obligations on labelling.
Table 21.1 List of food ingredients and substances provisionally excluded from Annex
IIIa of Directive 2000/13/EC
finished product, even in altered form, must be indicated on the label with a
clear reference to the name of the ingredient. This means that any allergenic
ingredient used in production and present in the foodstuff has to be indicated
on the label even if a list of ingredients is not needed. This requirement also
applies to the ingredients of a compound ingredient, food additives, a component
of a food additive, flavourings, a component of a flavouring, processing aids,
and carriers, solvents or media for additives, flavours or processing aids.
Indication is not required if the name under which the foodstuff is sold
clearly refers to the ingredient concerned.
21.4.1 Considerations
Where Member States have different legislation on product safety, this not
only affects the levels of protection provided to consumers, but also tends to
create barriers to trade and distortion of competition within the European
Community market. The Community aims to protect the health and safety of
consumers, and Community-wide legislation introducing general product
safety requirements and containing provisions on the general obligations of
producers and distributors should contribute to that aim. Because it is very
difficult to adopt Community-wide legislation for every product, there is a
need for a broad-based, legislative framework that establishes general safety
requirements for any product.
EU regulation of undeclared allergens in food products 387
21.5.1 Considerations
The free movement of safe and wholesome food can be achieved only if food
and feed safety requirements do not differ significantly from Member State
to Member State. Experience has shown that measures are needed to guarantee
that unsafe food is not placed on the market, and that systems exist to
identify and respond to food safety problems. To ensure the safety of food,
all aspects of the food production chain must be considered, from and including
primary production and the production of animal feed, up to and including
sale or supply of food to the consumer, because each element may have a
potential impact on food safety. Community policies aim to ensure a high
level of protection of human life and health, and Community food policies
form a common basis for measures governing food and feed taken by individual
Member States and at Community level.
EU regulation of undeclared allergens in food products 389
21.5.5 Traceability
Food and any other substance intended to be, or expected to be, incorporated
into a food shall be traceable through all stages of production, processing
and distribution. Traceability is of great importance for product liability. In
the case a defective product, the plaintiff can make each producer in the
chain liable, from raw material to end-product. Producers therefore need to
have a traceability system in place that enables them to check whether the
cause of a defective product lies in their own factory, is due to a defective
raw material, or arose later with a user in the chain. In the latter two cases the
producer can take recourse against the supplier or user. Food which is placed
on the market or is likely to be placed on the market in the Community shall
be adequately labelled or identified to facilitate its traceability, through relevant
documentation or information.
390 Detecting allergens in food
21.5.7 Liability
The provisions stated in Chapter 2 of the Regulation (EC) No 2002/178/EC
concerning General Requirements of Food Law, Food Safety Requirements,
Responsibilities and Traceability, shall be without prejudice to Directive 85/
374/EEC18 concerning liability for defective products. This means that the
rules concerning Food Safety Requirements, Responsibilities and Traceability,
and those concerning defective products, exist side by side.
21.6.1 Considerations
Different laws among Member States concerning the liability of the producer
for damage caused by a defective product may distort competition and affect
the free movement of goods. National laws also result in differing levels of
consumer protection against damage to health or property caused by a defective
product. Standardisation of laws across the Community is not and cannot be
total, but this Directive lays down the principle of strict liability for the
producer. Even in the absence of fault, the producer may be held responsible for
damage caused by a defective product that the producer has put on the market.
EU regulation of undeclared allergens in food products 391
21.6.4 Producer
On the basis of Article 3, ‘Producer’ means the manufacturer of a finished
product, the producer of any raw material or the manufacturer of a component
part and any person who, by putting his name, trademark or other distinguishing
feature on the product presents himself as its producer. In addition, any
person who imports a product into the Community for sale, hire, lease or any
form of distribution in the course of his business shall be deemed to be a
producer within the meaning of the Directive and shall be responsible as a
producer. The producers are liable jointly and severally, and the victim of
damage may make a claim against any of these persons for compensation,
whether they are the actual producer or not. If the producer of the product
cannot be identified, each supplier of the product shall be treated as its
producer unless he informs the injured person, within a reasonable time, of
the identity of the producer or of the person who supplied him with the
product.
392 Detecting allergens in food
21.6.5 Damage
The scope of the damage to be compensated is restricted according to Article
9, to include damage caused by death or by personal injuries, and damage to,
or destruction of, any item of property other than the defective product itself,
with a lower threshold of 500 ECU (Euro 500). The item of property should
be of a type ordinarily intended for private use or consumption. This directive
does not cover damage to professional property, where national laws concerning
liability are applicable. National laws are also needed for damage that is not
covered by the scope of product liability, such as the lower threshold of Euro
500. As described above, damage resulting from death or personal injury and
caused by identical items with the same defect can be limited to an amount
not less than 70 million ECU (Euro 70 million).
times during the production and distribution process. It is only when the
commodity is in circulation (and if it is defective) that the producer will be
liable. During storage and processing the product is deemed not to be in
circulation and, based on the legislation concerning product liability, the
producer cannot be liable. However, producer liability is possible based on
other legislation such as, for example, labour laws. Processing aids are not
deemed to have been put into circulation if they are used for the preparation
of foodstuffs, but only if they have been passed along the chain.
The expectations of the consumer play an important part in determining
whether a product is defective or not. Hence other circumstances under
which a product is considered defective relate to the nature of the product
(for instance, a medicine has a different nature to a foodstuff), the advantages
and the usefulness of the product, the chance or frequency of appearance of
the danger, the appearance of the harmful additional workings, and the
knowableness of the danger. A product is not considered defective if there is
a mere lack of conformity to specifications, stemming from regulations or
from a contract, and the safety of the product is not at stake.
The cause of a product defect can be reduced to faults in production,
where there are three kinds of faults.
• Design defects. This kind of fault leads to categorical defects, and therefore
to a series of defective products. The fault only can be removed by a new
or changed design.
• Manufacturing faults. This kind of fault occurs during production, and
results in one or more products which deviate from other products made
according to the same specifications. Producers have a duty to check
products against specifications and ensure that defective products are not
put into circulation.
• Inadequate instructions or warnings. If the instructions or warnings
included in the advertisement of the product, on the label, in the instructions
and on the directions for use do not give any or enough information about
the safe use of the product, then they are inadequate.
The appearance of one of these faults does not mean that the product is
defective, unless the safety of the product is at stake.
Products may become unsafe after they are put into circulation, which is
not covered by this Directive. If a product becomes defective after being put
into circulation, the claim for damages must comply with national rules
based on fault. The producer has a duty of care as laid down in Directive
2001/95/EC21, and must monitor products accordingly, warning consumers
if the product, or its use, becomes unsafe.
very difficult for the plaintiff to prove, and the Directive does not allow a
conversion of the burden of proof. But it is within the scope of the Directive
for a judge to charge the producer with contradicting the thesis of the plaintiff,
if the plaintiff has provided a sufficient part of the proof.
Directive 2001/95/EC33
Product definitions
Foodstuffs with one or more allergenic ingredients are products for the purpose
of this directive, under the definitions listed in Section 21.4.2. A foodstuff
containing one or more allergenic ingredients which are not indicated on the
label is not a safe product under the definitions listed in Section 21.4.2.
396 Detecting allergens in food
Product recall
Producers of foodstuffs have an obligation to monitor their products and to
recall products from the market if they present risks (Section 21.4.5). If an
allergenic ingredient is not indicated on the label of a foodstuff, its presence
forms a risk for some consumers and hence the product is dangerous. Once
the producer is aware of this risk, the foodstuff must be withdrawn from the
market. Producers and distributors are also obliged to recall dangerous products
from consumers. In some cases, other appropriate actions may include
adequately and effectively warning consumers. Governments can oblige
producers and distributors to recall a product, from the market and/or consumers.
Regulation 178/2002/EC34
Definition of food
Foodstuffs with one or more allergenic ingredients belong to food under the
definition of food for this regulation (Section 21.5.2).
Traceability
Producers of all foodstuffs, with and without allergenic ingredients, indicated
on the label or not, must meet traceability requirements as listed in Section
21.5.5
Directive 85/374/EEC35
Requirements for liability
The producer shall be liable for damage caused by a defect in his product
under the requirements described in Section 21.6.3.
Producer
The definition ‘Producer’ as described in Section 21.6.4 applies to the producer
of a foodstuff containing an allergenic ingredient that is not indicated on the
label.
Product
Foodstuffs with and without allergenic ingredients are products in the sentence
of this directive (Section 21.6.6).
Damage
A plaintiff can claim damage as described in Section 21.6.5.
Defective product
A foodstuff containing an allergenic ingredient is a defective product if the
allergenic ingredient is not indicated on the label, because the allergenic
ingredient presents a risk to some consumers (Section 21.6.6). A product
containing an allergenic ingredient may also be defective if there are faults
during production, such as defects in the design of the product and the label.
If the instructions or warning concerning the indication on the label do not
give enough or any information about one or more allergenic ingredients,
then the warnings or instructions are inadequate.
Causal relationship
The plaintiff has to prove the damage, that the product is defective, and the
causal relationship between the defect and the damage (Section 21.6.7). The
398 Detecting allergens in food
proof of the damage and the defect will be relatively straightforward. The
causal relationship will be more difficult to prove, because it will be possible
that the plaintiff will have consumed other foodstuffs containing the same
allergenic ingredients.
Means of defence
There are a number of means of defence for a producer to prove that he is not
liable (Section 21.6.8). But, in the case of foodstuffs containing allergenic
ingredients, the defect which caused the damage will have existed at the time
when the product was put into circulation. In addition, it will be clear that the
product was manufactured for sale and distribution by the producer, that the
defect is not due to compliance with mandatory regulations issued by the
public authorities, that the state of scientific and technical knowledge at the
time when the product was put into circulation made it possible to discover
the existence of the defect, and that the producer is not the manufacturer of
a component. The defect is therefore attributable to the design of the product
made by the producer.
Limitation period
The limitation period of three years applies in the case of foodstuffs containing
allergens (Section 21.6.9).
Period of expiry
The 10-year period of expiry applies in the case of foodstuffs containing
allergens (Section 21.6.10).
Misleading information
Omitting to indicate the presence of allergenic ingredients could be qualified
as misleading because it concerns the characteristics of the foodstuff (Section
21.2.2).
List of ingredients
Ingredients must be indicated under the requirements to list ingredients (Section
21.2.5). In addition to this, allergic ingredients which are listed in the Annex
to Directive 2003/89/EC must be indicated in a prescribed manner on the
label (Sections 21.2.5 and 21.3.2).
QUID regulation
The QUID regulation is relevant if the quantity of an allergenic ingredient is
such that it must be stated (Section 21.2.6).
Net quantity
The requirements concerning the net quantity are not relevant for this subject
(Section 21.2.7)
400 Detecting allergens in food
• CIAA
Confédération des Industries Agro-Alimentaires de l’EU
http://www.ciaa.be/
• European Community
http://europa.eu.int/eur-lex/nl/index.html
• FNLI
Federatie Nederlandse Levensmiddelen Industrie
http://www.fnli.nl/
• TNO Quality of Life
http://www.voeding.tno.nl/
• VAI
Nederlandse Voedingsmiddelen Industrie
http://www.vai-voeding.nl/
• Europe
— Labelling
http://europa.eu.int/comm/food/food/labellingnutrition/foodlabelling/
index_en.htm
— Consumer affairs/product safety/product liability
http://europa.eu.int/comm/consumers/index_en.htm
— Public health
http://europa.eu.int/comm/health/horiz_publications_en.htm
— European Food Safety Authority (EFSA)
http://www.efsa.eu.int/index_en.html
21.10 References
1. Rewe-Central A G vs. BundesmonopolVerwaltung fúr Branntwein (Cassis de Dijon)
(1979) ECR 469.
2. OJEC 2002 L31/1
3. OJEC 2000 L109/29
4. OJEC 2003 L308/15
5. OJEC 1997 L043/1
6. OJEC 1985 L210/29
7. OJEC 1992 L228/24
8. OJEC 1999 L141/20
9. OJEC 2001 L011/4
10. OJEC 2000 L109/29
11. OJEC 1979 L33/1
12. OJEC 2003 L308/15
13. OJEC 2000 L109/29
14. OJEC 2005 L75/33
15. OJ 2001 L11/4
16. OJ 1992 L228/0024
17. OJ 2002 L031/001
18. OJ 1985 L210/29
19. OJ 1985 L210/29
20. OJEC 1999 L141/20
21. OJ 2001 L11/4
22. OJ 2001 L11/4
23. OJEC 1992 L228/24
404 Detecting allergens in food
Conclusions
S. Hefle, University of Nebraska, USA and S. Koppelman,
AllerTeQ and University Medical Centre Utrecht, The Netherlands
Such validation studies have been undertaken recently for some allergen
detection kits and several publications became available in 2005:
• A validation trial designed by the U.S. Food and Drug Administration
(Park et al., 2005) utilized three laboratories in a performance validation
of three commercial peanut ELISA kits at 0 and 5 ppm to be used for
screening purposes using peanut butter spiked into breakfast cereal, cookies,
ice cream, and milk chocolate spiked at 0 and 5 ppm peanut. They found
when two validated kits are used in parallel, the probability of detecting
5 ppm is >95% peanut protein in four different food matrixes.
• Another interlaboratory trial of five commercial ELISA kits for peanut in
biscuits (cookies) and dark chocolate model foods containing peanut at 0–
10 ppm was published by Poms et al. (2005). They found that, in general,
these five quantitative kits performed better in the 5–10 ppm range than at
lower ranges of 2–2.5 ppm, but in some cases, 2 ppm was below the
official detection limit of the kit, so the authors were pushing the technology
in some cases beyond what the kit was designed to do. Impressively, 34
laboratories participated in this trial, and results were obtained from 12–
14 laboratories for each commercial kit. For the biscuits (cookies), peanut
flour was mixed and then baked in household-size portions and equipment.
For dark chocolate, peanut-free chocolate was melted at 60 °C and then
peanut flour was mixed in. Average recoveries varied with all types of test
kits and matrices, and both the type of kit and the matrix seemed to be
important for recovery.
• More work has been performed on interlaboratory validations for allergen
detection methods. Akiyama et al. (2005) described two interlaboratory
trials on peanut and buckwheat ELISAs: extracts of biscuit, sauce, chocolate
and butter spiked with peanut standard protein at a level of 5–20 ng/mL
were found to be reproducible in the study. Mean recoveries of the peanut
standard protein from the food extracts were over 40% using two ELISA
methods. The detection limits of both ELISA methods were 2–2.5 ng/mL
in sample solutions. Inter-laboratory evaluation studies were conducted
for the ELISA methods for buckwheat. Extracts of snacks, buns and udon
noodles spiked with buckwheat protein at a level of 5–20 ng/mL as sample
solutions were analyzed in replicate at 10 laboratories. Mean recoveries
of the buckwheat standard protein from the food extracts were over 40%
in the two ELISA methods. The detection limits of both ELISA buckwheat
methods were 1 ng/mL.
With the data of the validation studies available, we believe that the detection
and quantification of peanut residue in food products, as desired by the food
industry and food regulators, is possible for the majority of food products.
One should, however, always be aware of specific situations that may disturb
proper analysis. Recovery from the food matrix may be the most important
issue here, and some food matrices are particularly notorious. Chocolate, for
example, is a difficult matrix to extract peanut residue from. Akkerdaas et al.
Conclusions 407
22.5 Summary
The unintended presence of allergenic residue in foods is, and will be in the
future, an issue that affects the food allergic consumer, and thereby the food
industry. Data on threshold levels of an allergen to elicit an allergic reaction
in food-allergic patients are available for a selected number of allergens, and
based on these threshold levels, a sensible detection limit for food allergen
detection assays can be set. We propose that this detection limit should be
between 1 and 10 ppm, providing that the recovery of the analytical procedure
is at least 50% for the food products where the test is applied, and preferably
higher. Since the early 1990s many methods have been published to detect
allergenic residue in food and food ingredients. Some assays have been
validated in multi-laboratory trials (peanut) and have been shown to be useful
for assessing the amount of allergenic residue in foods. For some allergens
Conclusions 411
other than peanut such validation studies are in progress, and for other allergens,
methods need to optimized before validation studies can be considered.
Trace contamination can arise from practices in the food industry, restaurants,
and in homes and schools. Food-service employees and food processors
alike must be very vigilant about the use of shared utensils and equipment.
Most of the deaths from inadvertent exposure to allergenic foods have occurred
in food-service situations (restaurants, schools, homes). However, in many
countries, food-service establishments do not have specific regulations about
handling or labeling allergens. The increased demand among the general
consuming public for a wide variety of food products, and the ever-increasing
consumption of food prepared outside the home, can assist in producing
reactions to hidden food allergens. While the majority of reactions occur
after ingestion of a food that is prepared in a food-service environment rather
than packaged food products, the food processing industry has been subjected
to increasing scrutiny of its allergen controls. The resulting impact has been
remarkable: the food industry has made significant investment, effort and
improvements in allergen control in recent years. Food processors often use
shared equipment and facilities for the production of processed foods with a
variety of formulations. This is often out of necessity rather than just fueled
by economic reasons. Due to various limitations and practices in food
processing, foods may occasionally contain trace residues of other foods that
may not be declared on the ingredient label. The food industry began investing
a significant amount of time, money, and effort to attain better allergen
control in manufacturing facilities in the late 1980s. Since the late 1990s, the
commercialization of some test methods for the detection of some allergenic
foods has allowed food manufacturers to use these tests to create important
data to assess and design allergen control strategies and protocols.
Debate and discussion about the effects of processing on allergenicity and
allergenic residue detection in foods will be ongoing. In addressing new
labeling legislation in the EU and the US, analytical methods are being used
to assess allergenicity of some ingredients. However, analysis of some allergen-
derived ingredients can pose serious challenges. Problematic ingredients
include oils and lecithin due to their oily nature, and also hydrolyzed proteins
and residues of allergenic growth substrates in gums, starter cultures, enzymes,
etc. Standard ELISAs cannot be used in the case of hydrolyzed/proteolyzed/
fermented ingredients and PCR cannot be used to assess how allergenic the
ingredient is. Methods involving IgE obtained from individuals allergic to
the particular food are the only way to determine allergenicity of these types
of ingredients with the exception of oral challenge studies in allergic individuals.
Although there is much work to do to deal with the issue of detecting
allergenic residues in food, we believe that the enormous output of many
investigators and test kit manufacturers, and the use of test kits by the food
industry, has already succeeded in providing better protection for the food-
allergic consumer. This is a solid basis for the further development of tools
for the detection of allergenic residues.
412 Detecting allergens in food
22.6 References
Akkerdaas, J H, Wensing, M, Knulst, A C, Stephan, O, Hefle, S L, Aalberse, R C, van
Ree, R (2004) ‘A novel approach for the detection of potentially hazardous pepsin
stable hazelnut proteins as contaminants in chocolate-based food’, J Agric Food Chem
52, 7726–7731.
Aranishi, F, Okimoto, T (2004) ‘PCR-based detection of allergenic mackerel ingredients
in seafood’, J Genetics 83, 193–195.
Akiyama, H, Nakamura, K, Harikai, N, Watanabe, H, Iijima, K, Yamakawa, H, Mizuguchi,
Y, Yoshikawa, R, Yamamoto, M, Sato, H, Watai, M, Arakawa, F, Ogasawara, T, Nishihara,
R, Kato, H, Yamauchi, A, Takahata, Y, Morimatsu, F, Mamegoshi, S, Muraoka, S,
Honjoh, T, Watanabe, T, Sakata, K, Imamura, T, Toyoda, M, Matsuda, R, Maitani, T
(2004) ‘Inter-laboratory evaluation studies for establishment of notified ELISA methods
for allergic substances (peanuts)’, (in Japanese) Shokuhin Eiseigaku Zasshi 45, 325–
331.
Akiyama, H, Nakamura, K, Harikai, N, Watanabe, H, Iijima, K, Yamakawa, H, Mizuguchi,
Y, Yoshikawa, R, Yamamoto, M, Sato, H, Watai, M, Arakawa, F, Ogasawara, T, Nishihara,
R, Kato, H, Yamauchi, A, Takahata, Y, Morimatsu, F, Mamegoshi, S, Muraoka, S,
Honjoh, T, Watanabe, T, Sakata, K, Imamura, T, Toyoda, M, Matsuda, R, Maitani, T
(2004) ‘Inter-laboratory evaluation studies for establishment of notified ELISA methods
for allergic substances (buckwheat)’, (in Japanese) Shokuhin Eiseigaku Zasshi 45,
313–318.
Ben Rejeb, S, Abbott, M, Davies, D, Cleroux, C, Delahaut, P (2005) ‘Multi-allergen
screening immunoassay for the detection of protein markers of peanut and four tree
nuts in chocolate’, Food Addit Contam 22, 709–715.
Brzezinski, J L (2005) ‘Detection of crustacean DNA and species identification using a
PCR-restriction fragment length polymorphism method’, J Food Prot 68, 1866–1873.
Hirao, T, Imai, S, Sawada, H, Shiomi, N, Hachimura, S, Kato, H (2005) ‘PCR method for
detecting trace amounts of buckwheat (Fagopyrum spp.) in food’, Biosc Biotechnol
Biochem 69, 724–731.
Holden, L, Faeste, C K, Egaas, E (2005) ‘Quantitative sandwich ELISA for the determination
of lupine (Lupinus spp.) in foods’, J Agric Food Chem 53, 5866–5871.
Park, D L, Coates, S, Brewer, V A, Garber, E A, Abouzied, M, Johnson, K, Ritter, B,
McKenzie, D (2005) ‘Performance Tested Method multiple laboratory validation study
of ELISA-based assays for the detection of peanuts in food’, J AOAC Int 88, 156–160.
Poms, R E, Agazzi, M E, Bau, A, Nrohee, M, Capelletti, C, Norgaard, J V, and Anklam,
E (2005) ‘Inter-laboratory validation study of five commercial ELISA test kits for the
determination of peanut proteins in biscuits and dark chocolate’, Food Addit Contam
22, 104–112.
Sforza, S, Scaravelli, E, Corradini, R, Marchelli, R (2005) ‘Unconventional method based
on circular dichroism to detect peanut DNA in food by means of a PNA probe and a
cyanine dye’, Chirality 17, 515–521.
Stephan, O, Weisz, N, Vieths, S, Weiser, T, Rabe, B, Vatterott, W (2004) ‘Protein
quantification, sandwich ELISA, and real-time PCR used to monitor industrial cleaning
procedures for contamination with peanut and celery allergens’, J AOAC Int 87, 1448–
1457.
Zuidmeer, L, van Leeuwen, W A, Budde, I K, Cornelissen, J, Bulder, I, Rafalska, I,
Besoli, N T, Akkerdaas, J H, Asero, R, Rivas, M F, Mancebo, E G, van Ree, R (2005)
‘Lipid transfer proteins from fruit: cloning, expression and quantification’, Int Arch
Allergy Immunol 137, 273–281.
Index
A
adjuvants 69
adulteration regulations 363
advertising and promotion of foods 319
agricultural biotechnology 153
albumins 41 221 247
alpha-lactalbumin 40 221
bovine serum albumin 221
ovalbumin 43 227
parvalbumins 38 284
serum albumins 41
2S albumins 27
allergen labelling directive 267 382 398 401
allergen management 318
Allergen Nomenclature Subcommittee 44
allergosorbents 87
almond detection 210
alpha-amylase and protease inhibitors 29
alpha-caseins 41
alpha-lactalbumin 40 221
analytes 325 353
anaphylactoid reactions 5
anaphylaxis 150 152
This page has been reformatted by Knovel to provide easier navigation. 413
414
Index terms Links
animal derived allergens 37
ATP: guanido phosphotransferases 39
calcium-binding EF-hand proteins 38
caseins 41 221 224
glycoside hydrolases 40
immunoglobulins 41
Kazal-type serine protease inhibitors 42
lipocalins 40
serum albumins 41
transferrins 42
tropomyosins 14 37 39 172
282 283
annealing process 126 132
anti-food allergen antibody specificity 300
anti-food protein antisera 299
antibodies 65 110 176
antigen-antibody interactions 66
see also cross-reactions
classes of 65 112
engineered antibodies 72
IgA class 9
IgE class see IgE antibodies
IgG class 65 71
IgM class 71
immobilization 161
immunogen tolerance 72
immunoglobin structural model 65 110
monoclonal antibodies 67 73 75 257
one-step tests 176
polyclonal antibodies 67 75 257
production of 66 68
recognition of antigens 68
second antibodies 68
B
baker’s asthma 149 244
beef allergy 38 41
C
calcium-binding EF-hand proteins 38
carageenan 368
caseins 41 221 224
cashew nut detection 211
CATH protein structure classification 45
celery detection 133
cell-mediated reactions 4 7 10
cereals
alpha-amylase and protease inhibitors 29
and coeliac disease 10 30 244 247
265
oats 247 262
PCR detection 133
prolamins 26 29
D
dairy and egg residues 219
bioactivity of allergens 228
biosensors 235
blotting transfer 231
commercial tests 235
diffusion-in-gel detection 228
E
EAST inhibition 228
edible oils 13
F
fast assays 344
fish 283
allergens 14 37 284
collagen 14
detection methods 285
gelatin 14
monospecificity of allergies 284
prevalence of allergies 283
fluorescent dyes 146 149
food allergies, definition 4
food idiosyncrasies 5
food intolerances 4 5
food processing see processed foods
food safety 388 395
definition of food 389
product recalls 357 364 388 395
396
responsibilities of business operators 389 390
traceability requirements 389 397
unsafe foods 389
see also product safety
food sensitivity 4
G
Gee, Samuel 245
gelatin 14
H
hazard characterisation 317
hazard identification 316
hazelnuts
in chocolate products 206
detection 206
proteomics 150
PCR analysis 133 210
surface plasmon immunoassay (SPR) 166 168 169
threshold levels 203
heat treatments
and egg proteins 227
and impaired extraction efficiency 255
and milk 223
and nut and seed allergenicity 204
and peanuts 339
and wheat gluten 250
Hevein-like chitinases 33
histamine 5
hydrogen bonding 67
hydrolysates 14 293 294
hydrolyzed infant formulae 221
I
IEF power supplies 145
IFA (incomplete Freund’s adjuvant) 70
IgA antibodies 9
IgE antibodies 8 9 79 88
K
Kazal-type serine protease inhibitors 42
kiwi fruit 31
Kunitz-type protease inhibitors 36 278
M
McDonald’s 367
MALDI analysis 147 263
manufacturing controls 297 310 319
marker compounds 275 322 348
mass spectrometry analyses 147 194 263
matrix interference 326
metabolic food disorders 5
method validation 350 352
microfluidic devices 155
milk allergies see cows’ milk allergies
minimum eliciting doses 11
misbranding regulations 358
misleading information 380
mono-specific antibody reagents 101
monoclonal antibodies 67 73 75 257
mustard detection 212
O
oats 247 262
one-step tests 176
optical density (OD) 335 341
oral allergy syndrome (OAS) 6 35 150
organoleptic techniques 109
osteopenia 246
ovalbumin 43 227
ovine and bovine milk cross-reactions 224
ovomucoids 42 166 225
Q
quality assurance 349
dairy and egg residues 236
hypoallergenic infant formulas (HIF) 293
IgE antibody-based in vitro assays 91
IgE antibody-based in vivo assays 83
quality control materials (QCM) 351 353
see also effectiveness of allergen detection
quantitative ingredients declaration (QUID) 381 399
quantitative tests 145 235
R
random errors 331
rapid screening tests 266
RAST (radioallergosorbent test) 85 88 91 187
188
dairy and egg residues 228
strengths 94
weaknesses 95
see also IgE antibody-based in vitro assays
reaginic factor 7
real-time PCR 129
recognition of antigens 68
reference materials 81 351
reference matrix 355
regeneration solutions 161
regulations and standards 89 303 326
gluten-free foods 248 267
S
SAFE EU project 44
samples
accuracy of known samples 342
extraction procedures 160 166 298 337
integrity 303
serum samples 71
spiking 163 342
sandwich ELISA 112 114 331
sandwich surface plasmon immunoassay (SPR) 162 168
SCOP database 45
SDS-PAGE 98 144 259
seafood see crustaceans; fish
second antibodies 68
seed allergies see nut and seed allergies
sensitivity targets see threshold levels
separation methods 144 145
serpins 42
serum albumins 41
serum samples 71
T
T-cells 9 68 248 265
T lymphocytes 10
tandem mass spectrometers 147 194
targeted proteome method 149
test characteristics 335
test performance 332
test times 114 176 332
thaumatin-like proteins (TLPs) 33
U
Unilever 319
United States regulations 357
adulteration provisions 363
de minimis concept 359
misbranding provisions 358
product recalls 357 364
unsafe foods 389
W
walnut detection 211
Western blotting see immunoblotting
wheat gluten 26 244 340
choice of detection method 264
and coeliac disease 10 30 244 247
265
electrophoretic separations 259
ELISA detection 250
gluten-free food standards 248 260
and heat processing 250
Maldi-TOF detection 263
PCR detection 262
proteins in 244 246
and proteomics 149
rapid screening tests 266
threshold values 249
Win-like chitinases 33
Y
yolk protein 227