You are on page 1of 6

Molecular Immunology 100 (2018) 107–112

Contents lists available at ScienceDirect

Molecular Immunology
journal homepage: www.elsevier.com/locate/molimm

Meat allergy and allergens T



Jeffrey M. Wilson, Thomas A.E. Platts-Mills
Division of Allergy & Immunology, University of Virginia, Charlottesville, VA, USA

A R T I C LE I N FO A B S T R A C T

Keywords: IgE-mediated hypersensitivity to ingested animal products, including both mammalian and avian sources, is
Meat allergy increasingly appreciated as an important form of food allergy. Traditionally described largely in children, it is
Alpha-Gal now clear that allergy to meat (and animal viscera) impacts both children and adults and represents a hetero-
Pork-Cat geneous group of allergic disorders with multiple distinct syndromes. The recognition of entities such as pork-cat
Albumin
syndrome and delayed anaphylaxis to red meat, i.e- the α-Gal syndrome, have shed light on fundamental, and in
some cases newly appreciated, features of allergic disease. These include insights into routes of exposure and
mechanisms of sensitization, as well as the realization that IgE-mediated reactions can be delayed by several
hours. Here we review mammalian and avian meat allergy with an emphasis on the molecular allergens and
pathways that contribute to disease, as well as the role of in vitro IgE testing in diagnosis and management.

1. Introduction Reactions to mammalian meat are more common than for avian meat,
at least anecdotally, but neither is common. Mammalian meat allergy
Despite the fact that some animal products are well established food was once largely thought to be restricted to children, most commonly
allergens, such as milk and eggs, allergy to meat itself has historically those with atopic dermatitis or cow’s milk allergy (Werfel et al., 1997),
been considered to be quite rare. Case reports of allergy to mammalian but now is equally appreciated in adults. Part of the explanation relates
and avian meat became more commonplace starting about 20 years to the fact that several different forms of meat allergy have now been
ago, which in part also coincides with an increasing appreciation of recognized. There is significant regional variation in meat allergy,
food allergy in general (Platts-Mills, 2015). IgE-mediated reactions to which is likely a function of differences in local dietary habits, but other
many different types of meat have now been reported. The list includes environmental factors are also important. This is dramatically high-
beef, pork, lamb, and poultry, but also a host of others including kan- lighted by the realization that IgE sensitization to α-Gal is mediated by
garoo (Boyle et al., 2007), whale (Moore et al., 2007), seal (Moore bites from certain hard ticks. Thus, for example, there is a markedly
et al., 2007) and crocodile (Ballardini et al., 2017). A number of re- higher rate of allergic reactions to mammalian meat in the southeastern
levant allergens have been identified and characterized, and we have an United States, an area endemic with Amblyomma americanum (lone star
increasing appreciation of the natural history of meat allergy and re- ticks), as compared to other parts of the country (Commins et al., 2011).
levant cross-sensitizations. About 10 years ago a new form of meat al- The mechanisms and routes of exposure that lead to anaphylactic
lergy was recognized, which involves delayed anaphylaxis to mam- sensitization have been an active area of inquiry for over a century
malian meat, that relates to the oligosaccharide Gal-α1,3Gal- dating back to the pioneering work of Richet and Portier (Cohen and
β1,4GlcNAcR (α-Gal) (Commins et al., 2009). This allergy, which is Zelaya-Quesada, 2002). For some food allergens, such as peanut, there
often known as the α-Gal syndrome, has challenged many traditional has been convincing evidence that allergy results from epicutaneous
paradigms of how we think about food allergy (Wilson et al., 2017). The sensitization (Du Toit et al., 2008; Tordesillas et al., 2014), but for
present review considers various forms of meat allergy with a special many food allergens the route of sensitization is incompletely under-
emphasis on mammalian meat, aiming to highlight several advances stood. For ‘primary’ mammalian and avian meat allergy the suggestion
over the last decade. is that the inciting exposure is via the GI tract. However, many allergens
can also be present in airborne particles or skin products (Kligman and
2. Immunology and epidemiology Papa, 1965) leading to the possibility of respiratory or cutaneous sen-
sitization. Indeed, examples of syndromes where sensitization is es-
Good estimates of the prevalence of meat allergy do not exist. tablished to have occurred outside the GI tract include: pork-cat


Corresponding author at: FRS: Asthma and Allergic Diseases Center, University of Virginia, P.O. Box 801355, Charlottesville, VA, 22908-1355, USA.
E-mail address: tap2z@virginia.edu (T.A.E. Platts-Mills).

https://doi.org/10.1016/j.molimm.2018.03.018
Received 2 January 2018; Received in revised form 19 March 2018; Accepted 20 March 2018
Available online 21 April 2018
0161-5890/ © 2018 Elsevier Ltd. All rights reserved.
J.M. Wilson, T.A.E. Platts-Mills Molecular Immunology 100 (2018) 107–112

Table 1
Meat allergy syndromes.
Source Allergy syndrome Major Allergen(s) Route/Mode

Mammalian Meat Pork-cat Serum albumins Respiratory exposure to cat serum albumin in dander
α-Gal Galα1-3 Galβ1-4GlcNAcR Skin via tick bites
Avian meat Bird-egg Serum albumin Respiratory exposure to bird serum albumin in feathers
Fish-chicken Parvalbumin, Aldolase, Enolase Oral

(Posthumus et al., 2013), bird-egg (Hemmer et al., 2016) and α-Gal Table 3
syndromes (Commins et al., 2011) (see Table 1). Generally these forms Common serum albumin allergens in animals (from allergen.org, WHO/IUIS).
of allergy disproportionately impact adults and older children com- Common name Species Allergen Molecular Weight (kD)
pared to primary meat allergy, however young children can also be
*
affected. Fish-chicken syndrome is a more recently described entity that Domestic cattle Bos domesticus Bos d 6 67
likely involves cross-sensitization from GI exposure (Kuehn et al., Dog Canis familiaris Can f 3 69
Guinea pig Cavia porcellus Cav p 4 66
2016). Domestic horse Equus caballus Equ c 3 67
Cat Felis domesticus Fel d 2 69
Chicken Gallus domesticus Gal d 5 69
3. Biology and biochemistry
Domestic pig Sus scrofa Sus s 1 60

Serum albumin constitutes one of the most important contributors * traditionally referred to as Bos Taurus, but studies have not been done on
to both mammalian and avian meat allergy. In contrast, the oligo- native cow species.
saccharide α-Gal is selectively present only on mammalian tissue and
IgE antibody to an equivalent oligosaccharide has not been described in Thus, cases of albumin-related allergy to both mammal and bird pro-
avian allergy. Other less commonly identified allergens include im- ducts are very rare (Restani et al., 1997; Cahen et al., 1998). Serum
munoglobulin, myosin light chain kinase, parvalbumin, enolase and albumin cross-reactivity is a key feature in pork-cat syndrome, where
aldolase (see Table 2) although this list is certainly not complete primary sensitization to cat serum albumin, also known as Fel d 2, leads
(Restani et al., 2009). to allergic reactions upon ingestion of pork products containing pork
serum albumin, i.e. – Sus s 1. Interestingly, some of these subjects also
3.1. Albumins react to beef, which likely reflects further epitope spreading of the IgE
response to include Bos d 6 (Posthumus et al., 2013). Although his-
Serum albumins are ∼70 kD α-helical proteins that are highly torically the syndrome has been called ‘pork-cat’, some have advocated
conserved in sequence and conformation across many animals, in- that because cat sensitization precedes the allergic reaction to pork,
cluding mammals and birds (Chruszcz et al., 2013) (see Table 3). Serum that ‘cat-pork’ would be a more apt name (Hilger et al., 2017; Popescu,
albumins have multiple biologic functions but importantly they can 2015). Other examples of clinically relevant albumin cross-reactivity
cross capillary endothelia and are present in epithelia. Thus, in addition have been described in case reports. One such recent example involved
to being present in mammalian foods such as meat, milk and eggs, a woman with respiratory allergy to dog, who reported anaphylaxis to
animal pelts and bird feathers also contain serum albumins, with the horse meat. Her ensuing work-up revealed elevated IgE responses to
implication that inhalant and cutaneous exposure can occur (Liccardi dog extract as well as the serum albumins to dog (Can f 3) and horse
et al., 2011). There are several consequences that can result from the (Equ c 3). Supporting a diagnosis that would be consistent with ‘dog-
multitude of different sources of animal albumin. One is that it is horse’ is the fact that inhibition studies supported primary sensitization
common for subjects with beef allergy to have a co-existing milk al- to Can f 3 (Morisset et al., 2016).
lergy. Indeed, this was reflected among 28 young Italian children with Serum albumins are generally considered heat labile, and as such
beef allergy where 26 were sensitized specifically to Bos d 6 and all of the frequency and severity of reactions are likely reduced by consuming
these had immediate reactions upon milk challenge (Martelli et al., well-cooked animal products (Werfel et al., 1997; Fiocchi et al., 1998).
2002). Bird-egg syndrome represents a situation where primary sensi- Other approaches, such as freeze-drying, may be even more helpful for
tization to an avian serum albumin occurs via a respiratory route but reducing allergenicity (Fiocchi et al., 1998; Restani et al., 2004).
subsequently subjects develop allergic symptoms upon ingestion of
poultry (Szepfalusi et al., 1994; Quirce et al., 2001). 3.2. α-Gal
Cross-reactivity between albumins from different species is
common, but most often involves phylogenetically similar sources. When considering α-Gal it is important to realize that it was first
appreciated as a ‘B like’ blood group antigen by Landsteiner
Table 2 (Landsteiner and Miller, 1925). Indeed, it shares structural features
Important allergens from representative mammalian and avian meat sources. with the blood group B antigen (Fig. 1), and is the target of abundant
Source Allergen name Biochemical name ‘natural’ IgM, IgG and IgA antibodies in immunocompetent humans
(Hamadeh et al., 1995). The oligosaccharide is present in many mam-
Bovine Bos d 6 Serum albumin
malian foods, including meat, internal organs (such as kidney or tripe),
Bos d 7 Immunoglobulin
α-Gal Gal-α1,3 Gal-β1,4GlcNAcR* milk and other dairy, and gelatin (Mullins et al., 2012), but also other
Chicken Gal d 5 Serum albumin products such as the monoclonal antibody cetuximab, anti-venom and
Gal d 7 Myosin light chain kinase the zoster vaccine (Chung et al., 2008; Fischer et al., 2017a; Stone et al.,
Gal d 8 α-parvalbumin 2017). Among the features that distinguish α-Gal syndrome from other
Gal d 9 β-enolase
Gal d 10 Aldolase
IgE-mediated meat allergies (see Table 4) is the fact that reactions are
delayed, typically occurring 3–6 h after a relevant exposure (Commins
* α-Gal linkages have been described on a number of mammalian glyco- et al., 2014). From a clinical perspective this is an important char-
proteins and glycolipids (Chruszcz et al., 2013; Liccardi et al., 2011; Martelli acteristic and helps distinguish reactions related to α-Gal from those
et al., 2002). caused by IgE to other allergens. Anecdotally, we have seen several

108
J.M. Wilson, T.A.E. Platts-Mills Molecular Immunology 100 (2018) 107–112

Table 6
α-Gal linked glycoproteins recognized by IgE in subjects with red meat
allergy (Apostolovic et al., 2014).
Protein Molecular Weight (kD)

Alpha-enolase* 47.3
Beta-enolase 47.1
Aspartate aminotransferase 46.4
Creatine kinase M-type 43
Lactate dehydrogenase A 35.6
Carbonic anhydrase 3 29.4
Triosephosphate isomerase 26.7

* Glycoproteins in bold retained IgE binding after heat treatment.

Indeed, this is why we often refer to the α-Gal epitope as galactose-α-


Fig. 1. Comparison of structure of α-Gal and blood Group B antigen. 1,3-galactose. It should be pointed out, however, that the antibody
repertoire to α-Gal is broad and, at least in studies that investigated
Table 4
anti-Gal IgG, some antibodies can also bind the B antigen (Galili et al.,
Ways that α-Gal syndrome differs from traditional IgE-mediated food allergies. 1987; Milland et al., 2007; Galili, 2017). An implication of the het-
erogeneous specificity in anti-Gal antibodies is that differences in the
I Primary sensitization is mediated through the skin via tick bites (not oral
quantity and/or quality of the IgE antibody repertoire may impact
exposure)
II Allergy onset is usually in adults whether a sensitized subject experiences allergic symptoms upon a re-
III The major B cell epitope is an oligosaccharide levant ingestion. The point here is really two-fold: i) in population
IV Anaphylactic reactions are delayed, usually > 2 h studies many individuals produce IgE to α-Gal but do not have allergic
V Skin prick tests are not sufficiently sensitive symptoms, and ii) the extent of IgE affinity maturation and epitope
spreading is likely a factor in distinguishing allergic subjects from those
that are sensitized but tolerant. The former point is perhaps best ex-
patients in our clinic for evaluation of possible α-Gal syndrome where
emplified by a recent report of high-risk forest workers from southwest
the correct diagnosis involved IgE to bovine serum albumin, pork serum
Germany where 58 of 300 subjects were sensitized to α-Gal (cut-off of
albumin or gelatin. The case of gelatin is notable because some pre-
0.35 IU/mL), but only 5 of these had symptoms consistent with α-Gal
parations contain α-Gal and therefore it is possible to have IgE-medi-
syndrome, i.e – over 90% of the sensitized subjects in the cohort did not
ated reactions occurring to either the gelatin itself or the α-Gal com-
report relevant symptoms (Fischer et al., 2017b). The latter point is
ponent (Mullins et al., 2012; Stone et al., 2017; Retterer et al., 2018).
suggested by recent work from Jappe et al. where subjects with α-Gal
In the ten years since α-Gal was first identified as an important meat
syndrome had broad reactivity to a number of different α-Gal-con-
allergen there remain several important unanswered questions (see
taining epitopes (Jappe et al., 2018). Another possibility that could
Table 5) The mechanisms that contribute to the delay in clinical
explain why many subjects who are sensitized to α-Gal do not report
symptoms with α-Gal remain poorly understood. Importantly, this
symptoms, or do not report symptoms with every meat ingestion, is that
delay has been demonstrated in prospective meat challenge studies
there can be significant heterogeneity in the complexity of α-Gal linked
where ex vivo basophil activation occurred with similar kinetics
oligosaccharide structures. For example, α-Gal can be present on
(Commins et al., 2014). The explanation that seems most plausible in-
mono-, bi- or tri- antennary oligosaccharides, as shown in Fig. 3 for
volves the time required for processing, digestion and transit of α-Gal
Cetuximab. It is possible that IgE binding is favored when multiple α-
epitopes to target tissues. While a number of recent studies have fo-
Gal epitopes are in close proximity, which was supported by in vitro
cused on α-Gal containing glycoproteins, including in meat (see
experiments that compared IgE binding to cetuximab F(ab’)2 and Fab
Table 6) (Apostolovic et al., 2014; Hilger et al., 2016; Apostolovic et al.,
fragments with purified Gal-α1,3Gal-β1,4GlcNAc polysaccharide.
2017), α-Gal linked glycolipids are also well established in other
However, the details of the complexity of α-Gal-linked oligosaccharides
mammalian cells and tissues (Galili et al., 1987). The kinetics of lipid
have not been established for meat itself.
metabolism, which involves packaging into chylomicrons and transit
Any discussion about the relevance of a food allergen needs to
through lymphatics and the thoracic duct before entering the blood-
consider the stability of the epitope during food preparation and transit
stream, suggests the possibility that α-Gal-containing lipids are parti-
of the digestive tract. Results of prick-to-prick tests comparing raw or
cularly important in the delayed allergic response. Indeed, this hy-
cooked meat (beef and pork) in α-Gal subjects suggest that heating may
pothesis also fits with the observation that lean meat, particularly
have some effect on allergenicity (Fischer et al., 2014); on the other
venison, is less likely to trigger reactions in α-Gal allergic subjects than
hand α-Gal epitopes on glycoproteins in pork kidney retained reactivity
fatty cuts.
to a specific monoclonal antibody despite heating for 10 min at 95° C
The complete α-Gal epitope is considered to be the trisaccharide
(Hilger et al., 2016). Using beef thyroglobulin as a model, Apostolovic
form (i.e - Gal-α1,3Gal-β1,4GlcNAcR), however multiple studies have
et al. have shown that α-Gal peptides remain intact after in vitro pepsin
shown that the two terminal galactoses are the major binding de-
digestion. Not only was IgE binding maintained, but the glycopeptides
terminant (Milland et al., 2007; Plum et al., 2011; Rispens et al., 2013).
also stimulated basophils obtained from α-Gal allergic (but not non-
allergic) subjects (Apostolovic et al., 2017).
Table 5
Unanswered questions regarding the mechanism of reactions in α-Gal syn- 3.3. Immunoglobulin
drome.
Does the delay reflect time required for processing and digestion? A few studies have described immunoglobulin as a target of IgE in
Is there a difference between the response to α-Gal-containing glycolipid and meat allergic subjects, however the clinical relevance is less clear-cut
glycoproteins?
than for albumin and α-Gal (Werfel et al., 1997; Han et al., 2000). For
Is the complexity of the oligosaccharide, i.e. - mono vs bi-antennary, relevant?
Are multiple adjacent α-Gal moieties necessary for FcεRI cross-linking? example, among ten Japanese children with atopic dermatitis and re-
ported beef allergy, seven had a strong signal to BSA using IgE

109
J.M. Wilson, T.A.E. Platts-Mills Molecular Immunology 100 (2018) 107–112

immunoblots but three did not (Han et al., 2000). The sera from these
three subjects recognized ∼60 and 200 kD glycoproteins. The fact that
binding was inhibited by bovine gamma globulin suggested that im-
munoglobulin was the target of this IgE. One possibility is that a specific
glycosylation(s) could explain this binding, although this has not been
directly addressed (Raju et al., 2000). Alpha-Gal could represent one
such glycosylation, although the evidence for its presence on mammal
(non-human) IgA or IgM is stronger than for IgG (Adedoyin et al., 2006;
Gronlund et al., 2009).

4. Allergen sources

The majority of studies investigating meat allergy have relied on


natural sources of allergens. One exception, which relates to pork-cat
syndrome, is that a recombinant feline serum albumin, i.e. - Fel d 2, has
been developed by Phadia/Thermo-Fisher. As such, commercial assays
that use ImmunoCAP currently incorporate a recombinant Fel d 2 that Fig. 2. Correlation of sIgE to cetuximab (IU/mL) and beef thyroglobulin (IU/
has been expressed in a yeast system. This recombinant is absent of any mL) in 34 subjects with α-Gal syndrome and 11 control subjects. Modeled with
N-linked glycosylation and is reported to have a native folding pattern linear regression (p < 0.001).
(Jonas Lidholm, PhD, personal communication).
The assay for α-Gal warrants additional consideration. The com-
5. Approaches to meat allergy testing with focus on in vitro mercially available assay involves beef thyroglobulin conjugated on the
diagnostics solid-phase and is available through Phadia/Thermo-Fisher, and in the
United States through Viracor-IBT (Lees Summit, MO). Beef thyr-
Investigation of a suspected case of meat allergy often requires a oglobulin is heavily glycosylated with reports suggesting the possibility
multi-faceted approach, with component diagnostics playing an im- of 8–11 α-Gal linkages (Apostolovic et al., 2017; Thall and Galili,
portant role. Skin testing can be helpful, although sensitivity can be 1990). Research studies have also often used the monoclonal antibody
limiting with standard prick testing. Alternatives include attempting cetuximab conjugated by the streptavidin technique to the solid-phase.
prick-to-prick with fresh food sources or cautious use of intradermal Importantly, the performance of the two assays correlate closely as
testing. A shortcoming of the prick-to-prick approach is that there can demonstrated in Fig. 2 and reported by European colleagues (Jappe
be substantial variability in food preparations and results are not well et al., 2018). Despite the close correlation, the conclusion of Jappe et al.
validated. In our experience intradermal testing with commercially was that the cetuximab assay may be the more sensitive of the two
available beef, pork and lamb extracts can be done safely and correlates assays (Jappe et al., 2018). Another assay that has been used experi-
well with clinically relevant α-Gal allergy. However, in vitro tests can mentally uses α-Gal-conjugated to human serum albumin on the solid-
eliminate the risks associated with intradermal testing and variability phase, and this too had similar performance with the cetuximab assay
with prick-to-prick testing, and can be required to confirm a diagnosis (Lammerts van Bueren et al., 2011).
when identifying a specific allergen is important.
There are nuances of in vitro IgE testing which are important to
consider. For example, there are unique strengths and weaknesses that 6. Vaccine candidates
come with the use of extract versus component assays, or singleplex
versus multiplex assays (see Table 7). For extract tests an important The idea of desensitization to food allergens has recently gained
caveat relates to the fact that many meat allergens represent a minor traction in the allergy community, though meat has been little studied
fraction of the extract. As a consequence, the result of the extract assay in this regard. While we are aware of a recent case report describing
may be an underestimation of the magnitude of the IgE response to the successful desensitization in two α-Gal cases (Unal et al., 2017), we
specific allergen (Schuyler et al., 2017). Component tests do not suffer have not undertaken it and would not recommend desensitization
this limitation and, additionally, can be particularly helpful for iden- outside of research settings. An intriguing question is whether the
tifying the relevant epitope of a mammalian meat allergy. This is also continued consumption of foods that contain small amounts of α-Gal,
true for identifying allergens that may be involved in cross-sensitiza- such as some dairy products, could be protective for those that are al-
tion. In addition to extract tests, component tests that are particularly lergic to beef, but this has not been adequately addressed.
helpful in the evaluation of a putative mammalian meat allergy include:
α-Gal, Bos d 6, Sus s 1, Fel d 2 and gelatin.

Table 7
Considerations for in vitro IgE testing in meat allergy.
Extract –vs.– Component Singleplex – vs. – Multiplex

Strength • Readily
assays
available in commercial • Greater sensitivity and specificity
for identifying IgE epitope
• More allergen on the solid-phase &
thus greater sensitivity
• Can
test
test > 100 allergens with single

• Useful for initial screen • Helpful for establishing syndromes


with cross-sensitization
• Both extracts and components can
be conjugated to solid-phase
• Standard
albumins
panel has multiple serum

• Quantitative
Weakness • IgE result can be underestimate
for minor allergens
• Not all components are
commercially available
• Some components are not
available in this format
• Less sensitive, particularly in the
presence of high levels of specific IgG
• Cannot identify specific epitopes • Minor • Multiple tests likely required
4
impurities can be amplified or IgG
• Cannot
1
(in singleplex format) use extracts on solid-phase
• Standard panel may not include α-Gal
• Semi-quantitative
110
J.M. Wilson, T.A.E. Platts-Mills Molecular Immunology 100 (2018) 107–112

Fig. 3. Cetuximab is a chimeric IgG1 mono-


clonal antibody produced in a mouse cell line
(SP2/0). Demonstrated here are representative
bi- and tri-antennary glycans with terminal α-
Gal epitopes that are often present on the
variable region of the heavy chain (VH). Other
glycans with α-Gal are also possible. Glycans
are very common on the constant heavy chain
domain 2 (CH2) but only rarely do these have
terminal α-Gal epitopes. When present on the
Fc α-Gal epitopes are most frequently mono-
antennary. Anti-Gal IgE can bind to α-Gal epi-
topes on Fab, but not Fc because the tertiary
structure of the antibody precludes exposure of
these epitopes (Lammerts van Bueren et al.,
2011; Qian et al., 2007).

7. Conclusion and future directions Chung, C.H., Mirakhur, B., Chan, E., Le, Q.T., Berlin, J., Morse, M., Murphy, B.A.,
Satinover, S.M., Hosen, J., Mauro, D., Slebos, R.J., Zhou, Q., Gold, D., Hatley, T.,
Hicklin, D.J., Platts-Mills, T.A., 2008. Cetuximab-induced anaphylaxis and IgE spe-
Despite traditionally being considered rare, meat allergy is being cific for galactose-alpha-1,3-galactose. N. Engl. J. Med. 358, 1109–1117.
increasingly recognized in subjects of all ages. In part this may reflect Cohen, S.G., Zelaya-Quesada, M., 2002. Portier, Richet, and the discovery of anaphylaxis:
an increasing incidence, but also an appreciation that regional differ- a centennial. J. Allergy Clin. Immunol. 110, 331–336.
Commins, S.P., Satinover, S.M., Hosen, J., Mozena, J., Borish, L., Lewis, B.D., Woodfolk,
ences in exposure can have a major impact on prevalence of the disease. J.A., Platts-Mills, T.A., 2009. Delayed anaphylaxis, angioedema, or urticaria after
The increase has occurred at the same time as increases in other allergic consumption of red meat in patients with IgE antibodies specific for galactose-alpha-
diseases. The development of in vitro diagnostics has helped define 1,3-galactose. J. Allergy Clin. Immunol. 123, 426–433.
Commins, S.P., James, H.R., Kelly, L.A., Pochan, S.L., Workman, L.J., Perzanowski, M.S.,
important syndromes in meat allergy, i.e.-α-Gal and pork-cat, and has Kocan, K.M., Fahy, J.V., Nganga, L.W., Ronmark, E., Cooper, P.J., Platts-Mills, T.A.,
been an important tool in clinical practice for confirming diagnosis. 2011. The relevance of tick bites to the production of IgE antibodies to the mam-
Identification of relevant allergens has had important consequences for malian oligosaccharide galactose-alpha-1,3-galactose. J. Allergy Clin. Immunol. 127,
1286–1293 e1286.
disease management. This includes tailored dietary information to the
Commins, S.P., James, H.R., Stevens, W., Pochan, S.L., Land, M.H., King, C., Mozzicato,
patient, but also insight into the exposures and underlying mechanisms S., Platts-Mills, T.A., 2014. Delayed clinical and ex vivo response to mammalian meat
that lead to and/or promote ongoing sensitization. in patients with IgE to galactose-alpha-1,3-galactose. J. Allergy Clin. Immunol. 134,
108–115.
Du Toit, G., Katz, Y., Sasieni, P., Mesher, D., Maleki, S.J., Fisher, H.R., Fox, A.T., Turcanu,
Funding sources V., Amir, T., Zadik-Mnuhin, G., Cohen, A., Livne, I., Lack, G., 2008. Early con-
sumption of peanuts in infancy is associated with a low prevalence of peanut allergy.
TAEPM has a patent on an IgE assay for α-Gal, has received assay J. Allergy Clin. Immunol. 122, 984–991.
Fiocchi, A., Restani, P., Riva, E., Mirri, G.P., Santini, I., Bernardo, L., Galli, C.L., 1998.
support from Thermo-Fisher/Phadia and has a grant from the National Heat treatment modifies the allergenicity of beef and bovine serum albumin. Allergy
Institute of Health, AI-20565. JMW has no disclosures. 53, 798–802.
Fischer, J., Hebsaker, J., Caponetto, P., Platts-Mills, T.A., Biedermann, T., 2014.
Galactose-alpha-1,3-galactose sensitization is a prerequisite for pork-kidney allergy
References and cofactor-related mammalian meat anaphylaxis. J. Allergy Clin. Immunol. 134,
755–759 e751.
Adedoyin, J., Johansson, S.G., Gronlund, H., van Hage, M., 2006. Interference in im- Fischer, J., Eberlein, B., Hilger, C., Eyer, F., Eyerich, S., Ollert, M., Biedermann, T., 2017a.
munoassays by human IgM with specificity for the carbohydrate moiety of animal Alpha-gal is a possible target of IgE-mediated reactivity to antivenom. Allergy 72,
proteins. J. Immunol. Methods 310, 117–125. 764–771.
Apostolovic, D., Tran, T.A., Hamsten, C., Starkhammar, M., Cirkovic Velickovic, T., van Fischer, J., Lupberger, E., Hebsaker, J., Blumenstock, G., Aichinger, E., Yazdi, A.S., Reick,
Hage, M., 2014. Immunoproteomics of processed beef proteins reveal novel ga- D., Oehme, R., Biedermann, T., 2017b. Prevalence of type I sensitization to alpha-gal
lactose-alpha-1,3-galactose-containing allergens. Allergy 69, 1308–1315. in forest service employees and hunters. Allergy 72, 1540–1547.
Apostolovic, D., Krstic, M., Mihailovic, J., Starkhammar, M., Cirkovic Velickovic, T., Galili, U., 2017. The Natural Anti-Gal Antibody as Foe Turned Friend in Medicine, 1st
Hamsten, C., van Hage, M., 2017. Peptidomics of an in vitro digested alpha-Gal edition. Elsevier, Cambridge, MA.
carrying protein revealed IgE-reactive peptides. Sci. Rep. 7, 5201. Galili, U., Basbaum, C.B., Shohet, S.B., Buehler, J., Macher, B.A., 1987. Identification of
Ballardini, N., Nopp, A., Hamsten, C., Vetander, M., Melen, E., Nilsson, C., Ollert, M., erythrocyte Gal alpha 1-3Gal glycosphingolipids with a mouse monoclonal antibody,
Flohr, C., Kuehn, A., van Hage, M., 2017. Anaphylactic reactions to novel foods: case Gal-13. J. Biol. Chem. 262, 4683–4688.
report of a child with severe crocodile meat allergy. Pediatrics 139. Gronlund, H., Adedoyin, J., Commins, S.P., Platts-Mills, T.A., van Hage, M., 2009. The
Boyle, R.J., Russo, V.C., Andaloro, E., Mehr, S.M., Tang, M.L., 2007. Anaphylaxis to carbohydrate galactose-alpha-1,3-galactose is a major IgE-binding epitope on cat IgA.
kangaroo meat: identification of a new marsupial allergen. Allergy 62, 209–211. J. Allergy Clin. Immunol. 123, 1189–1191.
Cahen, Y.D., Fritsch, R., Wuthrich, B., 1998. Food allergy with monovalent sensitivity to Hamadeh, R.M., Galili, U., Zhou, P., Griffiss, J.M., 1995. Anti-alpha-galactosyl im-
poultry meat. Clin. Exp. Allergy 28, 1026–1030. munoglobulin a (IgA), IgG, and IgM in human secretions. Clin. Diagn. Lab. Immunol.
Chruszcz, M., Mikolajczak, K., Mank, N., Majorek, K.A., Porebski, P.J., Minor, W., 2013. 2, 125–131.
Serum albumins-unusual allergens. Biochim. Biophys. Acta 1830, 5375–5381. Han, G.D., Matsuno, M., Ito, G., Ikeucht, Y., Suzuki, A., 2000. Meat allergy: investigation

111
J.M. Wilson, T.A.E. Platts-Mills Molecular Immunology 100 (2018) 107–112

of potential allergenic proteins in beef. Biosci. Biotechnol. Biochem. 64, 1887–1895. Posthumus, J., James, H.R., Lane, C.J., Matos, L.A., Platts-Mills, T.A., Commins, S.P.,
Hemmer, W., Klug, C., Swoboda, I., 2016. Update on the bird-egg syndrome and genuine 2013. Initial description of pork-cat syndrome in the United States. J. Allergy Clin.
poultry meat allergy. Allergo J. Int. 25, 68–75. Immunol. 131, 923–925.
Hilger, C., Fischer, J., Swiontek, K., Hentges, F., Lehners, C., Eberlein, B., Morisset, M., Qian, J., Liu, T., Yang, L., Daus, A., Crowley, R., Zhou, Q., 2007. Structural character-
Biedermann, T., Ollert, M., 2016. Two galactose-alpha-1,3-galactose carrying pepti- ization of N-linked oligosaccharides on monoclonal antibody cetuximab by the
dases from pork kidney mediate anaphylactogenic responses in delayed meat allergy. combination of orthogonal matrix-assisted laser desorption/ionization hybrid quad-
Allergy 71, 711–719. rupole-quadrupole time-of-flight tandem mass spectrometry and sequential enzy-
Hilger, C., Hemmer, W., Swoboda, I., Morisset, M., Fischer, J., Tripathi, A., Platts-Mills, matic digestion. Anal. Biochem. 364, 8–18.
T., Biedermann, T., 2017. Molecular and extract-based diagnostics in meat allergy. In: Quirce, S., Maranon, F., Umpierrez, A., de las Heras, M., Fernandez-Caldas, E., Sastre, J.,
Kleine-Tebbe, J., Jakob, T. (Eds.), Molecular Allergy Diagnostics: Innovation for a 2001. Chicken serum albumin (Gal d 5*) is a partially heat-labile inhalant and food
Better Patient Management. Springer International Publishing, Cham, pp. 305–326. allergen implicated in the bird-egg syndrome. Allergy 56, 754–762.
Jappe, U., Minge, S., Kreft, B., Ludwig, A., Przybilla, B., Walker, A., Varga, R., Seidel, P., Raju, T.S., Briggs, J.B., Borge, S.M., Jones, A.J., 2000. Species-specific variation in gly-
Biedermann, T., Anemuller, W., Kromminga, A., Rueff, F., Merk, H., Wagner, N., cosylation of IgG: evidence for the species-specific sialylation and branch-specific
Treudler, R., Worm, M., Waldmann, I., Saloga, J., Becker, W.M., Goldmann, T., Platts- galactosylation and importance for engineering recombinant glycoprotein ther-
Mills, T.A., Homann, A., 2018. Meat allergy associated with galactosyl-alpha-(1,3)- apeutics. Glycobiology 10, 477–486.
galactose (alpha-Gal)-Closing diagnostic gaps by anti-alpha-Gal IgE immune pro- Restani, P., Fiocchi, A., Beretta, B., Velona, T., Giovannini, M., Galli, C.L., 1997. Meat
filing. Allergy 73, 93–105. http://dx.doi.org/10.1111/all.13238. allergy: III–Proteins involved and cross-reactivity between different animal species. J.
Kligman, A.P., Papa, C.hristopher M., 1965. Albumin As an antiwrinkling cosmetic. J. Am. Coll. Nutr. 16, 383–389.
Soc. Cosmet. Chem. 1965, 557–562. Restani, P., Ballabio, C., Cattaneo, A., Isoardi, P., Terracciano, L., Fiocchi, A., 2004.
Kuehn, A., Codreanu-Morel, F., Lehners-Weber, C., Doyen, V., Gomez-Andre, S.A., Characterization of bovine serum albumin epitopes and their role in allergic reac-
Bienvenu, F., Fischer, J., Ballardini, N., van Hage, M., Perotin, J.M., Silcret-Grieu, S., tions. Allergy 59 (Suppl. 78), 21–24.
Chabane, H., Hentges, F., Ollert, M., Hilger, C., Morisset, M., 2016. Cross-reactivity to Restani, P., Ballabio, C., Tripodi, S., Fiocchi, A., 2009. Meat allergy. Curr. Opin. Allergy
fish and chicken meat - a new clinical syndrome. Allergy 71, 1772–1781. Clin. Immunol. 9, 265–269.
Lammerts van Bueren, J.J., Rispens, T., Verploegen, S., van der Palen-Merkus, T., Stapel, Retterer, M.K.C., Workman, L.J., Bacon, J.R., Platts-Mills, T.A.E., 2018. Specific IgE to
S., Workman, L.J., James, H., van Berkel, P.H., van de Winkel, J.G., Platts-Mills, T.A., gelatin as a cause of anaphylaxis to zoster vaccine. J. Allergy Clin. Immunol. http://
Parren, P.W., 2011. Anti-galactose-alpha-1,3-galactose IgE from allergic patients does dx.doi.org/10.1016/j.jaci.2017.08.047.
not bind alpha-galactosylated glycans on intact therapeutic antibody Fc domains. Rispens, T., Derksen, N.I., Commins, S.P., Platts-Mills, T.A., Aalberse, R.C., 2013. IgE
Nat. Biotechnol. 29, 574–576. production to alpha-gal is accompanied by elevated levels of specific IgG1 antibodies
Landsteiner, K., Miller, C.P., 1925. Serological studies on the blood of the primates : Iii. and low amounts of IgE to blood group B. PLoS One 8, e55566.
Distribution of serological factors related to human isoagglutinogens in the blood of Schuyler, A.J., Tripathi, A., Workman, L.J., Wilson, J.M., Erwin, E.A., Lawrence, M.G.,
Lower monkeys. J. Exp. Med. 42, 863–872. McGowan, E.C., Hamilton, R.G., Platts-Mills, T.A.E., 2017. Underestimation of spe-
Liccardi, G., Asero, R., D’Amato, M., D’Amato, G., 2011. Role of sensitization to mam- cific IgE measurements using extract-based assays on undiluted sera revealed through
malian serum albumin in allergic disease. Curr. Allergy Asthma Rep. 11, 421–426. dilution. J. Allergy Clin. Immunol. Pract. http://dx.doi.org/10.1016/j.jaip.2017.10.
Martelli, A., De Chiara, A., Corvo, M., Restani, P., Fiocchi, A., 2002. Beef allergy in 037.
children with cow’s milk allergy; cow’s milk allergy in children with beef allergy. Stone Jr., C.A., Hemler, J.A., Commins, S.P., Schuyler, A.J., Phillips, E.J., Peebles Jr., R.S.,
Ann. Allergy Asthma Immunol. 89, 38–43. Fahrenholz, J.M., 2017. Anaphylaxis after zoster vaccine: implicating alpha-gal al-
Milland, J., Yuriev, E., Xing, P.X., McKenzie, I.F., Ramsland, P.A., Sandrin, M.S., 2007. lergy as a possible mechanism. J. Allergy Clin. Immunol. 139, 1710–1713 e1712.
Carbohydrate residues downstream of the terminal Galalpha(1,3)Gal epitope mod- Szepfalusi, Z., Ebner, C., Pandjaitan, R., Orlicek, F., Scheiner, O., Boltz-Nitulescu, G.,
ulate the specificity of xenoreactive antibodies. Immunol. Cell. Biol. 85, 623–632. Kraft, D., Ebner, H., 1994. Egg yolk alpha-livetin (chicken serum albumin) is a cross-
Moore, L.M., Rathkopf, M.M., Sanner, C.J., Whisman, B.A., Demain, J.G., 2007. Seal and reactive allergen in the bird-egg syndrome. J. Allergy Clin. Immunol. 93, 932–942.
whale meat: two newly recognized food allergies. Ann. Allergy Asthma Immunol. 98, Thall, A., Galili, U., 1990. Distribution of Gal alpha 1—3Gal beta 1—4GlcNAc residues on
92–96. secreted mammalian glycoproteins (thyroglobulin, fibrinogen, and immunoglobulin
Morisset, M., Arumugam, K., Ollert, M., Hilger, C., 2016. Horse-meat allergy mediated by G) as measured by a sensitive solid-phase radioimmunoassay. Biochemistry 29,
dog-allergy: a case report and review of the literature. Allergo J. 25, 30–35. 3959–3965.
Mullins, R.J., James, H., Platts-Mills, T.A., Commins, S., 2012. Relationship between red Tordesillas, L., Goswami, R., Benede, S., Grishina, G., Dunkin, D., Jarvinen, K.M., Maleki,
meat allergy and sensitization to gelatin and galactose-alpha-1,3-galactose. J. Allergy S.J., Sampson, H.A., Berin, M.C., 2014. Skin exposure promotes a Th2-dependent
Clin. Immunol. 129, 1334–1342 e1331. sensitization to peanut allergens. J. Clin. Invest. 124, 4965–4975.
Platts-Mills, T.A., 2015. The allergy epidemics: 1870-2010. J. Allergy Clin. Immunol. 136, Unal, D., Coskun, R., Demir, S., Gelincik, A., Colakoglu, B., Buyukozturk, S., 2017.
3–13. Successful beef desensitization in 2 adult patients with a delayed-type reaction to red
Plum, M., Michel, Y., Wallach, K., Raiber, T., Blank, S., Bantleon, F.I., Diethers, A., meat. J. Allergy Clin. Immunol. Pract. 5, 502–503.
Greunke, K., Braren, I., Hackl, T., Meyer, B., Spillner, E., 2011. Close-up of the im- Werfel, S.J., Cooke, S.K., Sampson, H.A., 1997. Clinical reactivity to beef in children
munogenic alpha1,3-galactose epitope as defined by a monoclonal chimeric im- allergic to cow’s milk. J. Allergy Clin. Immunol. 99, 293–300.
munoglobulin E and human serum using saturation transfer difference (STD) NMR. J. Wilson, J.M., Schuyler, A.J., Schroeder, N., Platts-Mills, T.A., 2017. Galactose-alpha-1,3-
Biol. Chem. 286, 43103–43111. Galactose: atypical food allergen or model IgE hypersensitivity? Curr. Allergy Asthma
Popescu, F.D., 2015. Cross-reactivity between aeroallergens and food allergens. World J. Rep. 17, 8.
Methodol. 5, 31–50.

112

You might also like