ArticlePDF Available

A Molecular Diagnostic Algorithm to Guide Pollen Immunotherapy in Southern Europe: Towards Component-Resolved Management of Allergic Diseases

Authors:

Abstract

Correct identification of the culprit allergen is an essential part of diagnosis and treatment in immunoglobulin E (IgE)-mediated allergic diseases. In recent years, molecular biology has made important advances facilitating such identification and overcoming some of the drawbacks of natural allergen extracts, which consist of mixtures of various proteins that may be allergenic or not, specific for the allergen source or widely distributed (panallergens). New technologies offer the opportunity for a more accurate component-resolved diagnosis, of benefit especially to polysensitized allergic patients. The basic elements of molecular diagnostics with potential relevance to immunotherapy prescription are reviewed here, with a focus on Southern European sensitization patterns to pollen allergens. We propose a basic algorithm regarding component-resolved diagnostic work-up for pollen allergen-specific immunotherapy candidates in Southern Europe; this and similar algorithms can form the basis of improved patient management, conceptually a 'Component-Resolved Allergy Management'.
E-Mail karger@karger.com
Original Paper
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
A Molecular Diagnostic Algorithm to Guide
Pollen Immunotherapy in Southern Europe:
Towards Component-Resolved Management
of Allergic Diseases
NikolaosDouladiris SavvasSavvatianos IreneRoumpedaki
ChrysanthiSkevaki DimitriosMitsias NikolaosG.Papadopoulos
Allergy Department, 2nd Pediatric Clinic, University of Athens, Athens , Greece
Introduction
Immunoglobulin E (IgE)-mediated allergic respirato-
ry diseases, namely allergic rhinitis/rhinoconjunctivitis
and asthma, represent a major health issue in most devel-
oped countries
[1, 2] . From a mechanistic point of view,
the central role of inhalant allergens in both disease
pathogenesis and the elicitation of symptoms are indis-
putable. However, allergen avoidance is generally thought
to be impractical and/or ineffective, both in the preven-
tion of allergy and the induction of clinical remission in
established disease, as most trials have failed to show sig-
nificant benefit from avoidance measures
[3, 4] .
Given the limited effectiveness of such a directly caus-
al approach, pharmacotherapy, mainly the use of anti-
histamines, bronchodilators and anti-inflammatory
agents, is considered the cornerstone of treatment
[1, 5] .
Nevertheless, allergen-specific immunotherapy (SIT)
represents an important intervention, currently adjunc-
tive to pharmacological treatment, which confers spe-
cific advantages over conventional management. SIT
has been shown to exhibit disease-modifying potential,
with a clinical benefit that outlasts treatment duration
Key Words
Allergen-specific immunotherapy · Molecular diagnosis ·
Panallergens · Pollen
Abstract
Correct identification of the culprit allergen is an essential
part of diagnosis and treatment in immunoglobulin E (IgE)-
mediated allergic diseases. In recent years, molecular biolo-
gy has made important advances facilitating such identifica-
tion and overcoming some of the drawbacks of natural al-
lergen extracts, which consist of mixtures of various proteins
that may be allergenic or not, specific for the allergen source
or widely distributed (panallergens). New technologies offer
the opportunity for a more accurate component-resolved di-
agnosis, of benefit especially to polysensitized allergic pa-
tients. The basic elements of molecular diagnostics with po-
tential relevance to immunotherapy prescription are re-
viewed here, with a focus on Southern European sensitization
patterns to pollen allergens. We propose a basic algorithm
regarding component-resolved diagnostic work-up for pol-
len allergen-specific immunotherapy candidates in South-
ern Europe; this and similar algorithms can form the basis of
improved patient management, conceptually a ‘Compo-
nent-Resolved Allergy Management’.
Copyright © 2013 S. Karger AG, Basel
Received: February 20, 2013
Accepted after revision: May 17, 2013
Published online: July 31, 2013
Correspondence to: Dr. Savvas Savvatianos
Allergy Department, 2nd Pediatric Clinic, University of Athens
41, Fidippidou Str.
GR–11527 Athens (Greece)
E-Mail ss
@ allergy.gr
© 2013 S. Karger AG, Basel
1018–2438/13/1622–0065$38.00/0
www.karger.com/iaa
N.D. and S.S. contributed equally to this manuscript.
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
Douladiris/Savvatianos/Roumpedaki/
Skevaki/Mitsias/Papadopoulos
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
66
[6, 7] , and has been attributed preventive capacity against
the progression of allergic rhinitis to asthma and the de-
velopment of IgE-sensitization to additional allergens
[8, 9] .
Although much remains to be elucidated regarding
underlying mechanisms in different types of SIT, it is a
common understanding that SIT acts in an allergen-spe-
cific manner
[10] . It should therefore be anticipated that
the efficacy of SIT may be highly dependent on both the
ability to correctly identify the culprit allergen(s) and on
the availability and concentration of the relevant aller-
gens in therapeutic extracts. This represents a consider-
able challenge as diagnostic extracts, whether intended
for serological (in vitro) or skin-prick (in vivo) testing, are
traditionally based on crude extracts of the allergen
source, thus containing a mixture of allergens, relevant
and not relevant. The same applies for immunotherapy
vaccines, as recombinant allergens are currently not li-
censed for therapeutical use in humans, and all commer-
cial immunotherapy products are extracts of the raw al-
lergenic materials, containing variable levels of both al-
lergenic and nonallergenic substances. Moreover,
information is scarce regarding the relative concentration
of specific allergens in natural extracts, either diagnostic
or therapeutic, while methodological heterogeneity is the
rule for the sporadically published reports
[11–13] on the
topic; this further complicates the issue of matching im-
munotherapy prescription to specific IgE-sensitization
patterns and might at least partially account for some ob-
served ineffectiveness in immunotherapy trials and in
routine practice.
Recently, a major step forward in the direction of di-
agnostic optimization was signaled by the introduction of
component-resolved diagnostics (CRD)
[14] . Analyzing
patient serum IgE-specificity to the level of single aller-
gens is now possible, enhancing our overview of the IgE-
sensitization profile; this would be expected to facilitate
selection of candidates for immunotherapy and adminis-
tration of the appropriate extracts. However, to our
knowledge, since the introduction of component-re-
solved diagnosis, only a few published articles exist on
how to use the corresponding results for the optimal se-
lection of immunotherapy candidates
[15–18] . This is of
prime importance in the Mediterranean region; in con-
trast to the more or less clearly separated pollination pe-
riods of major allergenic plants in the north of Europe,
the overlapping flowering periods of allergenic plants in
the south, in conjunction with an abundance of multisen-
sitization profiles, frequently hamper diagnosis and make
it difficult to assess the clinical relevance of a given IgE
sensitization
[15, 16] . It is possible that such a diagnostic
work-up may also be applicable in regions across the
world with similar aerobiological characteristics, for ex-
ample, in parts of Australia or in the southern USA (e.g.
Florida).
Although the published reports provide a valuable
first insight in the utilization of these exciting new tech-
niques, they mainly focus on distinguishing patients
sensitized to species-specific allergens from those sensi-
tized to cross-reactive allergens. The basic principle is
that identification of IgE-sensitization to a species-spe-
cific allergen is a marker of genuine sensitization, rules
out positivity due to cross-reacting components and is
therefore a prerequisite for the selection of a therapeuti-
cal extract of the corresponding natural source. It is pos-
sible, nonetheless, that other elements may also be incor-
porated in such an analysis, like available published data
on the relative allergen representation in commercial ex-
tracts, confounding factors in in vitro diagnosis and po-
tential markers indicating a reduced anticipated re-
sponse to specific immunotherapy. By taking the above
points into account, we attempt to build upon existing
algorithms and propose a practical framework for opti-
mized immunotherapy candidate selection, with a focus
on the most important allergenic pollen sources in the
Mediterranean. This and other similar algorithms can
form the basis of improved patient management, con-
ceptually a ‘Component-Resolved Allergy Management’
(CRM).
P o l l e n A l l e r g y
There is general consensus that SIT should be indi-
cated in cases of established clinical relevance of an aller-
gen source
[19] . When seasonal symptoms point to pol-
len allergy, in vivo and/or in vitro testing typically con-
firm the presence of IgE to the specific pollen. In cases of
IgE-sensitization to more than one pollen source, it is im-
portant to identify the clinically significant pollens and
exclude any source that may appear positive exclusively
by cross-reactivity, as misrecognizing the primary sensi-
tizing source(s) may in turn compromise immunological
responses to SIT. This is especially relevant in southern
Europe, where a clinical history may be suboptimally in-
formative, owing to the partial overlap of the pollinating
periods of grasses, trees and weeds.
A number of assays are now routinely available, de-
tecting IgE towards pollen allergen components. These
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
CRM of Allergic Disease in Southern
Europe
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
67
can be classified to those signifying genuine sensitization
to a given source (species-specific allergens) and others,
indicating broad cross-sensitization.
Markers of Genuine Sensitization to Major Pollen
Sources
Grasses
Grasses are the only allergenic pollen species with
ubiquitous representation and clinical significance across
Europe. Therefore, it is not surprising that half of all com-
mercially marketed CRD assays for inhalant sources in-
volve grass allergens. Among these, Phl p 5 and Phl p 6 do
not exhibit significant serological cross-reactivity to pol-
len allergens outside the Pooideae subfamily, and are thus
regarded as species-specific allergens for Timothy grass
(Phleum pratense) and all other Pooideae members, in-
cluding common meadow grass (Poa pratensis) , orchard
grass (Dactylis glomerata) and perennial ryegrass (Lolium
perenne)
[20] . IgE to Phl p 2 may also be regarded as fair-
ly specific for patients sensitized to grass species of the
Pooideae subfamily; although Phl p 2 can also be found
in non-Pooideae grass species (e.g. Zea mays and Cyn-
odon dactylon ), its immunological importance seems to
be confined to the Pooideae subfamily
[21] .
When IgE specific to one or more of these component
allergens is detected and the clinical history suggests
grass allergy, grass pollen immunotherapy is indicated.
Identification of the exact Pooideae member inducing
primary sensitization may not be as important; both
mixed grass pollen extracts and single representative spe-
cies ( P. pratense pollen) have been argued to be effective
[22–25] .
Phl p 1 represents the archetype of group 1 grass pollen
allergens and is the most prevalent allergen among grass-
allergic patients
[26, 27] . However, the corresponding IgE
is only partially specific for the Pooideae grass subfamily,
dominating in Europe; partial cross-reactivity between
Phl p 1 and Cyn d 1, the group 1 major allergen in Ber-
muda grass (C. dactylon) , frequently hampers correct
identification of the sensitizing allergen source
[28] . Spe-
cifically, when testing for Pooideae-specific molecules
(e.g. rPhl p 2, rPhl p 5 and rPhl p 6) is negative, relatively
higher levels of IgE specific to nCyn d 1 than to rPhl p 1
have been suggested to be indicative of primary sensitiza-
tion to Bermuda grass
[29] ; a SIT extract containing Ber-
muda grass pollen might then be suitable, especially as
such an extract may be quantitatively standardized ac-
cording to group 1 allergen (nCyn d 1) content. In con-
trast, if antibodies against Pooideae-specific molecules
(e.g. rPhl p 5) are positive and specific IgE levels against
rPhl p 1 are higher than those to nCyn d 1, the case is most
probably primary sensitization to Pooideae grasses and
Bermuda pollen representation can be omitted from the
SIT regimen. Finally, if both conditions are present (i.e.
group 5 antibodies-positive and specific IgE against nCyn
d 1 higher than to rPhl p 1), the clinician may be facing a
true double sensitization.
It should not be overlooked that clinical response to
immunotherapy relies on adequate representation of sen-
sitizing allergens in SIT extracts; in this regard, the con-
cept of maximum tolerated extract dose is advocated
[30] .
Most manufacturers determine the concentration of only
one major allergen as part of the standardization process
[12] ; information regarding additional allergen content is
generally not available. It is expected, therefore, that if
IgE-sensitization proves positive against the same major
allergen that is employed in extract standardization, this
would ensure the enhanced possibility of response to SIT.
This is underscored by the fact that the levels of major al-
lergen content and the biological potency of extracts have
repeatedly been found to correlate well
[13, 31] . In this
context, detection of anti-rPhl p 5 IgE, compared to other
grass-specific markers, has added value in terms of asso-
ciated grass-immunotherapy outcome, as rPhl p 5 quan-
tification is the rule for such standardization by compa-
nies and Phl p 5 is typically found in abundance in ex-
tracts
[12, 13, 28, 31, 32] .
T r e e s
In a similar fashion to grass pollen CRD, markers for
better response to immunotherapy can be utilized for the
most important allergenic tree pollen in southern Europe.
nOle e 1/rOle e 1 and nCup a 1 are specific markers for
olive and cypress pollen, respectively, and are available
for routine testing. Potential positivity suggests authentic
sensitization and validates selection of the corresponding
natural SIT extract, when compatible symptoms are pres-
ent. Briefly, Ole e 1 is the most frequent sensitizing aller-
gen in olive pollen
[33] ; it is also typically used in the
standardization process of olive pollen diagnostic and
therapeutic extracts, therefore it is not surprising that Ole
e 1 is the only allergen yet proven to induce immunolog-
ical changes during olive pollen SIT
[34] . Ole e 1-like mol-
ecules are also present in grass (Phl p 11), chenopod (Che
a 1), English plantain (Pla l 1) and saltwort pollen (Sal k
5)
[35] . It seems, however, that cross-reactivity of Ole e 1
with the molecules outside the Oleaceae family is limited,
probably nonsignificant
[36, 37] . Importantly, and owing
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
Douladiris/Savvatianos/Roumpedaki/
Skevaki/Mitsias/Papadopoulos
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
68
to the high cross-reactivity between Ole e 1-like mole-
cules within the Oleaceae family, positivity to rOle e 1
may serve as a marker of the suitability of ash or privet
pollen SIT in olive-free areas with a high exposure to pol-
len of these closely related Oleaceae members
[36–38] .
nCup a 1 is the only available diagnostic marker for
primary sensitization to pollen of the Cupressaceae fam-
ily. Due to the structural homology of their major aller-
gens, genuine sensitization to either Cupressus arizonica
and/or C. sempervirens , both prevalent in the Mediterra-
nean basin, may be confirmed by this single assay
[39] . In
this case, the high level of cross-antigenicity between
members of the Cupressaceae family may justify thera-
peutic administration of a single representative pollen ex-
tract, even if it is not the same as the originally sensitizing
species
[40] .
Plane tree (Platanus acerifolia) pollen can be a cause of
respiratory allergy in some patients. Major allergens rPla
a 1 and nPla a 2 are commercially available only through
the ISAC allergen biochip immunoassay (Thermo Fisher
Scientific, Uppsala, Sweden) which, unfortunately, is not
very widely used. Positive specific IgE (sIgE) against these
components may serve as a marker of primary sensitiza-
tion to plane tree pollen
[41, 42] . In contrast, rPla a 3 is a
minor allergen that belongs to the nonspecific lipid trans-
fer protein (LTP) family and has been linked with IgE-
sensitization to plant-food LTPs
[43] .
W e e d s
Wall pellitory ( Parietaria judaica and P. officinalis ),
mugwort (Artemisia vulgaris) and, lately, saltwort (S.
kali) are regarded as the most prevalent allergenic weeds
in the Mediterranean
[44–46] . With respect to wall pelli-
tory allergy, Par j 1 and Par j 2 have been described as
major allergens in P. judaica pollen
[47, 48] . Although
they both belong to the largely cross-reactive LTP aller-
gen group, they in fact lack significant cross-reactivity to
other pollen or plant-food LTPs, thus conferring specific-
ity of induced IgE for the Parietaria genus
[49–51] . How-
ever, only rPar j 2 is commercially available for routine
testing. Theoretically, this would have an impact on the
assay’s sensitivity, i.e. the assay’s ability to detect weed-
allergic patients genuinely sensitized to Parietaria pollen.
Nonetheless, detection of IgE to rPar j 2 alone has been
proven to be sufficient in more than 80% of cases
[48] ,
probably owing to Par j 1 and Par j 2 sharing at least three
identical allergenic epitopes
[47] . The reported structural
similarity is primarily held to account for the demonstrat-
ed potency of Par j 1-standardized Parietaria pollen ex-
tracts in anti-rPar j 2-IgE-positive patients
[52] . By taking
the above into account, clinicians may use rPar j 2 as an
efficient Parietaria -specific marker when considering
weed-specific immunotherapy.
Although both nArt v 1 and nArt v 3 have been re-
ported to be species-specific molecules in mugwort-aller-
gic patients and potential markers of positive response to
SIT
[29] , the role of sensitization to the nsLTP Art v 3 may
not be as straightforward. There are conflicting data re-
garding the role of Art v 3 as a primary sensitizer. The
selection of the population is a probable reason for this,
but what seems to be of prime importance is the geo-
graphic area under study; in European (Mediterranean)
populations, Art v 3 may be of little clinical relevance,
generally considered to reflect primary sensitization to
cross-reacting plant-food LTPs
[53–56] . In contrast,
cross-reactivity between plant-food and mugwort LTP
has been shown to be inversely directed in selected pa-
tients and in patients in northern China
[57, 58] . In any
case, the total content of Art v 3 in crude pollen material
has been shown to strikingly vary from batch to batch,
hardly being detected in some
[51] ; this would put into
question the reliability/sufficiency of SIT natural extracts
for patients sensitized only to Art v 3. For the above rea-
sons, the role of detecting specific IgE to nArt v 3 in se-
lecting immunotherapy candidates remains rather con-
troversial at the moment.
On the other hand, nArt v 1 is considered a true major
mugwort pollen allergen, accounting for IgE-sensitiza-
tion in approximately 80% of mugwort-pollen-allergic
patients
[59, 60] . Importantly, it is an effective marker of
genuine sensitization to mugwort pollen, even when
cross-reactivity to minor ragweed allergen Amb v 4 oc-
curs
[61] . Furthermore, when major allergen quantifica-
tion is involved in the process of mugwort pollen extract
standardization, Art v 1 is typically used, suggesting a po-
tential increase in SIT efficacy in anti-Art v 1-IgE-positive
patients
[12] .
Finally, commercially marketed nSal k 1-IgE assay
may provide a species-specific marker for primary salt-
wort pollen allergy, which is steadily gaining significance
across extensive areas in southern Europe due to ongoing
desertification
[46, 62] . Quite importantly, detection of
specific IgE to nSal k 1 is able to distinguish the majority
of patients sensitized to saltwort from those sensitized to
closely related chenopod (Chenopodium album) pollen
[63] . As both weeds pollinate at overlapping periods and
often appear double-positive in extract-based diagnos-
tics, identification of the primary sensitizing source may
be of added value, allowing more specific immunothera-
py
[63] .
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
CRM of Allergic Disease in Southern
Europe
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
69
Markers of Cross-Reactivity: Panallergens
In essentially birch-free southern Europe, profilin and
polcalcin molecules are the main cause of in vitro and in
vivo cross-reactivity across a broad spectrum of allergen-
ic pollen. The highly conserved structure and ubiquitous
distribution of these molecules among plant species are
the basis for their ascribed ‘pan-allergenicity’
[64] . Unfor-
tunately, this presents additional challenges, when it
comes to evaluating the sensitization profiles of immuno-
therapy candidates; profilin- and polcalcin-sensitized pa-
tients tend to recognize these molecules in numerous
sources and, as a consequence, score positive in extract-
based diagnostic testing with most pollen species
[65–67] .
Analysis by CRD partially resolves this problem. In fact,
only a limited number of pollen panallergens are available
for routine use (grass profilin, rPhl p 12, and birch pro-
filin, rBet v 2; grass polcalcin, rPhl p 7 and birch polcalcin,
rBet v 4), but due to marked structural homology among
allergenic species, these serve as efficient markers of hy-
persensitivity to the entire group of homologous proteins,
with the possible exception of Parietaria and cypress pro-
filins
[68–72] . The fact that grass pollen is the most prev-
alent cause of both profilin and polcalcin sensitization in
the Mediterranean, along with an excellent cross-reactiv-
ity spectrum of both molecules, favors the use of the
grass-derived allergens over the birch-derived ones, at
least in this geographic region
[66, 68, 70, 73] .
The presence of IgE-sensitization to such panallergens
has been suggested to be a marker of poor outcome in
pollen immunotherapy, especially in the absence of IgE
to species-specific molecules
[16, 20] . In our opinion,
sensitization to pollen profilins and/or polcalcins would
be expected to decrease, at least to some extent, the effi-
cacy of SIT for the following reasons.
i) Whether profilin and polcalcin are clinically rele-
vant allergens is an ongoing matter of debate; recently, it
was demonstrated that they may actually elicit symptoms
in sensitized patients
[74–76] . Moreover, the content in
various pollen SIT extracts, at least for profilin, has been
shown to be remarkably low
[74] , meaning that profilin-
sensitized patients are expected to receive a therapeutic
extract of little immune-modulatory capacity against
profilin.
ii) Sensitization to both profilin and/or polcalcin al-
lergens has been associated with a longer duration of al-
lergic disease and with resulting cosensitization to a larg-
er number of species-specific allergen molecules, com-
pared to patients that do not recognize panallergens
[46,
65, 66]
. This increases the number of genuine sensitiza-
tions to different pollen and would theoretically require
multiple pollen representation for immunotherapy to be
effective.
iii) Sensitization to both profilin and/or polcalcin al-
lergens typically follows previous cosensitization to other
molecular allergens from the same pollen source, as has
been elegantly shown in the grass pollen model
[77, 78] .
It is proposed that, at least for grass pollen allergy, panal-
lergens are only being recognized at the late stage of this
‘molecular spreading’, where the pattern of molecular
sensitization is more complex and the favorable window
of ‘early immunological intervention’ has probably passed
[77, 79] . This is especially relevant, since therapeutic ex-
tracts do not contain all relevant allergens at high doses
[32] ; at best, the concentration of just one major allergen
is ensured
[12] .
In summary, there are theoretical grounds to suggest
that the detection of anti-rPhl p 12-IgE and/or anti-rPhl
p 7-IgE may reduce the anticipated response to pollen
SIT, due to associated molecular multisensitization and
the practical inability to administer an ‘appropriate’, al-
lergen-matching therapeutic extract. However, in case
sIgE against major allergens is present, SIT with extracts
containing these allergens can be administered, especial-
ly as the clinical relevance of profilins and polcalcins is
still arguable.
The Role of Cross-Reactive Carbohydrate
Determinants
Cross-reactive carbohydrate determinants (CCDs) are
not only present in the glycoproteins of virtually all pol-
len, but are regarded as the most frequently encountered
individual epitope structures for IgE
[80] . Nonetheless,
after years of debate about their actual participation in the
induction of symptoms, there is now enough evidence to
support the view that IgE-sensitization to CCDs is clini-
cally insignificant, as these carbohydrates seem to be able
to activate basophils by IgE-binding only ex vivo
[80, 81] .
Either due to insufficient representation, low binding-af-
finity of IgE to carbohydrate epitopes or the presence of
CCD-blocking IgG4 antibodies, glycans do not behave as
allergens in vivo
[80, 81] . In contrast, IgE to CCDs does
become evident by ‘contaminating’ extract-based in vitro
specific-IgE testing; widespread positivity is then the rule
[82] .
If these apparently multisensitized patients undergo
CRD, there is a similar possibility of misleading, false-
positive results when glycosylated component allergens
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
Douladiris/Savvatianos/Roumpedaki/
Skevaki/Mitsias/Papadopoulos
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
70
are utilized. Such CCD-containing natural purified gly-
coproteins include nCyn d 1, nOle e 1, nCup a 1, nSal k 1,
nPla a 2 and nArt v 1, used for the detection of primary
sensitization to Bermuda grass, olive, cypress, saltwort,
plane tree and mugwort pollen, respectively. On the con-
trary, recombinant forms of proteins are typically pro-
duced in the reducing cytoplasmic environment of Esch-
erichia coli bacteria, which is unable to host posttransla-
tional modification, and thus the glycosylation of proteins
[83] . As a result, IgE to CCDs does not interfere with re-
combinant CRD pollen allergens. It must be noted, none-
theless, that the lack of posttranslational modifications in
E. coli -produced recombinant allergens may theoretically
result in improper folding of the allergen protein, leading
to potentially false-negative results due to reduced aller-
gen recognition by the patient IgE
[83] .
When considering CRD results in the formulation of
SIT extracts for apparently multisensitized patients, it is
imperative to exclude potentially CCD-restricted IgE-
sensitization to natural glycosylated species-specific al-
lergens, i.e. the presence of IgE directed only to the carbo-
hydrate moiety in the absence of IgE specific for the pro-
tein fragment. Available markers of sensitization to CCDs
are bromelain (nAna c 2) and MUXF3, the purified N-
glycan from bromelain (on a peptide backbone)
[29] .
These are able to detect IgE to N-glycans in most pollen
sources. When the clinician is confronted with positive in
vitro results to a natural glycoprotein, negative sIgE re-
sults to CCD markers reveal the protein nature of IgE
epitopes. An exception is the case of positivity to nArt v
1; IgE-sensitization to CCD may not be excluded by test-
ing with bromelain or MUXF3, as these markers detect
IgE to N-glycans, whereas nArt v 1 is known to primarily
contain O-glycans
[84] . On the contrary, positive sIgE re-
sults to CCD markers should optimally be accompanied
by assessment of biological activity, i.e. by positive skin-
prick testing or nasal/conjunctival challenge with the al-
lergen source
[80, 81] . This approach may serve to prove
the concomitant presence of sIgE against protein epit-
opes, and thus validate the inclusion of the allergen source
in the SIT regimen.
A Clinical Algorithm for Using CRD in Pollen-Specific
Immunotherapy: Introducing ‘Component-Resolved
Management’
The use of a panel of species-specific allergen markers,
representing the most common allergenic species in the
Mediterranean, along with the panallergen screening
molecules rPhl p 7 and rPhl p 12, may facilitate the selec-
tion of those immunotherapy candidates who would have
an increased probability of benefitting from SIT. A pro-
posed clinical algorithm for the assessment of pollen-al-
lergic patients suitable for extract-based immunotherapy
in the birch-pollen-free areas of southern Europe is de-
picted in figure 1 .
Initially, a basic diagnostic discrimination between
mono/oligo- and multisensitization may be performed,
based on skin test readings and/or sIgE in vitro results us-
ing common pollen extracts. In the case of mono-
or oli-
gosensitization, an individual assessment of patient suit-
ability for SIT is carried out on the basis of sensitization
to major species-specific molecules that are contained in
commercial therapeutical extracts in as close to ‘high
dose’ as possible.
In a multisensitized pollen-allergic patient, an addi-
tional assessment of (co)sensitization to panallergens is
important, as detection of sIgE directed towards profilins
and/or polcalcins would possibly mean a poorer outcome
of immunotherapy for the reasons discussed above. With
respect to species-specific markers, when nonglycosylat-
ed recombinant allergens are used in in vitro tests, posi-
tive sIgE results to major grass-specific rPhl p 5/rPhl p 1,
Parietaria -specific rPar j 2 or olive-specific rOle e 1 may
justify immunotherapy with the respective pollen extract.
In the case of positive in vitro results for Bermuda grass,
cypress, olive, saltwort or mugwort pollen major aller-
gens (nCyn d 1, nCup a 1, nOle e 1, nSal k 1, and nArt v
1 respectively) in multisensitized patients, sensitization
restricted to the CCD component must be optimally
ruled out, as these molecules are used in diagnostics as
purified natural CCD-rich isoforms. As the role of CCDs
in clinical allergy is generally disputed, it would be of
questionable value to specifically target these molecules;
only in the case of concurrent sensitization to a protein
component would immunotherapy to the allergen source
be a prudent choice.
C o n c l u s i o n
We propose a CRM plan for pollinosis in southern
Europe, based on molecular sensitization patterns for the
most common pollens in the area. At the moment, there
are no prospective studies to evaluate the added benefit of
applying CRD in terms of SIT efficacy. However, in a pro-
spective study, it has recently been shown that the incor-
poration of CRD results was able to alter initial SIT indi-
cation and allergen prescription in more than half of the
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
CRM of Allergic Disease in Southern
Europe
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
71
patients studied [85] . Moreover, results from a retrospec-
tive analysis of SIT efficacy with respect to molecular sen-
sitization patterns favor such an approach and are com-
patible with the underlying rationale
[18] .
Of course, for the moment, these remain mainly theo-
retical considerations; significant limitations include in-
complete and sometimes conflicting published data, in-
adequate information about the allergen content of com-
mercial SIT extracts as well as reported qualitative and
quantitative variability between different SIT products
and manufacturers
[12, 31, 86–88] . In addition, taking
into account the limited number of allergen components
currently available for clinical use and the fact that exact
matching of natural SIT extracts to individual sensitiza-
tion profiles may probably be a Utopian goal, one could
reasonably argue against the practical value of such an at-
tempt. However, until patient-tailored recombinant ex-
tracts are licensed for clinical use and until prospective
controlled studies provide solid data on all significant as-
pects, we argue that CRM, based on the best-available sci-
entific knowledge, may enhance our diagnostic and ther-
apeutic accuracy.
Fig. 1. Proposal for a basic CRD work-up for pollen SIT candidates
in southern Europe. For the purposes of the current figure, com-
ponent allergens’ names denote the detection of corresponding
specific IgE. * In cases with nCyn d 1 (+) > rPhl p 1 (+) and nega-
tive species-specific allergen markers for Timothy grass (e.g. rPhl
p 2, rPhl p 5 and rPhl p 6), a Bermuda grass standardized extract
may be sufficient for the treatment of clinical allergy to grass pol-
len (please refer to text). * * Both natural nOle e 1 and recombinant
rOle e 1 are commercially available in Europe. * * * For optimal SIT
efficacy, the use of standardized extracts containing the species-
specific major allergen at high-dose concentration may be re-
quired. * * * * rPhl p 7, as a polcalcin marker and rPhl p 12, as a pro-
filin marker are typically used. In areas with a high prevalence of
birch pollen allergy (not typical of the Mediterranean region), rBet
v 4 and rBet v 2 may respectively be used, along with the essential
rBet v 1 birch pollen major allergen.
Color version available online
Clinical history
SPT/In vitro extract-based slgE assay
Mono-/Oligo-sensitization
Check for major species-specific
allergen markers
rPhl p 1*
rPhl p 5
rPar j 2
rOle e 1**
rPla a 1
Uncertain SIT efficacy
Non-glycosylated
allergen markers
nCyn d 1*
nCup a 1
nOle e1**
nArt v 1
nSal k 1
nPla a 2
Natural purified
glycoproteins
E\²DQWL&&',J(
E\H[WUDFWVNLQWHVWLQJ
E\QDVDOFRQMXQFWLYDODOOHUJHQ
challenge
Assess protein nature of epitope(s)
in multi-sensitized patients
Suitability for SIT***
Check for sensitization
to panallergens****
Multi-sensitization
²
²


SIT efficacy expected
rPhl p 7/12

rPhl p 7/12
²
rPhl p 7/12
²
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
Douladiris/Savvatianos/Roumpedaki/
Skevaki/Mitsias/Papadopoulos
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
72
References
1 Bousquet J, Khaltaev N, Cruz AA, Denburg J,
Fokkens WJ, Togias A, Zuberbier T, Baena-
Cagnani CE, Canonica GW, van Weel C,
Agache I, Ait-Khaled N, Bachert C, Blaiss MS,
Bonini S, Boulet LP, Bousquet PJ, Camargos
P, Carlsen KH, Chen Y, Custovic A, Dahl R,
Demoly P, Douagui H, Durham SR, van Wijk
RG, Kalayci O, Kaliner MA, Kim YY, Kowal-
ski ML, Kuna P, Le LT, Lemiere C, Li J, Lock-
ey RF, Mavale-Manuel S, Meltzer EO, Mo-
hammad Y, Mullol J, Naclerio R, O’Hehir RE,
Ohta K, Ouedraogo S, Palkonen S, Papado-
poulos N, Passalacqua G, Pawankar R, Popov
TA, Rabe KF, Rosado-Pinto J, Scadding GK,
Simons FE, Toskala E, Valovirta E, van Cau-
wenberge P, Wang DY, Wickman M, Yawn
BP, Yorgancioglu A, Yusuf OM, Zar H, Anne-
si-Maesano I, Bateman ED, Ben Kheder A,
Boakye DA, Bouchard J, Burney P, Busse
WW, Chan-Yeung M, Chavannes NH,
Chuchalin A, Dolen WK, Emuzyte R, Grouse
L, Humbert M, Jackson C, Johnston SL, Keith
PK, Kemp JP, Klossek JM, Larenas-
Linnemann D, Lipworth B, Malo JL, Marshall
GD, Naspitz C, Nekam K, Niggemann B, Ni-
zankowska-Mogilnicka E, Okamoto Y, Orru
MP, Potter P, Price D, Stoloff SW, Vanden-
plas O, Viegi G, Williams D: Allergic Rhinitis
and its Impact on Asthma (ARIA) 2008 up-
date (in collaboration with the World Health
Organization, GA(2)LEN and AllerGen). Al-
lergy 2008;
63(suppl 86): 8–160.
2 Masoli M, Fabian D, Holt S, Beasley R: The
global burden of asthma: executive summary
of the GINA Dissemination Committee re-
port. Allergy 2004;
59: 469–478.
3 Woodcock A, Lowe LA, Murray CS, Simpson
BM, Pipis SD, Kissen P, Simpson A, Custovic
A: Early life environmental control: effect on
symptoms, sensitization, and lung function at
age 3 years. Am J Respir Crit Care Med 2004;
170: 433–439.
4 Custovic A, Wijk RG: The effectiveness of
measures to change the indoor environment
in the treatment of allergic rhinitis and asth-
ma: ARIA update (in collaboration with
GA(2)LEN). Allergy 2005;
60: 1112–1115.
5 Papadopoulos NG, Arakawa H, Carlsen KH,
Custovic A, Gern J, Lemanske R, Le Souef P,
Makela M, Roberts G, Wong G, Zar H, Akdis
CA, Bacharier LB, Baraldi E, van Bever HP, de
Blic J, Boner A, Burks W, Casale TB, Castro-
Rodriguez JA, Chen YZ, El-Gamal YM, Eve-
rard ML, Frischer T, Geller M, Gereda J, Goh
DY, Guilbert TW, Hedlin G, Heymann PW,
Hong SJ, Hossny EM, Huang JL, Jackson DJ,
de Jongste JC, Kalayci O, Ait-Khaled N, Kling
S, Kuna P, Lau S, Ledford DK, Lee SI, Liu AH,
Lockey RF, Lodrup-Carlsen K, Lotvall J, Mor-
ikawa A, Nieto A, Paramesh H, Pawankar R,
Pohunek P, Pongracic J, Price D, Robertson
C, Rosario N, Rossenwasser LJ, Sly PD, Stein
R, Stick S, Szefler S, Taussig LM, Valovirta E,
Vichyanond P, Wallace D, Weinberg E,
Wennergren G, Wildhaber J, Zeiger RS: Inter-
national consensus on (ICON) pediatric asth-
ma. Allergy 2012;
67: 976–997.
6 Durham SR, Walker SM, Varga EM, Jacobson
MR, O’Brien F, Noble W, Till SJ, Hamid QA,
Nouri-Aria KT: Long-term clinical efficacy of
grass-pollen immunotherapy. N Engl J Med
1999;
341: 468–475.
7 Eng PA, Reinhold M, Gnehm HP: Long-term
efficacy of preseasonal grass pollen immuno-
therapy in children. Allergy 2002;
57: 306–312.
8 Jacobsen L, Niggemann B, Dreborg S, Fer-
dousi HA, Halken S, Host A, Koivikko A,
Norberg LA, Valovirta E, Wahn U, Moller C:
Specific immunotherapy has long-term pre-
ventive effect of seasonal and perennial asth-
ma: 10-year follow-up on the PAT study. Al-
lergy 2007;
62: 943–948.
9 Pajno GB, Barberio G, De Luca F, Morabito L,
Parmiani S: Prevention of new sensitizations
in asthmatic children monosensitized to
house dust mite by specific immunotherapy.
A six-year follow-up study. Clin Exp Allergy
2001;
31: 1392–1397.
10 Dreborg S, Lee TH, Kay AB, Durham SR: Im-
munotherapy is allergen-specific: a double-
blind trial of mite or Timothy extract in mite
and grass dual-allergic patients. Int Arch Al-
lergy Immunol 2012;
158: 63–70.
11 Larenas-Linnemann D, Esch RE, Guidos-Fo-
gelbach G, Rodriguez-Perez N: A comparison
of in vitro potency between European and
Mexican allergen extracts and US (CBER/
FDA) reference extracts. Allergol Immuno-
pathol (Madr) 2010;
38: 170–173.
12 Larenas-Linnemann D, Cox LS: European al-
lergen extract units and potency: review of
available information. Ann Allergy Asthma
Immunol 2008;
100: 137–145.
13 Larenas-Linnemann D, Esch R, Plunkett G,
Brown S, Maddox D, Barnes C, Constable D:
Maintenance dosing for sublingual immuno-
therapy by prominent European allergen
manufacturers expressed in bioequivalent al-
lergy units. Ann Allergy Asthma Immunol
2011;
107: 448–458 e443.
14 Valenta R, Lidholm J, Niederberger V, Hayek
B, Kraft D, Gronlund H: The recombinant al-
lergen-based concept of component-resolved
diagnostics and immunotherapy (CRD and
CRIT). Clin Exp Allergy 1999;
29: 896–904.
15 Valenta R, Twaroch T, Swoboda I: Compo-
nent-resolved diagnosis to optimize allergen-
specific immunotherapy in the Mediterra-
nean area. J Investig Allergol Clin Immunol
2007;
17(suppl 1):36–40.
16 Asero R: Component-resolved diagnosis-as-
sisted prescription of allergen-specific immu-
notherapy: a practical guide. Eur Ann Allergy
Clin Immunol 2012;
44: 183–187.
17 Pittner G, Vrtala S, Thomas WR, Weghofer
M, Kundi M, Horak F, Kraft D, Valenta R:
Component-resolved diagnosis of house-dust
mite allergy with purified natural and recom-
binant mite allergens. Clin Exp Allergy 2004;
34: 597–603.
18 Schmid-Grendelmeier P: Recombinant aller-
gens. For routine use or still only science?
Hautarzt 2010;
61: 946–953.
19 European Declaration on Allergen Immuno-
therapy. European Academy of Allergy and
Clinical Immunology, 2011. Available from
eaaci.net.
20 Kazemi-Shirazi L, Niederberger V, Linhart B,
Lidholm J, Kraft D, Valenta R: Recombinant
marker allergens: diagnostic gatekeepers for
the treatment of allergy. Int Arch Allergy Im-
munol 2002;
127: 259–268.
21 Niederberger V, Laffer S, Froschl R, Kraft D,
Rumpold H, Kapiotis S, Valenta R, Spitzauer
S: IgE antibodies to recombinant pollen aller-
gens (Phl p 1, Phl p 2, Phl p 5, and Bet v 2)
account for a high percentage of grass pollen-
specific IgE. J Allergy Clin Immunol 1998;
101: 258–264.
22 Johansen N, Weber RW, Ipsen H, Barber D,
Broge L, Hejl C: Extensive IgE cross-reactivity
towards the Pooideae grasses substantiated
for a large number of grass-pollen-sensitized
subjects. Int Arch Allergy Immunol 2009;
150:
325–334.
23 Hejl C, Wurtzen PA, Kleine-Tebbe J, Johan-
sen N, Broge L, Ipsen H: Phleum pratense
alone is sufficient for allergen-specific im-
munotherapy against allergy to Pooideae
grass pollens. Clin Exp Allergy 2009;
39: 752–
759.
24 Moingeon P, Hrabina M, Bergmann KC, Jae-
ger S, Frati F, Bordas V, Peltre G: Specific im-
munotherapy for common grass pollen aller-
gies: pertinence of a five-grass pollen vaccine.
Int Arch Allergy Immunol 2008;
146: 338–
342.
25 Chabre H, Gouyon B, Huet A, Baron-Bodo
V, Nony E, Hrabina M, Fenaille F, Lautrette
A, Bonvalet M, Maillere B, Bordas-Le Floch
V, Van Overtvelt L, Jain K, Ezan E, Batard
T, Moingeon P: Molecular variability of
group 1 and 5 grass pollen allergens be-
tween Pooideae species: implications for
immunotherapy. Clin Exp Allergy 2010;
40:
505–519.
26 Andersson K, Lidholm J: Characteristics and
immunobiology of grass pollen allergens. Int
Arch Allergy Immunol 2003;
130: 87–107.
27 Tripodi S, Frediani T, Lucarelli S, Macri F,
Pingitore G, Di Rienzo Businco A, Dondi A,
Pansa P, Ragusa G, Asero R, Faggian D, Ple-
bani M, Matricardi PM: Molecular profiles of
IgE to Phleum pratense in children with grass
pollen allergy: implications for specific im-
munotherapy. J Allergy Clin Immunol 2012;
129: 834–839 e838.
28 Duffort O, Quintana J, Ipsen H, Barber D,
Polo F: Antigenic similarity among group
1 allergens from grasses and quantitation
ELISA using monoclonal antibodies to Phl p
1. Int Arch Allergy Immunol 2008;
145: 283–
290.
29 Sastre J: Molecular diagnosis in allergy. Clin
Exp Allergy 2010;
40: 1442–1460.
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
CRM of Allergic Disease in Southern
Europe
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
73
30 Calderon MA, Larenas D, Kleine-Tebbe J, Ja-
cobsen L, Passalacqua G, Eng PA, Varga EM,
Valovirta E, Moreno C, Malling HJ, Alvarez-
Cuesta E, Durham S, Demoly P: European
Academy of Allergy and Clinical Immunolo-
gy Task Force report on ‘dose-response rela-
tionship in allergen-specific immunothera-
py’. Allergy 2011;
66: 1345–1359.
31 Sander I, Fleischer C, Meurer U, Bruning T,
Raulf-Heimsoth M: Allergen content of grass
pollen preparations for skin prick testing and
sublingual immunotherapy. Allergy 2009;
64:
1486–1492.
32 Schmidt H, Gelhaus C, Nebendahl M, Janssen
O, Petersen A: Characterization of Phleum
pratense pollen extracts by 2-D DIGE and al-
lergen immunoreactivity. Proteomics 2010;
10: 4352–4362.
33 Quiralte J, Palacios L, Rodriguez R, Cardaba
B, Arias de Saavedra JM, Villalba M, Florido
JF, Lahoz C: Modelling diseases: the allergens
of Olea europaea pollen. J Investig Allergol
Clin Immunol 2007;
17(suppl 1):24–30.
34 Barber D, Moreno C, Ledesma A, Serrano P,
Galan A, Villalba M, Guerra F, Lombardero
M, Rodriguez R: Degree of olive pollen expo-
sure and sensitization patterns. Clinical im-
plications. J Investig Allergol Clin Immunol
2007;
17(suppl 1):11–16.
35 Lombardero M, Obispo T, Calabozo B, Leza-
un A, Polo F, Barber D: Cross-reactivity be-
tween olive and other species. Role of Ole e
1-related proteins. Allergy 2002;
57(suppl
71):29–34.
36 Rodriguez R, Villalba M, Batanero E, Palo-
mares O, Quiralte J, Salamanca G, Sirvent S,
Castro L, Prado N: Olive pollen recombinant
allergens: value in diagnosis and immuno-
therapy. J Investig Allergol Clin Immunol
2007;
17(suppl 1):4–10.
37 Asero R: Analysis of hypersensitivity to ole-
aceae pollen in an olive-free and ash-free area
by commercial pollen extracts and recombi-
nant allergens. Eur Ann Allergy Clin Immu-
nol 2011;
43: 77–80.
38 Palomares O, Swoboda I, Villalba M, Balic N,
Spitzauer S, Rodriguez R, Valenta R: The ma-
jor allergen of olive pollen Ole e 1 is a diagnos-
tic marker for sensitization to Oleaceae. Int
Arch Allergy Immunol 2006;
141: 110–118.
39 Aceituno E, Del Pozo V, Minguez A, Arrieta
I, Cortegano I, Cardaba B, Gallardo S, Rojo M,
Palomino P, Lahoz C: Molecular cloning of
major allergen from Cupressus arizonica pol-
len: Cup a 1. Clin Exp Allergy 2000;
30: 1750–
1758.
40 Charpin D, Gouitaa M, Dron-Gonzalvez M,
Fardeau MF, Massabie-Bouchat YP, Hugues
B, Fabre C, Vivinus S, Pegliasco H, Andre C:
Immunotherapy with an aluminum hydrox-
ide-adsorbed Juniperus ashei foreign pollen
extract in seasonal indigenous cypress pollen
rhinoconjunctivitis. A double-blind, placebo-
controlled study. Int Arch Allergy Immunol
2007;
143: 83–91.
41 Asturias JA, Ibarrola I, Bartolome B, Ojeda I,
Malet A, Martinez A: Purification and charac-
terization of Pla a 1, a major allergen from
Platanus acerifolia pollen. Allergy 2002;
57:
221–227.
42 Ibarrola I, Arilla MC, Martinez A, Asturias
JA: Identification of a polygalacturonase as a
major allergen (Pla a 2) from Platanus aceri-
folia pollen. J Allergy Clin Immunol 2004;
113:
1185–1191.
43 Lauer I, Miguel-Moncin MS, Abel T, Foetisch
K, Hartz C, Fortunato D, Cistero-Bahima A,
Vieths S, Scheurer S: Identification of a plane
pollen lipid transfer protein (Pla a 3) and its
immunological relation to the peach lipid-
transfer protein, Pru p 3. Clin Exp Allergy
2007;
37: 261–269.
44 D’Amato G, Liccardi G: Pollen-related allergy
in the European Mediterranean area. Clin
Exp Allergy 1994;
24: 210–219.
45 D’Amato G, Cecchi L, Bonini S, Nunes C, An-
nesi-Maesano I, Behrendt H, Liccardi G, Pop-
ov T, van Cauwenberge P: Allergenic pollen
and pollen allergy in Europe. Allergy 2007;
62:
976–990.
46 Barber D, de la Torre F, Feo F, Florido F,
Guardia P, Moreno C, Quiralte J, Lombardero
M, Villalba M, Salcedo G, Rodriguez R: Un-
derstanding patient sensitization profiles in
complex pollen areas: a molecular epidemio-
logical study. Allergy 2008;
63: 1550–1558.
47 Asturias JA, Gomez-Bayon N, Eseverri JL,
Martinez A: Par j 1 and Par j 2, the major al-
lergens from Parietaria judaica pollen, have
similar immunoglobulin E epitopes. Clin Exp
Allergy 2003;
33: 518–524.
48 Stumvoll S, Westritschnig K, Lidholm J, Spit-
zauer S, Colombo P, Duro G, Kraft D, Geraci
D, Valenta R: Identification of cross-reactive
and genuine Parietaria judaica pollen aller-
gens. J Allergy Clin Immunol 2003;
111: 974–
979.
49 Tordesillas L, Sirvent S, Diaz-Perales A, Vil-
lalba M, Cuesta-Herranz J, Rodriguez R, Sal-
cedo G: Plant lipid transfer protein allergens:
no cross-reactivity between those from foods
and olive and Parietaria pollen. Int Arch Al-
lergy Immunol 2011;
156: 291–296.
50 Salcedo G, Sanchez-Monge R, Barber D, Di-
az-Perales A: Plant non-specific lipid transfer
proteins: an interface between plant defence
and human allergy. Biochim Biophys Acta
2007;
1771: 781–791.
51 Gadermaier G, Harrer A, Girbl T, Palazzo P,
Himly M, Vogel L, Briza P, Mari A, Ferreira
F: Isoform identification and characterization
of Art v 3, the lipid-transfer protein of mug-
wort pollen. Mol Immunol 2009;
46: 1919–
1924.
52 Rossi RE, Monasterolo G, Coco G, Operti D:
Possible relationship between systemic side
effects and sensitization to rPar j 2 in allergic
patients submitted to an ultra-rush (20 min)
sublingual immunotherapy and selected by
component-resolved diagnosis. Int Arch Al-
lergy Immunol 2005;
138: 105–110.
53 Pastorello EA, Pravettoni V, Farioli L, Rivolta
F, Conti A, Ispano M, Fortunato D, Bengtsson
A, Bianchi M: Hypersensitivity to mugwort
(Artemisia vulgaris) in patients with peach al-
lergy is due to a common lipid transfer pro-
tein allergen and is often without clinical ex-
pression. J Allergy Clin Immunol 2002;
110:
310–317.
54 Pascal M, Munoz-Cano R, Reina Z, Palacin A,
Vilella R, Picado C, Juan M, Sanchez-Lopez J,
Rueda M, Salcedo G, Valero A, Yague J, Bartra
J: Lipid transfer protein syndrome: clinical
pattern, cofactor effect and profile of molecu-
lar sensitization to plant foods and pollens.
Clin Exp Allergy 2012;
42: 1529–1539.
55 Asero R, Pravettoni V: Anaphylaxis to plant
foods and pollen allergens in patients with
lipid transfer protein syndrome. Curr Opin
Allergy Clin Immunol 2013, E-pub ahead of
print.
56 Asero R: Co-recognition of lipid trasfer pro-
tein in pollen and foods in northern Italy: cli-
nician’s view. Eur Ann Allergy Clin Immunol
2010;
42: 205–208.
57 Gao ZS, Yang ZW, Wu SD, Wang HY, Liu
ML, Mao WL, Wang J, Gadermaier G, Fer-
reira F, Zheng M, van Ree R: Peach allergy in
China: a dominant role for mugwort pollen
lipid transfer protein as a primary sensitizer.
J Allergy Clin Immunol 2013;
131: 224–226
e223.
58 Lombardero M, Garcia-Selles FJ, Polo F, Ji-
meno L, Chamorro MJ, Garcia-Casado G,
Sanchez-Monge R, Diaz-Perales A, Salcedo G,
Barber D: Prevalence of sensitization to Arte-
misia allergens Art v 1, Art v 3 and Art v 60
kDa. Cross-reactivity among Art v 3 and oth-
er relevant lipid-transfer protein allergens.
Clin Exp Allergy 2004;
34: 1415–1421.
59 Oberhuber C, Ma Y, Wopfner N, Gadermaier
G, Dedic A, Niggemann B, Maderegger B, Gr-
uber P, Ferreira F, Scheiner O, Hoffmann-
Sommergruber K: Prevalence of IgE-binding
to Art v 1, Art v 4 and Amb a 1 in mugwort-
allergic patients. Int Arch Allergy Immunol
2008;
145: 94–101.
60 Gadermaier G, Wopfner N, Wallner M, Egger
M, Didierlaurent A, Regl G, Aberger F, Lang
R, Ferreira F, Hawranek T: Array-based pro-
filing of ragweed and mugwort pollen aller-
gens. Allergy 2008;
63: 1543–1549.
61 Leonard R, Wopfner N, Pabst M, Stadlmann
J, Petersen BO, Duus JO, Himly M, Radauer
C, Gadermaier G, Razzazi-Fazeli E, Ferreira F,
Altmann F: A new allergen from ragweed
(Ambrosia artemisiifolia) with homology to
art v 1 from mugwort. J Biol Chem 2010;
285:
27192–27200.
62 Arilla MC, Ibarrola I, Brena S, Martinez A,
Colas C, Asturias JA: The Russian thistle (Sal-
sola kali) pollen major allergen, Sal k 1, can be
quantified in allergenic extracts and airborne
pollen. Int Arch Allergy Immunol 2010;
152:
319–326.
63 Barderas R, Garcia-Selles J, Salamanca G, Co-
las C, Barber D, Rodriguez R, Villalba M: A
pectin methylesterase as an allergenic marker
for the sensitization to Russian thistle (Salsola
kali) pollen. Clin Exp Allergy 2007;
37: 1111–
1119.
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
Douladiris/Savvatianos/Roumpedaki/
Skevaki/Mitsias/Papadopoulos
Int Arch Allergy Immunol 2013;162:65–74
DOI: 10.1159/000353113
74
64 Hauser M, Roulias A, Ferreira F, Egger M: Pa-
nallergens and their impact on the allergic
patient. Allergy Asthma Clin Immunol 2010;
6: 1.
65 Barber D, de la Torre F, Lombardero M, An-
tépara I, Colas C, Dávila I, Tabar AI, Vidal C,
Villalba M, Salcedo G, Rodríguez R: Compo-
nent-resolved diagnosis of pollen allergy
based on skin testing with profilin, polcalcin
and lipid transfer protein pan-allergens. Clin
Exp Allergy 2009;
39: 1764–1773.
66 Villalta D, Asero R: Analysis of the allergenic
profile of patients hypersensitive to pollen
pan-allergens living in two distinct areas of
northern Italy. Eur Ann Allergy Clin Immu-
nol 2011;
43: 54–57.
67 Mari A: Multiple pollen sensitization: a mo-
lecular approach to the diagnosis. Int Arch
Allergy Immunol 2001;
125: 57–65.
68 Asero R, Monsalve R, Barber D: Profilin sen-
sitization detected in the office by skin prick
test: a study of prevalence and clinical rele-
vance of profilin as a plant-food allergen. Clin
Exp Allergy 2008;
38: 1033–1037.
69 Asero R, Mistrello G, Roncarolo D, Amato
S: Parietaria profilin shows only limited
cross-reactivity with birch and grass profil-
ins. Int Arch Allergy Immunol 2004;
133:
121–124.
70 Santos A, Van Ree R: Profilins: mimickers of
allergy or relevant allergens? Int Arch Allergy
Immunol 2011;
155: 191–204.
71 Grote M, Westritschnig K, Valenta R: Immu-
nogold electron microscopic localization of
the 2 EF-hand calcium-binding pollen aller-
gen Phl p 7 and its homologues in pollens of
grasses, weeds and trees. Int Arch Allergy Im-
munol 2008;
146: 113–121.
72 Ledesma A, Barderas R, Westritschnig K,
Quiralte J, Pascual CY, Valenta R, Villalba M,
Rodriguez R: A comparative analysis of the
cross-reactivity in the polcalcin family includ-
ing Syr v 3, a new member from lilac pollen.
Allergy 2006;
61: 477–484.
73 Tinghino R, Twardosz A, Barletta B, Puggioni
EMR, Iacovacci P, Butteroni C, Afferni C,
Mari A, Hayek B, Di Felice G, Focke M, Wes-
tritschnig K, Valenta R, Pini C: Molecular,
structural, and immunologic relationships
between different families of recombinant
calcium-binding pollen allergens. J Allergy
Clin Immunol 2002;
109: 314–320.
74 Ruiz-Garcia M, Garcia Del Potro M, Fernan-
dez-Nieto M, Barber D, Jimeno-Nogales L,
Sastre J: Profilin: a relevant aeroallergen? J Al-
lergy Clin Immunol 2011;
128: 416–418.
75 Asero R, Jimeno L, Barber D: Preliminary re-
sults of a skin prick test-based study of the
prevalence and clinical impact of hypersensi-
tivity to pollen panallergens (polcalcin and
profilin). J Investig Allergol Clin Immunol
2010;
20: 35–38.
76 Nunez R, Carballada F, Lombardero M, Jime-
no L, Boquete M: Profilin as an aeroallergen
by means of conjunctival allergen challenge
with purified date palm profilin. Int Arch Al-
lergy Immunol 2012;
158: 115–119.
77 Hatzler L, Panetta V, Lau S, Wagner P, Berg-
mann RL, Illi S, Bergmann KE, Keil T, Hof-
maier S, Rohrbach A, Bauer CP, Hoffman U,
Forster J, Zepp F, Schuster A, Wahn U, Mat-
ricardi PM: Molecular spreading and predic-
tive value of preclinical IgE response to Phle-
um pratense in children with hay fever. J Al-
lergy Clin Immunol 2012;
130: 894–901 e895.
78 Mari A: Skin test with a Timothy grass (Phle-
um pratense) pollen extract vs. IgE to a Timo-
thy extract vs. IgE to rPhl p 1, rPhl p 2, nPhl p
4, rPhl p 5, rPhl p 6, rPhl p 7, rPhl p 11, and
rPhl p 12: epidemiological and diagnostic
data. Clin Exp Allergy 2003;
33: 43–51.
79 Valenta R: The future of antigen-specific im-
munotherapy of allergy. Nat Rev Immunol
2002;
2: 446–453.
80 Altmann F: The role of protein glycosylation
in allergy. Int Arch Allergy Immunol 2007;
142: 99–115.
81 Mari A, Ooievaar-de Heer P, Scala E, Giani M,
Pirrotta L, Zuidmeer L, Bethell D, van Ree R:
Evaluation by double-blind placebo-con-
trolled oral challenge of the clinical relevance
of IgE antibodies against plant glycans. Aller-
gy 2008;
63: 891–896.
82 Malandain H, Giroux F, Cano Y: The influ-
ence of carbohydrate structures present in
common allergen sources on specific IgE re-
sults. Eur Ann Allergy Clin Immunol 2007;
39:
216–220.
83 Demain AL, Vaishnav P: Production of re-
combinant proteins by microbes and higher
organisms. Biotechnol Adv 2009;
27: 297–306.
84 Leonard R, Petersen BO, Himly M, Kaar W,
Wopfner N, Kolarich D, van Ree R, Ebner C,
Duus JO, Ferreira F, Altmann F: Two novel
types of O-glycans on the mugwort pollen al-
lergen Art v 1 and their role in antibody bind-
ing. J Biol Chem 2005;
280: 7932–7940.
85 Sastre J, Landivar ME, Ruiz-Garcia M, An-
dregnette-Rosigno MV, Mahillo I: How mo-
lecular diagnosis can change allergen-specific
immunotherapy prescription in a complex
pollen area. Allergy 2012;
67: 709–711.
86 Larenas-Linnemann D, Cruz AA, Gutierrez
IR, Rodriguez P, Shah-Hosseini K, Michels A,
Mosges R: European and Mexican vs. US di-
agnostic extracts of Bermuda grass and cat in
skin testing. Ann Allergy Asthma Immunol
2011;
106: 421–428.
87 Duffort O, Palomares O, Lombardero M, Vil-
lalba M, Barber D, Rodriguez R, Polo F: Vari-
ability of Ole e 9 allergen in olive pollen ex-
tracts: relevance of minor allergens in immu-
notherapy treatments. Int Arch Allergy
Immunol 2006;
140: 131–138.
88 Casset A, Mari A, Purohit A, Resch Y, Weg-
hofer M, Ferrara R, Thomas WR, Alessandri
C, Chen KW, de Blay F, Valenta R, Vrtala S:
Varying allergen composition and content af-
fects the in vivo allergenic activity of commer-
cial Dermatophagoides pteronyssinus extracts.
Int Arch Allergy Immunol 2012;
159: 253–262.
Downloaded by:
UCLA Biomedical Library
128.97.245.9 - 8/11/2013 8:07:37 PM
... The main reason of the increase in the incidence of pollen and the prevalence of its medium and heavy forms is the leadership of symptomatic pharmacotherapy over pathogenetic allergen-specific immunotherapy (ASIT) The most effective method of treatment for allergic diseases is allergen-specific immunotherapy, which affects all pathogenetic links of the allergic process and has a long-term preventive effect after the completion of treatment courses ( Only in the last two decades, these methods began to be widely used first in European countries, later in North and Central America, and in the last years methods are already used in Kazakhstan. A comparative study of the efficacy of oral and parenteral allergen-specific immunotherapy (ASIT) has not been adequately studied, mainly in European countries (Cheryl et al., 2013, Douladiris, 2013Kudabayeva et al., 2014;Kudabayeva et al., 2017;Valenta et al., 2014). In our republic, similar results of the study were not previously published. ...
... In the course of the work, 153 (67.1%) patients from the 1st group (SLIT) 95 (75.4%), from the 2nd group (PIT) 58 (56.9%) patients with pollinosis were examined by allergic molecular diagnostics and having positive skin scratch tests for native allergens with polysensitivity to mixes of weeds, meadow grasses, and trees (Douladiris, 2013). ...
... In both cases, ITA was recommended according to the main guidelines that were prevailing during the study period. [17][18][19] ...
Article
Full-text available
Background: Recognition of specific allergens triggering immune response is key for the appropriate prescription of allergen-specific immunotherapy (SIT). This study aimed at evaluating the impact of using the commercially available microarray ImmunoCAPTM ISAC 112 (Thermo Fisher Scientific) on the etiological diagnosis and SIT prescription compared to the conventional diagnostic methods in patients with allergic rhinitis/rhinoconjunctivitis and/or asthma. Methods: 300 patients with respiratory allergic disease, sensitized to three or more pollen aeroallergens from different species, as assessed by a skin prick test (SPT) and specific IgE assays (sIgE), were included in this multicentric, prospective observational study. SPT and a blood test were performed to all patients. Total serum IgE and sIgE (ImmunoCAPTM) for allergens found positive in the SPT and sIgE allergen components (ImmunoCAPTM ISAC 112) were measured. Results: According to SPT results, the most prevalent pollen sensitizers in our population were Olea europaea followed by grass, Platanus acerifolia and Parietaria judaica. The molecular diagnosis (MD) revealed Ole e 1 as the most prevalent pollen sensitizer, followed by Cup a 1, Phl p 1, Cyn d 1, Par j 2, Pla a 1, 2, and 3 and Phl p 5. Immunotherapy prescription changed, due to MD testing, in 51% of the cases, with an increase of prescription of SIT from 39% to 65%. Conclusion: The identification of the allergen eliciting the respiratory disease is essential for a correct immunotherapy prescription. The advances in allergen characterization using methods, such as the commercial microarray ImmunoCAPTM ISAC 112, can help clinicians to improve SIT prescription.
... In particular, the list of allergens to be included in an AIT treatment was modified by the identification of sensitisation to a genuine allergen or of sensitisation to one or more cross-reacting components. As a result of these studies, Douladiris et al. 8 described an algorithm that indicated the best AIT approach, which considered sensitisations to genuine and crossreacting components. Notably, this procedure also considered the possibility of false positives due to the presence of cross-reactive carbohydrate determinants (CCD) in the 'natural highly purified' component used in the assay. ...
Article
Full-text available
Emerging technologies are profoundly changing the landscape of allergy diagnoses and future allergy treatments. At the single patient level, the introduction of single components and allergen microarrays for allergy diagnoses has significantly modified treatment strategies. In epidemiological terms, the availability of information from large dataset analyses has allowed and, more importantly, will allow for changes in prophylaxis and treatment strategies in many patients. In this report, we describe the different fields where new technologies have had a significant effect on allergy management and identify new scenarios where the combination of data from basic, clinical, and epidemiological research will improve our knowledge of allergy diagnosis and treatment.
... In pollen allergy treatment, the content of commercial pollen allergen immunotherapy (AIT) extracts is not always known, but the presence of major allergens is often stated (5,6). Molecular diagnosis can improve allergy diagnosis (7), selecting patients eligible for AIT and allowing an accurate individual prescription, which will avoid inappropriate or ineffective therapies (8). ...
Article
Pollens are the main cause of respiratory allergies which prevalence is increasing. The most important cause of pollinosis in Europe and especially in the Mediterranean countries as Portugal is Poaceae family pollen. Timothy grass (Phleum pratense) is one of the most common pollen sources, and one of the best characterized allergenic grasses. The major allergens Phl p1 and Phl p5 are considered markers of genuine grass pollen sensitization. A characterization of Phl p1 and Phl p5 sensitization in the North-Central region of Portugal was made in children and adults. Phl p1 sensitization was the most frequent.
... It has been shown that continuous pollen monitoring could protect allergic patients from exposure and prophylactic medication, thus reducing the referral to medical centers by 50%. 43 Accordingly, evidence on seasonal aeroallergen levels can help generate an alarm system for sensitized individuals and doctors. ...
Article
Full-text available
Introduction and objectives: Pollen calendars have been proved clinically important in allergic disease management, as differences in airborne allergens exist not only between different countries but also between regions of the same country as well. This study aims to provide new data regarding the atmospheric pollen and fungi content of a Mediterranean region (Western Thrace/North-East Greece) and discuss seasonal trends, differences in pollen grains and fungi spores' circulation over the years, and correlations with climate parameters. Material and methods: A 7-day continuously running volumetric trap was used to collect circulating pollen grains and fungi spores. Pollen taxa and fungi were characterized by standard protocols and counted as grains /m3 and average total grains and spores. The primary allergenic pollen season was discovered, and their 10-day averages were measured over time. Correlations with temperature, rain, and humidity were assessed by single linear regression analysis. Results: Pollen grains from nine pollen families were identified, including five arboreal, two nonarboreal taxa, and spores from two fungi species. The three most prevalent taxa were Oleaceae, Poaceae, Pinaceae, and Cladosporium in the fungi. Peak pollen concentrations were detected during April and May, with daily averages exceeding 170 grains/m3. Poaceae presented the longest pollen season of 342 days and Oleaceae the shortest, extending to only 110 days, during the 3years. Cladosporium was the fungus with the highest spore concentration (180,129.9 spores) compared with Alternaria (28,026.1 spores). Correlations with meteorological parameters showed variable associations among different taxa, with rainfall and relative humidity being the most significant determinants. Conclusion: In this study, the pollenic and fungal spectrum of a Mediterranean region and information that can be proved clinically significant for the appropriate diagnostic and therapeutic approach of allergic patients was presented.
... Using both SPT and compound-specific IgE basically determines the same values with two highly overlapping diagnostic tools. This is, of course, not valid for the determination of monoclonal specific IgE, for which there is a good rationale supporting its use as a tool for confirmation and further diagnostic differentiation and for determining the usefulness and effectiveness of subcutaneous immunotherapy [30,31]. Symptoms reported by patients are very variable especially in a setting with polysensitized subjects where the point in time reported by a patient does not always coincide with the underlying multiple sensitizations. ...
Article
Full-text available
Background: Diagnosis of allergic rhinitis is achieved by a combination of patient history and different screening tools, followed by specific provocation testing. Screening tools usually involve a skin prick test (SPT), specific serum IgE or a combination of both. Objective: The purpose of this study was to evaluate the correlation of SPT, intradermal testing and specific serum IgE testing in certain allergens and to evaluate sensitization rates, symptom patterns and time of symptoms in a cohort of patients with suspected allergic rhinitis. Methods: Data on 4653 patients with suspected allergic rhinitis were included and divided into five groups: spring bloomers (birch, hazel, etc.), summer bloomers (grasses and rye), autumn bloomers (ribwort and mugwort), mites and mold. Correlation of SPT, intradermal testing and specific IgE test results using Cohen’s kappa and logistic regression were carried out to evaluate the probability of symptoms. Results: Comparison of SPT and specific serum IgE led to kappa coefficients between 0.33 and 0.47, corresponding to a minor to moderate concordance. Comparing the symptoms reported by patients with sensitization diagnosed by SPT, a correlation was only found for spring and summer bloomers with an odds ratio of 1.5 and 2.1, respectively. The most prevalent symptom in the study cohort was rhinitis, followed by others such as asthma, sense of smell and atopic dermatitis. Conclusions: SPT seems to be more sensitive than specific IgE for detection of sensitization. Patients’ symptoms as well as the timing of symptoms, especially for perennial allergies, are not always very pronounced.
... This is particularly true when allergen immunotherapy (AIT) is provided to the patient. Even though large discussions on this topic are still ongoing, suggestive data seems to indicate that AIT is probably more effective in patients sensitized to genuine allergens, [38][39][40][41][42][43]42,43 while its activity is less impressive in patients with a sensitization to cross-reacting components or pan-allergens. In this context, the in vitro evidence of specific IgE involvement in the patient's symptoms associated with the detection of IgE specific for genuine components seems to be the entry level diagnostic for patients that could have a real modification of their allergy by AIT. ...
... 309 However, molecular-based algorithms have been proposed that would optimize the prescription. [310][311][312] These algorithms all incorporate the idea that detecting sensitization to genuine components is essential. 313 Although AIT for pollen-associated food allergy has shown beneficial effects in some studies, particularly for oral symptoms, other clinical trials have not shown similar outcomes; thus AIT cannot be recommended as a treatment in these cases and should be considered only when respiratory symptoms are present. ...
Article
Full-text available
Since the discovery of immunoglobulin E (IgE) as a mediator of allergic diseases in 1967, our knowledge about the immunological mechanisms of IgE‐mediated allergies has remarkably increased. In addition to understanding the immune response and clinical symptoms, allergy diagnosis and management depend strongly on the precise identification of the elicitors of the IgE‐mediated allergic reaction. In the past four decades, innovations in bioscience and technology have facilitated the identification and production of well‐defined, highly pure molecules for component‐resolved diagnosis (CRD), allowing a personalized diagnosis and management of the allergic disease for individual patients. The first edition of the “EAACI Molecular Allergology User's Guide” (MAUG) in 2016 rapidly became a key reference for clinicians, scientists, and interested readers with a background in allergology, immunology, biology, and medicine. Nevertheless, the field of molecular allergology is moving fast, and after 6 years, a new EAACI Taskforce was established to provide an updated document. The Molecular Allergology User's Guide 2.0 summarizes state‐of‐the‐art information on allergen molecules, their clinical relevance, and their application in diagnostic algorithms for clinical practice. It is designed for both, clinicians and scientists, guiding health care professionals through the overwhelming list of different allergen molecules available for testing. Further, it provides diagnostic algorithms on the clinical relevance of allergenic molecules and gives an overview of their biology, the basic mechanisms of test formats, and the application of tests to measure allergen exposure.
Article
Full-text available
Background: Diagnosis and immunotherapy of house-dust mite (HDM) allergy is still based on natural allergen extracts. The aim of this study was to analyze commercially available Dermatophagoides pteronyssinus extracts from different manufacturers regarding allergen composition and content and whether variations may affect their allergenic activity. Methods: Antibodies specific for several D. pteronyssinus allergens (Der p 1, 2, 5, 7, 10 and 21) were used to analyze extracts from 10 different manufacturers by immunoblotting. Sandwich ELISAs were used to quantify Der p 1 and Der p 2 in the extracts. Mite-allergic patients (n = 45) were skin-tested with the extracts and tested for immunoglobulin E (IgE) reactivity to a panel of 10 mite allergens (Der p 1, 2, 4, 5, 7, 8, 10, 14, 20 and 21) by dot blot. Results: Only Der p 1 and Der p 2 were detected in all extracts but their concentrations and ratios showed high variability (Der p 1: 6.0-40.8 µg ml(-1); Der p 2: 1.7-45.0 µg ml(-1)). At least 1 out of 4 allergens (i.e. Der p 5, 7, 10 and 21) was not detected in 8 of the studied extracts. Mite-allergic subjects showed different IgE reactivity profiles to the individual mite allergens, the extracts showed different allergenic activity in skin-prick tests and false-negative results. Conclusions: Commercially available D. pteronyssinus extracts lack important allergens, show great variability regarding allergen composition and content and some gave false-negative diagnostic test results in certain patients.
Article
Purpose of review: Nonspecific lipid transfer protein (LTP) is the main cause of primary food allergy in adults living in the Mediterranean area. The way allergic patients get sensitized to this protein is all but established, and the clinical expression of sensitization is extremely variable, ranging from long-lasting symptomless sensitization to severe anaphylaxis. Such variability is seemingly due to the presence/absence of a number of cofactors. Recent findings: The possibility that LTP sensitization occurs via the inhalation of LTP-containing pollen particles seems unlikely; in contrast, peach particles containing the protein seem able to sensitize both via the airways and the skin. Cosensitization to pollen allergens as well as to labile plant food allergens makes LTP allergy syndrome less severe. In some LTP sensitized subjects clinical food allergy occurs only in the presence of cofactors such as exercise, NSAIDs, or chronic urticaria. Summary: Lipid transfer protein allergy syndrome shows some peculiarities that are unique in the primary food allergy panorama: geographical distribution, frequent asymptomatic sensitization, frequent need for cofactors, and reduced severity when pollen allergy is present. Future studies will have to address these points as the results may have favorable effects on other, more severe, types of food allergy.
Article
Allergen specific immunotherapy remains the only means to change the natural history of allergic disease. Thanks to the recent advances in molecular biology a large spectrum of purified allergen molecules are presently routinely available for diagnostic purposes. This review represents a practical guide on how to use these new diagnostic tools in order to detect precisely the primary sensitizing allergen sources in subjects showing a multiple sensitization to seasonal and/or perennial airborne allergens, thus avoiding the diagnostic mistakes that have been probably associated with the prescription and administration of several ineffective immunotherapies up to a recent past.
Article
Multiple plant-food sensitizations with a complex pattern of clinical manifestations are a common feature of lipid transfer protein (LTP)-allergic patients. Component-resolved diagnosis permits the diagnosis of the allergen sensitization profile. We sought to clinically characterize and describe the plant-food and pollen molecular sensitization profile in patients with LTP syndrome. Forty-five subjects were recruited, after being diagnosed with multiple plant-food allergies sensitized to LTP, but not to any other plant-food allergen, according to the molecular allergen panel tested (Pru p 3 (LTP), Pru p 1 (Bet v 1-like), Pru p 4 (profilin) and those included in a commercial microarray of 103 allergenic components). IgE-mediated food-allergy symptoms and pollinosis were collected. Patients were skin prick tested with a plant-food and pollens panel, and specific IgE to Tri a 14 was evaluated. A heterogeneous group of plant-foods was involved in local and systemic symptoms: oral allergy syndrome (75.6%), urticaria (66.7%), gastrointestinal disorders (55.6%) and anaphylaxis (75.6%), 32.4% of which were cofactor dependent (Non-Steroidal Anti-inflammatory Drugs, exercise). All tested subjects were positive to peach and Pru p 3, Tri a 14 and to some of the LTPs included in the microarray. Pollinosis was diagnosed in 75.6% of subjects, with a broad spectrum of pollen and pollen-allergen sensitization. Plane tree and mugwort were the statistically significant pollens associated with Pru p 3. Several plant-foods, taxonomically unrelated, independent of peach involvement, are implicated in LTP syndrome. Local symptoms should be evaluated as a risk marker for anaphylaxis because they are frequently associated with cofactor-dependent anaphylaxis. The association of these symptoms with pollinosis, especially plane tree pollinosis, could be part of this syndrome in our area.
Article
IgE sensitization against grass pollen is a cause of seasonal allergic rhinitis. We sought to investigate the evolution at the molecular level and the preclinical predictive value of IgE responses against grass pollen. The German Multicentre Allergy Study examined a birth cohort born in 1990. A questionnaire was administered yearly, and blood samples were collected at 1, 2, 3, 5, 6, 7, 10, and 13 years of age. Grass pollen-related seasonal allergic rhinitis (SARg) was diagnosed according to nasal symptoms in June/July. Serum IgE antibodies to Phleum pratense extract and 8 P pratense molecules were tested with immune-enzymatic singleplex and multiplex assays, respectively. One hundred seventy-seven of the 820 examined children had SARg. A weak monomolecular/oligomolecular IgE response to P pratense was observed very frequently before SARg onset. These initial IgE responses increased in concentration and molecular complexity during the preclinical and clinical process. A typical progression of IgE sensitization was observed: Phl p 1 (initiator in >75% of cases); then Phl p 4 and Phl p 5; then Phl p 2, Phl p 6, and Phl p 11; and then Phl p 12 and Phl p 7. At age 3 years, IgE sensitization predicted SARg by age 12 years (positive predictive value, 68% [95% CI, 50% to 82%]; negative predictive value, 84% [95% CI, 80% to 87%]). At this preclinical prediction time, the number of recognized molecules and the serum levels of IgE to P pratense were significantly lower than at 3 or more years after SARg onset. The IgE response against grass pollen molecules can start years before disease onset as a weak monosensitization or oligosensitization phenomenon. It can increase in serum concentration and complexity through a "molecular spreading" process during preclinical and early clinical disease stages. Testing IgE sensitization at a preclinical stage facilitates prediction of seasonal allergic rhinitis at its molecular monosensitization or oligosensitization stage.
Article
Allergic rhinitis is a symptomatic disorder of the nose induced after allergen exposure by an IgE-mediated inflammation of the membranes lining the nose. It is a global health problem that causes major illness and disability worldwide. Over 600 million patients from all countries, all ethnic groups and of all ages suffer from allergic rhinitis. It affects social life, sleep, school and work and its economic impact is substantial. Risk factors for allergic rhinitis are well identified. Indoor and outdoor allergens as well as occupational agents cause rhinitis and other allergic diseases. The role of indoor and outdoor pollution is probably very important, but has yet to be fully understood both for the occurrence of the disease and its manifestations. In 1999, during the Allergic Rhinitis and its Impact on Asthma (ARIA) WHO workshop, the expert panel proposed a new classification for allergic rhinitis which was subdivided into 'intermittent' or 'persistent' disease. This classification is now validated. The diagnosis of allergic rhinitis is often quite easy, but in some cases it may cause problems and many patients are still under-diagnosed, often because they do not perceive the symptoms of rhinitis as a disease impairing their social life, school and work. The management of allergic rhinitis is well established and the ARIA expert panel based its recommendations on evidence using an extensive review of the literature available up to December 1999. The statements of evidence for the development of these guidelines followed WHO rules and were based on those of Shekelle et al. A large number of papers have been published since 2000 and are extensively reviewed in the 2008 Update using the same evidence-based system. Recommendations for the management of allergic rhinitis are similar in both the ARIA workshop report and the 2008 Update. In the future, the GRADE approach will be used, but is not yet available. Another important aspect of the ARIA guidelines was to consider co-morbidities. Both allergic rhinitis and asthma are systemic inflammatory conditions and often co-exist in the same patients. In the 2008 Update, these links have been confirmed. The ARIA document is not intended to be a standard-of-care document for individual countries. It is provided as a basis for physicians, health care professionals and organizations involved in the treatment of allergic rhinitis and asthma in various countries to facilitate the development of relevant local standard-of-care documents for patients.
Article
Asthma is the most common chronic lower respiratory disease in childhood throughout the world. Several guidelines and/or consensus documents are available to support medical decisions on pediatric asthma. Although there is no doubt that the use of common systematic approaches for management can considerably improve outcomes, dissemination and implementation of these are still major challenges. Consequently, the International Collaboration in Asthma, Allergy and Immunology (iCAALL), recently formed by the EAACI, AAAAI, ACAAI, and WAO, has decided to propose an International Consensus on (ICON) Pediatric Asthma. The purpose of this document is to highlight the key messages that are common to many of the existing guidelines, while critically reviewing and commenting on any differences, thus providing a concise reference. The principles of pediatric asthma management are generally accepted. Overall, the treatment goal is disease control. To achieve this, patients and their parents should be educated to optimally manage the disease, in collaboration with healthcare professionals. Identification and avoidance of triggers is also of significant importance. Assessment and monitoring should be performed regularly to re-evaluate and fine-tune treatment. Pharmacotherapy is the cornerstone of treatment. The optimal use of medication can, in most cases, help patients control symptoms and reduce the risk for future morbidity. The management of exacerbations is a major consideration, independent of chronic treatment. There is a trend toward considering phenotype-specific treatment choices; however, this goal has not yet been achieved.
Article
Component-resolved diagnosis of allergies allows disease-specific patterns of sensitization in some conditions such as allergic bronchopulmonary aspergillosis ABPA). By determination of IgE against important pollen allergens such as Bet v 1, Ole e 1 or Phl p1/Phl p 5, more precise guidance for allergen-specific immunotherapy may be achieved, as pollen extracts contain mostly these major allergens. Sensitizations against minor allergens such as profilins or polcalcins influence the outcome of IgE measurements against full allergen sources, but are often of limited clinical relevance. In food allergy, frequent cross reactivity between pollens such as birch pollen via Bet v 1/PR10 proteins can be identified. Sensitization against some storage proteins such as peanut (Ara h 2) or lipid transfer proteins of peach (Pru p 3) or hazelnut (Cor a 8) may indicate an increased risk of severe anaphylactic reactions. Exercise-induced anaphylaxis, unclear sensitizations against latex or double-positivity in insect allergy are other useful indications for component-resolved diagnosis. Microarray-based allergen chip diagnosis makes possible today the detection of IgE against more than 100 allergens in tiny amounts of serum and is very promising, but still needs evaluation and optimization in regard to allergen selection and sensitivity.
Article
The identification of disease-eliciting allergens is a prerequisite for accurate prescription of allergen-specific immunotherapy (SIT). The aim of this study was to determine whether molecular diagnosis (MD) may change indication and allergen prescription of SIT. A total of 141 patients with allergic rhinoconjunctivitis and/or asthma sensitized to pollen with or without concomitant food allergy were included. Skin prick testing with a panel of aeroallergens and a microarray-based panel of allergens (ISAC(®); Phadia, Sweden) was performed in all patients. Prior to learning the results of molecular diagnosis, three of the authors reached a consensus on the indication of SIT and use of allergens following EAACI recommendations, basing their judgment on clinical history and skin prick test results before and after obtaining the ISAC results. The agreement coefficient (kappa index) was used to analyze the results. Fifty-nine percent of the patients were women with a mean age of 31 ± 13.63. Agreement in SIT indication before and after ISAC(®) results was found in only 62 (46%) patients (kappa = 0.1057 ± 0.0413). Concerning allergens used in the most common prescriptions before and after MD results, we obtained the following results: κ = 0.117 ± 0.0825 for grass; κ = 0.1624 ± 0.0639 for olive; κ = 0.0505 ± 0.0548 for olive and grass; κ = 0.1711 ± 0.0471 for grass and cypress; κ = 0.1897 ± 0.0493 for grass and London plane; κ = 1 ± 0.0842 for olive and cypress, and κ = 0.3586 ± 0.0798 for other combinations. There was very low agreement concerning indication and use of allergens for SIT before and after performing MD. This discrepancy emphasizes the usefulness of MD, at least in areas of complex sensitization to pollen, in determining correct indication of SIT.