covid
Buscar en
Allergologia et Immunopathologia
Toda la web
Inicio Allergologia et Immunopathologia Microarrays: Molecular allergology and nanotechnology for personalised medicine ...
Journal Information
Vol. 38. Issue 4.
Pages 217-223 (July - August 2010)
Share
Share
Download PDF
More article options
Vol. 38. Issue 4.
Pages 217-223 (July - August 2010)
Review
Full text access
Microarrays: Molecular allergology and nanotechnology for personalised medicine (II)
Visits
3972
J.M. Lucas
Section of Pediatric Allergy, Virgen de la Arrixaca University Hospital, Murcia, Spain
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Abstract

Progress in nanotechnology and DNA recombination techniques have produced tools for the diagnosis and investigation of allergy at molecular level. The most advanced examples of such progress are the microarray techniques, which have been expanded not only in research in the field of proteomics but also in application to the clinical setting.

Microarrays of allergic components offer results relating to hundreds of allergenic components in a single test, and using a small amount of serum which can be obtained from capillary blood. The availability of new molecules will allow the development of panels including new allergenic components and sources, which will require evaluation for clinical use.

Their application opens the door to component-based diagnosis, to the holistic perception of sensitisation as represented by molecular allergy, and to patient-centred medical practice by allowing great diagnostic accuracy and the definition of individualised immunotherapy for each patient.

The present article reviews the application of allergenic component microarrays to allergology for diagnosis, management in the form of specific immunotherapy, and epidemiological studies. A review is also made of the use of protein and gene microarray techniques in basic research and in allergological diseases. Lastly, an evaluation is made of the challenges we face in introducing such techniques to clinical practice, and of the future perspectives of this new technology.

Keywords:
Arrays
Microarrays
Diagnosis
Allergy
Full Text
The clinical use of allergenic component microarrays

Microarrays of allergic components offer results relating to hundreds of allergenic components in a single test, and using a small amount of serum which can be obtained from capillary blood. By increasing the level of analytical detail, allergenic component-resolved diagnosis (CRD) microarrays afford an image of patient sensitisation at molecular level – making it possible to distinguish between major and minor (but equally important in terms of disease) allergens. In this context, by also identifying the panallergens, the true sensitisation profile (mono-, oligo- or polysensitisation) can be established.

This broad, holistic view will make it possible to monitor the IgE reactivity profiles during the development and course of the disease and its treatment, evaluating the correlation between the changes in these profiles and the patient clinical parameters (e.g., progression of allergic rhinoconjunctivitis to asthma). Thanks to the use of molecular allergenic markers, such applications may help predict the risk of developing new allergic disorders or their intensity (e.g., increased risk of anaphylaxis due to rosacea associated to LTP,1 of asthma associated to Ole e 2 and Ole e 10,2 or a lesser risk in people allergic to latex if they are only allergic to Hev b 8),3 and establish the origin of sensitisation (e.g., latex, according to the pattern of its allergenic components).4,5

This knowledge of the molecular sensitisation profile can already be used to individualise the composition of extracts for specific immunotherapy with great accuracy, by discovering cross-sensitisations to panallergens which lack clinical relevance. Likewise, allergenic microarrays could be used in the context of a “pharmacoproteomic” approach to monitor the efficacy of specific immunotherapy, assessing the evolution of allergen-specific IgE/IgG or the appearance of new sensitisations during treatment which could have an impact upon the clinical course.

DiagnosisFood allergy

Many studies indicate that the molecular analysis of allergenic sensitisation patterns can be of increased clinical usefulness in diagnosing IgE-mediated allergic processes. Purified natural as well as recombinant allergens have been used to this effect, together with synthetic peptide panels.6

Different studies have shown microarrays to be useful in allergic patients for identifying cow’s milk and egg allergen patterns7,8. In this sense it is essential for the technique to be applicable to the management of patients with food allergy, with the prediction of when a given potentially allergic patient will develop a reaction after ingestion of the culprit food allergen.

These clinical decision points (CDPs) offering certain positive predictive values (PPVs) in application to oral provocation are regarded as important parameters for diagnosing paediatric food allergy,9–11 and the combined microarray findings for egg and cow’s milk in different age groups have been able to yield PPVs for 90%, 95% and 99% of the patients.8

Beyer et al. determined disease persistence by screening children with allergy to cow’s milk using a microarray casein peptide panel.12

The technique has also been shown to be useful in application to peanut allergy,13 where a correlation is observed between the diversity of specific IgE and the severity of the allergic clinical manifestations, as well as the different epitopes of these allergenic molecules.14 The microarray technique has been useful for studying the allergenic potential of two 2S albumins from soybean.15

Incorporation is required of new allergens such as Ara h 9, which in the peanut allergy population constitutes a minor allergen, but is more important in patients from south-eastern Europe, where sensitisation to the classical major allergens – Ara h 1, Ara h 2, and Ara h 3 – is less pronounced.16

The microarray technique as a screening test has served to classify the phenotype of pollen-food allergy syndrome in patients with oral allergy to soy.17

Allergy to aeroallergens

Deinhofer et al.18 demonstrated that for a well characterised and selected panel of recombinant pollen allergens, the microchip-based diagnosis of allergy can equal the performance of the well-established tests in terms of sensitivity and specificity – with excellent correlations for recombinant birch and grass allergens. Wöhrl et al. 19 in turn confirmed the diagnostic usefulness of a microarray involving a broader range of inhaled allergen components.

The pollens of Ambrosia and Artemisia share allergens – profilin, polcalcin and nonspecific lipid transfer protein (nsLTP) – which cause cross-reactivity between them.20 In a study of 509 patients, 93% of the individuals sensitised to Artemisia were also found to be sensitised to Ambrosia, while 48% of those sensitised to Ambrosia were also sensitised to Artemisia.21 In patients of this kind, the study of the allergenic components of both types of pollen based on microarray tests has been shown to be as useful as ELISA testing – yielding information on the differential diagnosis between one type of sensitisation and the other, of great importance for selecting the most appropriate allergenic source for immunotherapy.22

This technique has been used to study the sensitisation profiles pollen-vegetable food syndromes such as birch-apple syndrome, 23 or to establish the differential diagnosis of a type of occupational asthma (baker’s asthma).24

Microarrays have also been found to be useful in differentiating primary sensitisations to pollen, fruits and latex from the subclinical immune recognition of different prophyllins and LTP in paediatric patients.25

Allergy to insect venom

Protein microarrays can also be used for the study of allergy to bee, wasp and ant venoms.26

Immunotherapy

In allergen-specific immunotherapy, identification of the allergens causing the disease is a fundamental requirement for precise treatment prescription. This is particularly important when considering that over 60% of all pollen allergic patients are polysensitised. In this setting, distinguishing panallergens (profilins, polcalcins) and homologous molecules of limited presence (e.g., LTP) from highly selective components results in improved immunotherapy.27

A number of studies have shown that recombinant allergens can be used to distinguish patients who can benefit from immunotherapy (monosensitised patients) from those showing cross-reactions with an allergen from a different biological source.28,29 Furthermore, purified allergenic markers can be used to monitor the immune effects of treatment and prevent new sensitisations during immunotherapy.30,31

In all these aspects, allergenic component microarrays are of great importance. In addition, the fact that the molecular sensitisation profile of the patient can be known makes it logical to define the next step as the introduction of “molecular” immunotherapy adapted to the specific sensitisation profile of each individual patient.32–34

In this sense, clinical trials have been made using recombinant allergens as either the sum of recombinants that conform a natural extract,35 or as recombinant allergens modified to exhibit reduced allergenic activity 36–38– showing also that immunotherapy targeted to recombinant allergens can be both effective and safe. Recently, a new specific immunotherapeutic treatment targeted to Alternaria with purified native allergen (nAlt a 1) has been introduced on the market, showing good immunogenicity and safety.39

In this context it is necessary to demonstrate that such formulations possess the same potency as the complete natural allergens, and that the panel of recombinant allergens encompasses all the epitopes recognised by the B and T cells. In the case of gramineous species (grasses), it has been shown that a panel of 5 recombinants (Phl p 1, Phl p 2, Phl p 5a, Phl p 5b), or the mentioned panel plus Phl p 6, significantly reduces the symptoms and induces a specific and very intense response against these allergens.35 Birch allergens behave in a similar way.

It has been shown that recombinant hybrids from unrelated allergenic sources possess great immunogenic capacity.40

In patients sensitised to dust mites, the technique already allows us to determine whether they are sensitised to group 1 or to group 2 allergens, or whether in contrast the subjects are sensitised to tropomyosin – which would constitute a contraindication to immunotherapy with dust mite extracts containing group 1 and 2 allergens.26

Epidemiological studies

“Universal” allergenic microarrays may offer a means for determining specific IgE reactivity in concrete regions or populations, specific disease patterns, or the dynamic evolution of sensitisations – contributing much more detailed information.41

Given the increase in the prevalence of allergic disease in children and the possibility of sensitisation during the foetal period, studies have been made of the relationships between genes, and between these and the environment, based on the use of arrays. The findings suggest that the control of maternal atopy and the prenatal redox response may prevent IgE-mediated diseases.42

It has been shown that early sensitisation to a common allergen (e.g., egg, grass pollen, dust mites or cat dander) is associated with an increased risk of developing asthma.43 The Section on Pediatrics of the European Academy of Allergy and Clinical Immunology (EAACI) has established the criteria for allergy detection studies in children, with a view to defining a strategy which can help prevent the course of allergy.44

Since part of the primary preventive measures have been shown to be scantly effective,45 new approaches and lines of research are needed46 to understand which factors influence the development of these diseases. In this context, allergen microarrays, thanks to the extremely low volume of sample required (20μl) and the large number of allergens which can be studied, offer a tool for improved risk evaluation in atopic individuals in the early stages of life.

Use of gene and protein microarrays in allergology

Microarray technology has become the most potent instrument for analysing genic expression profiles and functional genomics. In this context, a range of basic and applied research lines have been developed in the field of allergic diseases, in which microarray testing is a critical tool.

Basic research

Microarray technology has been used for the study of interactions among allergenic proteins, immunoglobulins and T cell receptors, with a view to developing genetic modifications which can yield hypoallergenic variants of plant proteins.47

Microarrays have been applied in the investigation of clonal diversity48 and immune response heterogeneity among patients,49 and to establish the clinical correlations between antibody diversity and the allergic manifestations.50–52

Microarray technology has been shown to be very useful for mapping and characterising allergenic epitopes.53 An example is represented by cow’s milk allergens,54 with differentiation between IgE and IgG4 patterns for sequential epitopes of alpha(s1)-, alpha(s2)-, beta- and kappa-caseins and beta-lactoglobulin in reactive patients and tolerant individuals.55 However, further studies are needed to assess the potential usefulness of the technique in clinical practice.

In immunology, microarrays offer applications in the extensive field of genic expression in primary immune deficiencies.56

Atopic dermatitis

Noh et al.57 in studying the relationship between atopic dermatitis and food allergy, measured the concentration of IgE and IgG 4 specific of certain foods, using an allergenic protein chip (Allergy Chip). The authors suggested that this protein microarray technique could be one of the most useful methods for evaluating allergic condition in such diseases.

Its usefulness in diagnosing sensitisations in patients with atopic dermatitis has also been recently demonstrated by Ott et al.58

Likewise, antibody microarrays have been used to study the cytokines and chemokines implicated in the physiopathology of atopic dermatitis in children,59 with the use of biochips for the molecular genomic analysis of this disease.60

Contact dermatitis

Large-scale genetic expression analysis based on the use of DNA microarrays has been used to study the genes implicated in contact dermatitis. The detected genetic markers can be posteriorly included in microarrays on a smaller scale, to establish the definitive diagnostic test.61

Rhinitis

Microarrays have helped to determine genic expression in allergic rhinitis,62 to study the pathogenesis of the disease, and to evaluate the role of chemokines and their receptors via the expression of their complementary DNA (cDNA) in the nasal mucosa, using gene biochips.63

Asthma

In asthma, microarray technology has been used to study the pathogenesis of the disease, yielding new biological diagnostic markers and therapeutic targets, and furthering our understanding of the pathogenic mechanisms of bronchial asthma.64,65 In application to allergic asthma complex, microarrays have been shown to be valid and relevant in defining allergic asthma expression patterns, comparing two different types of biochips (U95Av2 and U133A Gene Chips) in the study of bronchial biopsy samples.66

Microarrays have been used to explore the importance of interleukin 13 (IL-13) in the pathogenesis of asthma.67 Another study, involving cytokine microarrays, has shown cytokine PARC/CCL18 to be a new factor implicated in the development of eosinophilic inflammation of the airways in asthma.68

Asthma exacerbations requiring hospital admission in children have been associated to viral infections occurring mainly in atopic individuals, although the significance of such comorbidities is not known. A study using flow cytometry and genic expression microarrays has suggested that viral infection in atopic children may trigger an atopic inflammatory cascade which amplifies and maintains the airway inflammation initiated by innate antiviral immunity via use of the underlying atopic mechanisms. These interactions may explain the susceptibility of atopic patients to severe asthma episodes induced by viral infections.69

Likewise, microarrays offer great potential for studying the pharmacogenomics of asthma in the genetic polymorphisms which influence the results of asthma therapy.70 Recent studies have revealed the genes associated to asthma and to a favourable response to inhaled corticosteroids in bronchial epithelial cells71 and in peripheral blood mononuclear cells.72 Likewise, if pharmacogenetic studies were able to establish the genetic differences in patients with a good response to fluticasone or montelukast,73,74 it could become possible to avoid the excessive or insufficient treatment of such individuals – microarray technology in turn facilitating the individualisation of therapy.

T cell microarrays are used to assess the activity of the genes mediating apoptosis in the context of tolerance – this being useful for evaluating the efficacy of immunotherapy, particularly as regards the sublingual route in peanut allergy.75

Challenges for clinical application

In order for microarray testing to become a routine technique in allergy, its performance and diagnostic accuracy must be confirmed by large-scale, population-based multicentre studies reflecting the prevalence of the disease and the setting in which the mentioned technology will be posteriorly used.

In a time of increasing control of healthcare expenditure, new technologies must be evaluated to confirm that they offer the expected added value and cost/benefit ratio. This technology uses allergens which must be purified or created with recombinant technology, and moreover requires sophisticated chip analytical instruments. However, with use, the cost of this technology will tend to decrease. On the other hand, the cost must be weighed against the great amount of information obtained, the small sample volume required, and particularly the probable benefit of determining the prognosis and contributing to prevention and the effectiveness and safety of treatment, by allowing selection of the most appropriate allergen composition for immunotherapy. Cost/utility studies are therefore needed, with the comparison of effectiveness versus the rest of the in vitro and in vivo diagnostic techniques currently available.76

In turn, it must be checked that transfer of the technology from the research setting to clinical practice (and from evidence to clinical application) proves satisfactory 77. For example: despite their amplitude, the existing allergen panels do not contain all the components of clinical interest of the respective allergen sources, or all the sources that are of clinical relevance. Additional clinical studies are needed to improve allergen panel definition.

Likewise, clinical decision support is needed, since the large volume of information obtained is complex to interpret. In this sense, the microarray test results should offer help for clinicians in the form of specific software, with direct access to expert databases and guides, making it possible to better interpret the tests and their application to therapy.

Conclusions and future perspectives

Allergenic component microarrays offer an elegant way to avoid the problem of allergen standardisation and false polysensitisation.

However, this novel technology has further implications: it represents a true change in paradigm, offering a different view of the aetiology of allergic disease and extending the approach from allergenic sources to allergenic molecules – from botanical classification to molecular classification.78 This systematisation in turn should prove manageable, since it involves a limited number of protein families and functions,79,80 thus allowing a holistic approach to the disease.

This technology opens the door to personalised medical practice, by allowing diagnosis and planning at molecular level, specific for each patient, with a known and balanced dosing of standardised allergens for immunotherapy. In this context, it is possible to include allergens that are not well represented in the natural extracts but which are nevertheless of clinical relevance. Alternatively, genetic engineering can be used to incorporate hypoallergenic variants with lesser IgE reactivity, but which result in equal or even superior T cell reactivity – thus separating immunogenicity from allergenicity, and making the technology both more effective and safer.

This novel diagnostic technology is minimally invasive, makes use of small sample volumes, offers quantitative results, and constitutes a multianalytic test – thus facilitating its incorporation to clinical use. However, not all patients will require such testing, since in clinical practice studies at molecular level are mainly recommended in individuals sensitised to panallergens detected by prick testing,81 or in polysensitised patients in which diagnostic doubts persist.

In an even broader sense, the use of microarrays for exploring these complex disease processes will reveal the aetiopathogenic implication of multiple genes, helping to clarify the many and varied mechanisms and associated pathological pathways – the complexity of which requires the application of network theory82 to allow understanding. This has given rise to a new field in biology – the biology of systems – which aims to decipher the cell and signal networks underlying these biological processes, using gene and protein microarrays as its principal tool.

Application of the new knowledge afforded by nanotechnology on an individualised basis in allergic patients requires specialised knowledge that does not form part of the standard curriculum of clinical allergologists at the present time. In this context, a training effort in both the clinical and research settings is warranted, with a view to developing this new era in allergology.

Conflict of interest

The author declares no conflicts of interest.

References
[1]
R. Asero, G. Mistrello, D. Roncarolo, S.C. de Vries, M.F. Gautier, C.L. Cuirana, et al.
Lipid transfer protein: a pan-allergen in plant-derived foods that is highly resistant to pepsin digestion.
Int Arch Allergy Immunol, 124 (2001), pp. 67-69
[2]
J. Quiralte, E. Llanes, P. Barral, J.M. Arias de Saavedra, B. Sáenz de San Pedro, M. Villalba, et al.
Ole e 2 and Ole e 10: new clinical aspects and genetic restrictions in olive pollen allergy.
[3]
O. Quercia, G.F. Stefanini, A. Scardovi, R. Asero.
Patients monosensitised to Hev b 8 (Hevea brasiliensis latex profilin) may safely undergo major surgery in a normal (non-latex safe) environment.
Eur Ann Allergy Clin Immunol, 41 (2009), pp. 112-116
[4]
H.Y. Yeang, K.F. Cheong, E. Sunderasan, S. Hamzah, N.P. Chew, S. Hamid, et al.
The 14.6 kd rubber elongation factor (Hev b 1) and 24 kd (Hev b 3) rubber particle proteins are recognized by IgE from patients with spina bifida and latex allergy.
J Allergy Clin Immunol, 98 (1996), pp. 628-639
[5]
A. Valls, C.Y. Pascual, M.T. Caballero, M. Martin Esteban.
Latex allergy.
Allergol Immunopathol, 32 (2004), pp. 295-305
[6]
J. Lidholm, B.K. Ballmer-Weber, A. Mari, S. Vieths.
Component resolved diagnostics in food allergy.
Curr Opin Allergy Clin Immunol, 6 (2006), pp. 234-240
[7]
J.C. Gaudin, H. Rabesona, Y. Choiset, G. Yeretssianw, J.M. Chobert, V. Sakanyanwz, et al.
Assessment of the immunoglobulin E-mediated immune response to milk-specific proteins in allergic patients using microarrays Clinical and Experimental.
Allergy, 38 (2008), pp. 686-693
[8]
H. Ott, J.M. Baron, R. Heise, C. Ocklenburg, S. Stanzel, H.F. Merk, et al.
Clinical usefulness of microarray-based IgE detection in children with suspected food allergy.
[9]
S. Celik-Bilgili, A. Mehl, A. Verstege, U. Staden, M. Nocon, K. Beyer, et al.
The predictive value of specific immunoglobulin E levels in serum for the outcome of oral food challenges.
Clin Exp Allergy, 35 (2005), pp. 268-273
[10]
H.A. Sampson.
Utility of food-specific IgE concentrations in predicting symptomatic food allergy.
J Allergy Clin Immunol, 107 (2001), pp. 891-896
[11]
M.C. Garcia-Ara, M.T. Boyano-Martinez, J.M. Diaz-Pena, M.F. Martin-Munoz, M. Martin-Esteban.
Cow´s milk-specific immunoglobulin E levels as predictors of clinical reactivity in the follow-up of the cow´s milk allergy infants.
Clin Exp Allergy, 34 (2004), pp. 866-870
[12]
K. Beyer, K.M. Jarvinen, L. Bardina, M. Mishoe, K. Turjanmaa, B. Niggemann, et al.
IgE-binding peptides coupled to a commercial matrix as a diagnostic instrument for persistent cow´s milk allergy.
J Allergy Clin Immunol, 116 (2005), pp. 704-705
[13]
W.G. Shreffler, K. Beyer, T.H. Chu, A.W. Burks, H.A. Sampson.
Microarray immunoassay: association of clinical history, in vitro IgE function, and heterogeneity of allergenic peanut epitopes.
J Allergy Clin Immunol, 113 (2004), pp. 776-782
[14]
A.E. Flinterman, E.F. Knol, D.A. Lencer, L. Bardina, C.F. den Hartog Jager, J. Lin, et al.
Peanut epitopes for IgE and IgG4 in Peanut sensitized children in relation to severity of peanut allergy.
J Allergy Clin Immunol, 121 (2008), pp. 737-743
[15]
J. Lin, P.R. Shewry, D.B. Archer, K. Beyer, B. Niggemann, H. Haas, et al.
The potential allergenicity of two 2S albumins from soybean (Glycine max): a protein microarray approach.
Int Arch Allergy Immunol, 141 (2006), pp. 91-102
[16]
S. Krause, G. Reese, S. Randow, D. Zennaro, D. Quaratino, P. Palazzo, et al.
Lipid transfer protein (Ara h 9) as a new peanut allergen relevant for a Mediterranean allergic population.
J Allergy Clin Immunol, 124 (2009), pp. 771-778
[17]
A. Yagami, Y. Inaba, Y. Kuno, K. Suzuki, A. Tanaka, S. Sjolander, et al.
Two cases of pollen-food allergy syndrome to soy milk diagnosed by skin prick test, specific serum immunoglobulin E and microarray analysis.
[18]
K. Deinhofer, H. Sevcik, N. Balic, C. Harwanegg, R. Hiller, H. Rumpold, et al.
Microarrayed allergens for IgE profiling.
Methods, 32 (2004), pp. 249-254
[19]
S. Wohrl, K. Vigl, S. Zehetmayer, R. Hiller, R. Jarisch, M. Prinz, et al.
The performance of a component-based allergen-microarray in clinical practice.
[20]
N. Wopfner, G. Gadermaier, M. Egger, R. Asero, C. Ebner, B. Jahn-Schmid, et al.
The Spectrum of Allergens in Ragweed and Mugwort Pollen.
Int Arch Allergy Immunol, 138 (2005), pp. 337-346
[21]
R. Asero, N. Wopfner, P. Gruber, G. Gadermaier, F. Ferreira.
Artemisia and Ambrosia hypersensitivity: co-sensitization or co-recognition?.
Clin Exp Allergy., 36 (2006), pp. 658-665
[22]
G. Gadermaier, N. Wopfner, M. Wallner, M. Egger, A. Didierlaurent, G. Regl, et al.
Array-based profiling of ragweed and mugwort pollen allergens.
[23]
D.G. Ebo, C.H. Bridts, M.M. Verweij, K.J. De Knop, M.M. Hagendorens, L.S. De Clerck, et al.
Sensitization profiles in birch pollen-allergic patients with and without oral allergy syndrome to apple: lessons from multiplexed component-resolved allergy diagnosis.
Clin Exp Allergy, 40 (2010), pp. 339-347
[24]
C. Constantin, S. Quirce, M. Poorafshar, A. Touraev, B. Niggemann, A. Mari, et al.
Micro-arrayed wheat seed and grass pollen allergens for component-resolved diagnosis.
[25]
M.J. Goikoetxea, P. Cabrera-Freitag, M.L. Sanz, M. Fernandez-Benitez.
The importance of in vitro component-resolved diagnosis in paediatric patients.
Allergol Immunopathol(Madr), 38 (2010), pp. 37-40
[26]
D.C. de Graaf, M. Aerts, E. Danneels, B. Devreese.
Bee, wasp and ant venomics pave the way for a component-resolved diagnosis of sting allergy.
J Proteomics, 72 (2009), pp. 145-154
[27]
C. Moreno-Aguilar.
Improving pollen immunotherapy: minor allergens and panallergens.
Allergol Immunopathol, 36 (2008), pp. 26-30
[28]
L. Kazemi–Shirazi, V. Niederberger, B. Linhart, J. Lidholm, D. Kraft, R. Valenta.
Recombinant marker allergens: diagnostic gatekeepers for the treatment of allergy.
Int Arch Allergy Immunol, 127 (2002), pp. 259-268
[29]
R. Movérare, K. Westritschnig, M. Svensson, B. Hayek, M. Bende, G. Pauli, et al.
Different IgE reactivity profiles in birch pollen-sensitive patients from six European populations revealed by recombinant allergens: an imprint of local sensitization.
Int Arch Allergy Immunol, 128 (2002), pp. 325-335
[30]
R. Movérare, L. Elfman, E. Vesterinen, T. Metso, T. Haahtela.
Development of new IgE specificities to allergenic components in birch pollen extract during specific immunotherapy studied with immunoblotting and Pharmacia CAP System.
Allergy, 57 (2002), pp. 423-430
[31]
R. Valenta, T. Twaroch, I. Swoboda.
Component-resolved diagnosis to optimize allergen-specific immunotherapy in the mediterranean area.
J Investig Allergol Clin Immunol, 17 (2007), pp. 36-40
[32]
T. Zeiler.
Recombinant allergen fragments as candidate preparations for allergen immunotherapy.
J Allergy Clin Immunol, 100 (1997), pp. 721
[33]
R. Valenta, D. Kraft.
From allergen structure to new forms of allergen specific immunotherapy.
Curr Opin Immunol, 14 (2002), pp. 718-727
[34]
R. Rodriguez, M. Villalba, E. Batanero, O. Palomares, J. Quiralte, G. Salamanca, et al.
Olive pollen recombinant allergens: Value in diagnosis and immunotherapy.
J Investig Allergol Clin Immunol, 17 (2007), pp. 4-10
[35]
M. Jutel, L. Jaeger, R. Suck, H. Meyer, H. Fiebig, O. Cromwell.
Allergen-specific immunotherapy with recombinant grass pollen allergens.
J Allergy Clin Immunol, 116 (2005), pp. 608-613
[36]
V. Niederberger, F. Horak, S. Vrtala, S. Spitzauer, M.T. Krauth, P. Valent, et al.
Vaccination with genetically engineered allergens prevents progression of allergic disease.
Proc Natl Acad Sci USA, 101 (2004), pp. 14677-14682
[37]
E. Pace.
Hypoallergenic fragment of par j 2 increases functional expression of toll-like receptors in atopic children.
[38]
R. González-Rioja, I. Ibarrola, M.C. Arilla, A. Ferrer, A. Mir, C. Andreu, et al.
Genetically engineered hybrid proteins from Parietaria judaica pollen for allergen-specific immunotherapy.
J Allergy Clin Immunol, 120 (2007), pp. 602-609
[39]
L. Prieto, D. Aldana, R. Palacios, C. Pérez, A. Ferrer.
Inmunoterapia con nAlt a 1 mediante pauta cluster: seguridad e inmunogenicidad.
Journal Investig Allergol Clin Immunology, 19 (2009), pp. 168
[40]
B. Linhart.
A hybrid molecule resembling the epitope spectrum of grass pollen for allergy vaccination.
Journal of Allergy and Clinical Immunology, 115 (2005), pp. 1010
[41]
E. Scala, D. Quaratino, A. Zaffiro, M.L. Bernardi, R. Ferrara, D. Zennaro, et al.
IgE proteomic in the diagnosis and epidemiology of allergic diseases: a population survey.
Allergy, 62 (2007), pp. S432
[42]
K.D. Yang, J.C. Chang, H. Chuang, H.M. Liang, H.C. Kuo, Y.S. Lee, et al.
Gene-gene and gene-environment interactions on IgE production in prenatal stage.
Allergy, (2009),
[43]
H.L. Rhodes, P. Thomas, R. Sporik, S.T. Holgate, J.J. Cogswell.
A birth cohort study of subjects at risk of atopy: twenty two-year follow-up of wheeze and atopic status.
Am J Respir Crit Care Med, 165 (2002), pp. 176-180
[44]
A. Høst, S. Andrae, S. Charkin, C. Díaz-Vázquez, S. Dreborg, P.A. Eigenmann, et al.
Allergy testing in children: why, who, when and how?.
Allergy, 58 (2003), pp. 559-569
[45]
I. Pali-Schöll, H. Renz, E. Jensen-Jarolim.
Update on allergies in pregnancy, lactation, and early childhood.
The Journal of allergy and clinical immunology, 123 (2009), pp. 1012-1021
[46]
E. Hamelmann, U. Herz, P. Holt, A. Høst, R.P. Lauener, P.M. Matricardi, et al.
New visions for basic research and primary prevention of pediatric allergy: an iPAC summary and future trends.
Pediatr Allergy Immunol, 19 (2008), pp. 4-16
[47]
M.B. Singh, N. de Weerd, P. Bhalla.
Genetically engineered plant allergens with reduced anaphylactic activity.
Int Arch Allergy Immunol, 119 (1999), pp. 75-85
[48]
C. Pinilla, R. Martin, B. Gran, J.R. Appel, C. Boggiano, D.B. Wilson, et al.
Exploring immunological specificity using synthetic peptide combinatorial libraries.
Curr Opin Immunol, 11 (1999), pp. 193-202
[49]
W. Shreffler, D. Lencer, L. Bardina, H. Sampson.
Epitope mapping by microarray peptide immunoassay and cluster analysis reveals interpatient heterogeneity and a more detailed map.
J Allergy Clin Immunol, 116 (2005), pp. 893-899
[50]
S. Mohapatra.
Determinant spreading: implications in allergic disorders.
Immunol Today, 15 (1994), pp. 596-597
[51]
K. Beyer, L. Ellman-Grunther, K.M. Jarvinen, R.A. Wood, J. Hourihane, H.A. Sampson.
Measurement of peptide-specific IgE as an additional tool in identifying patients with clinical reactivity to peanuts.
J Allergy Clin Immunol, 112 (2003), pp. 202-207
[52]
P. Chatchatee, K.M. Jarvinen, L. Bardina, K. Beyer, H.A. Sampson.
Identification of IgE- and IgG-binding epitopes on alpha(s1)-casein: differences in patients with persistent and transient cow’s milk allergy.
J Allergy Clin Immunol, 107 (2001), pp. 379-383
[53]
J. Lin, L. Bardina, W.G. Shreffler.
Microarrayed allergen molecules for diagnostics of allergy.
Methods Mol Biol, 524 (2009), pp. 259-272
[54]
J. Lin, L. Bardina, W.G. Shreffler, D.A. Andreae, Y. Ge, J. Wang, et al.
Development of a novel peptide microarray for large-scale epitope mapping of food allergens.
J Allergy Clin Immunol, 124 (2009), pp. 315-322
[55]
I. Cerecedo, J. Zamora, W.G. Shreffler, J. Lin, L. Bardina, M.C. Dieguez, et al.
Mapping of the IgE and IgG4 sequential epitopes of milk allergens with a peptide microarray-based immunoassay.
J Allergy Clin Immunol, 122 (2008), pp. 589-594
[56]
K.E. Blomberg, C.I. Smith, J.M. Lindvall.
Gene expression signatures in primary immunodeficiencies: the experience from human disease and mouse models.
Curr Mol Med, 7 (2007), pp. 555-566
[57]
G. Noh, H.S. Ahn, N.Y. Cho, S. Lee, J.W. Oh.
The clinical significance of food specific IgE/IgG4 in food specific atopic dermatitis.
Pediatr Allergy Immunol, 18 (2007), pp. 63-70
[58]
H. Ott, R. Fölster-Holst, H.F. Merk, J.M. Baron.
Allergen microarrays: a novel tool for high-resolution IgE profiling in adults with atopic dermatitis.
Eur J Dermatol, 20 (2010), pp. 54-61
[59]
K.L. Hon, G.K. Ching, K.Y. Wong, T.F. Leung, A.K. Leung.
A pilot study to explore the usefulness of antibody array in childhood atopic dermatitis.
J Natl Med Assoc, 100 (2008), pp. 500-504
[60]
H. Saito.
Much atopy about the skin: genome-wide molecular analysis of atopic eczema.
Int Arch Allergy Immunol, 137 (2005), pp. 319-325
[61]
S. Szameit, E. Weber, C. Noehammer.
DNA microarrays provide new options for allergen testing.
Expert Rev Mol Diagn, 9 (2009), pp. 843-850
[62]
M. Benson, L. Carlsson, G. Guillot, M. Jernås, M.A. Langston, M. Rudemo, et al.
A network-based analysis of allergen-challenged CD4+ T cells from patients with allergic rhinitis.
Genes Immun, 7 (2006), pp. 514-521
[63]
R.X. Zhang, S.Q. Yu, J.Z. Jiang, G.J. Liu.
Complementary DNA microarray analysis of chemokines and their receptors in allergic rhinitis.
J Investig Allergol Clin Immunol, 17 (2007), pp. 329-336
[64]
K. Izuhara, H. Saito.
Microarray-based identification of novel biomarkers in asthma.
Allergol Int, 55 (2006), pp. 361-367
[65]
M.S. Rolph, M. Sisavanh, S.M. Liu, C.R. Mackay.
Clues to asthma pathogenesis from microarray expression studies.
Pharmacol Ther, 109 (2006), pp. 284-294
[66]
A. Chamberland, A.M. Madore, K. Tremblay, M. Laviolette, C. Laprise.
A comparison of two sets of microarray experiments to define allergic asthma expression pattern.
Exp Lung Res, 35 (2009), pp. 399-410
[67]
D.A. Kuperman, C.C. Lewis, P.G. Woodruff, M.W. Rodríguez, Y.H. Yang, G.M. Dolganov, et al.
Dissecting asthma using focused transgenic modeling and functional genomics.
J Allergy Clin Immunol, 116 (2005), pp. 305-311
[68]
H.B. Kim, C.K. Kim, K. Iijima, T. Kobayashi, H. Kita.
Protein microarray analysis in patients with asthma: elevation of the chemokine PARC/CCL18 in sputum.
Chest, 135 (2009), pp. 295-302
[69]
L.S. Subrata, J. Bizzintino, E. Mamessier, A. Bosco, K.L. McKenna, M.E. Wikström, et al.
Interactions between innate antiviral and atopic immunoinflammatory pathways precipitate and sustain asthma exacerbations in children.
J Immunol, 183 (2009), pp. 2793-2800
[70]
A. Pahl, E. Benediktus, L. Chialda.
Pharmacogenomics of asthma.
Curr Pharm Des, 12 (2006), pp. 3195-3206
[71]
P.G. Woodruff, H.A. Boushey, G.M. Dolganov, C.S. Barker, Y.H. Yang, S. Donnelly, et al.
Genome-wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids.
Proc Natl Acad Sci U S A, 104 (2007), pp. 15858-15863
[72]
H. Hakonarson, U.S. Bjornsdottir, E. Halapi, J. Bradfield, F. Zink, M. Mouy, et al.
Profiling of genes expressed in peripheral blood mononuclear cells predicts glucocorticoid sensitivity in asthma patients.
Proc Natl Acad Sci USA, 102 (2005), pp. 14789-14794
[73]
R.S. Zeiger, S.J. Szefler, B.R. Phillips, M. Schatz, F.D. Martínez, V.M. Chinchilli, et al.
Response profiles to fluticasone and montelukast in mild-to-moderate persistent childhood asthma.
J Allergy Clin Immunol, 117 (2006), pp. 45-52
[74]
S.J. Szefler, B.R. Phillips, F.D. Martínez, V.M. Chinchilli, R.F. Lemanske, R.C Strunk, et al.
Characterization of within -subject responses to fluticasone and montelukast in childhood asthma.
J Allergy Clin Immunol, 115 (2005), pp. 233-242
[75]
S.M. Jones, L. Pons, J.L. Roberts, A.M. Scurlock, T.T. Perry, M. Kulis, et al.
Clinical efficacy and immune regulation with peanut oral immunotherapy.
J Allergy Clin Immunol, 124 (2009), pp. 292-300
[76]
H.C. Sox, S. Greenfield.
Comparative effectiveness research: a report from the Institute of Medicine.
Ann Intern Med, 151 (2009), pp. 203-205
[77]
A.D. Naik, L.A. Peterson.
The neglected purpose of comparative-effectiveness research.
N Engl J Med, 360 (2009), pp. 1929-1931
[78]
N. Mothes, F. Horak, R. Valenta.
Transition from a botanical to a molecular classification in tree pollen allergy: implications for diagnosis and therapy.
Int Arch Allergy Immunol, 135 (2004), pp. 357-373
[79]
C. Radauer, H. Breitender.
Pollen allergens are restricted to a few protein families and show distinct patterns of species distribution.
J Allergy Clin Immunol, 117 (2006), pp. 141-147
[80]
C. Radauer, M. Bublin, S. Wagner, A. Mari, H. Breiteneder.
Allergens are distributed into few protein families and possess a restricted number of biochemical functions.
J Allergy Clin Immunol, 121 (2008), pp. 847-852
[81]
D. Barber, F. de la Torre, M. Lombardero, I. Antépara, C. Colas, I. Dávila, et al.
Component-resolved diagnosis of pollen allergy based on skin testing with profilin, polcalcin and lipid transfer protein pan-allergens.
Clin Exp Allergy, 39 (2009), pp. 1764-1773
[82]
M. Benson, R. Breitling.
Network theory to understand microarray studies of complex diseases.
Curr Mol Med, 6 (2006), pp. 695-701
Copyright © 2010. SEICAP
Article options
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos

Quizás le interese:
10.1016/j.aller.2020.03.007
No mostrar más