Skip to main content

CSACI guidelines for the ethical, evidence-based and patient-oriented clinical practice of oral immunotherapy in IgE-mediated food allergy

Abstract

Background

Oral immunotherapy (OIT) is an emerging approach to the treatment of patients with IgE-mediated food allergy and is in the process of transitioning to clinical practice.

Objective

To develop patient-oriented clinical practice guidelines on oral immunotherapy based on evidence and ethical imperatives for the provision of safe and efficient food allergy management.

Materials and methods

Recommendations were developed using a reflective patient-centered multicriteria approach including 22 criteria organized in five dimensions (clinical, populational, economic, organizational and sociopolitical). Data was obtained from: (1) a review of scientific and ethic literature; (2) consultations of allergists, other healthcare professionals (pediatricians, family physicians, nurses, registered dieticians, psychologists, peer supporters), patients and caregivers; and patient associations through structured consultative panels, interviews and on-line questionnaire; and (3) organizational and economic data from the milieu of care. All data was synthesized by criteria in a multicriteria deliberative guide that served as a platform for structured discussion and development of recommendations for each dimension, based on evidence, ethical imperatives and other considerations.

Results

The deliberative grid included 162 articles from the literature and media reviews and data from consultations involving 85 individuals. Thirty-eight (38) recommendations were made for the practice of oral immunotherapy for the treatment of IgE mediated food allergy, based on evidence and a diversity of ethical imperatives. All recommendations were aimed at fostering a context conducive to achieving objectives identified by patients and caregivers with food allergy. Notably, specific recommendations were developed to promote a culture of shared responsibility between patients and healthcare system, equity in access, patient empowerment, shared decision making and personalization of OIT protocols to reflect patients’ needs. It also provides recommendations to optimize organization of care to generate capacity to meet demand according to patient choice, e.g. OIT or avoidance. These recommendations were made acknowledging the necessity of ensuring sustainability of the clinical offer in light of various economic considerations.

Conclusions

This innovative CPG methodology was guided by patients’ perspectives, clinical evidence as well as ethical and other rationales. This allowed for the creation of a broad set of recommendations that chart optimal clinical practice and define the conditions required to bring about changes to food allergy care that will be sustainable, equitable and conducive to the well-being of all patients in need.

Background

IgE-mediated food allergy is a condition that imposes food-related restrictions on patients and their caregivers in order to prevent allergic reactions. The burden of disease stems from both the actual and perceived risks of accidental ingestion, including the possibility of a life-threatening allergic reaction. This burden manifests itself in variable levels of anxiety and social limitations and significantly impacts quality of life. The current standard management for food allergy is complete avoidance of the offending allergen in the diet, combined with training on how to recognize and treat allergic reactions. And while avoidance is currently recognized as a safe approach, it has a limited ability to improve a patients’ perception of safety or sense of control over the condition and its associated limitations—this leads many to seek alternative management options [1,2,3,4,5,6].

Oral immunotherapy (OIT) consists of daily ingestion of the offending food allergen (food dosing), starting below a patient’s threshold dose (i.e. the minimum amount that would elicit a reaction), and increasing the dose over time with a goal of increasing clinical tolerance to that food. It had been proposed as a potential alternative to avoidance throughout the 20th and into the early 21st centuries [7], yet its development had been limited until recently. This is mainly due to the risk of allergic reactions associated with OIT, which is difficult to reconcile with current practice standards that focus on the avoidance of reactions at all cost and are arguably rooted in a culture of fear.

OIT can be viewed as a disruptive innovation as it challenges the current paradigm of care in food allergy and highlights its limitations in responding to patient needs. There is a need for patient-centered ethical clinical practice guidelines (CPGs) that include patients and caregivers as well as other stakeholders in the consultation and deliberation process, in order to develop best practice recommendations for providing OIT for food allergy. It is essential to understand patient and caregiver perspectives to ensure proper interpretation of published evidence and understand the ethical issues involved.

Previous CPGs on OIT [8,9,10] have used more traditional approaches, focusing mainly on quantitative clinical evidence. However, in order to ensure a clear vision of all that OIT entails and to benefit all patients with food allergy in need of care, CPGs must not only be based on experimental data (e.g., clinical trials) but also on observational, economic and sociopolitical data, as well as experiential narrative data from both patients and healthcare professionals. The optimal provision of OIT must also account for ethical and organizational data to promote the equitable, sustainable and responsible development of this treatment. In fact, evidence-based medicine has been defined as “the application of the best available research to clinical care, which requires the integration of evidence with clinical expertise and patient values [11].”

The development of these CPGs on OIT followed the Guidelines International Network 2015 recommendations including a policy for the management of conflicts of interests [12]. They were commissioned by the Canadian Society of Allergy and Clinical Immunology (CSACI), and was developed through a collaboration with the University Hospital Center Sainte-Justine and the methodological support of the Institut National d’Excellence en Santé et Services Sociaux (INESSS). CSACI acted as the sponsor and was responsible for creating a diversified Working Group. This working group and the executive team opted for a reflective multicriteria methodology, which has been used in the past to develop CPGs [13] using the EVIDEM (Evidence and Values Impact on Decision Making) ethical framework [14,15,16,17]. This approach encourages the collection of data from diverse sources, including both the scientific literature and consultations, thus offering a 360° view of the subject. This framework was also used to organize, analyze and ultimately integrate the data into a deliberation guide, providing an efficient and consistent approach throughout the project.

Materials and methods

Multidisciplinary team

A multidisciplinary team comprised of experts from a diversity of fields was assembled, including allergists and other healthcare professionals, ethicists, and those with expertise in multicriteria deliberation methodology, literature review, consultations and sociology.

Multicriteria methodology

The aim of the methodological approach was to support reflection by all stakeholders during the development of fair and reasonable recommendations. To this end, a patient-centered and ethics-based multicriteria framework was used throughout the process. The framework was adapted from EVIDEM, included three additional patient focused criteria and organized 22 criteria into five dimensions defined to cover all relevant aspects of OIT. The framework also builds on the Accountability for reasonableness (A4R) conditions set forth by Daniels and Sabin [18] and the Triple Aim of healthcare systems set forth by Berwick and colleagues [19]. The five dimensions of the framework include:

  • How the use of OIT can contribute to a socio-politico-cultural context conducive to The Common Good (sociopolitical dimension),

  • How OIT contributes to a fair and equitable distribution of services (populational dimension),

  • How OIT responds to a need for health and well-being in an adequate way (clinical dimension),

  • How the use of OIT can contribute to improve the quality, organization and governance of healthcare services (organizational dimension), and

  • How OIT optimizes the use of resources and the associated costs to ensure sustainability of healthcare systems (economic dimension).

Each dimension contains criteria for operationalization, organized into a multicriteria grid. Data from the literature review and consultations were collected and organized per criterion to allow for a comprehensive assessment of OIT and provide a knowledge platform for the development of the CPG recommendations.

All documentation was compiled using a data management software program (CITAVI), which was adapted to organize data by criteria and provided an interactive interface for review by the working group.

Conflict of interest management and respect of persons

To avoid the impact of COI or conflicts of roles on the process, explicit guidelines regarding their declaration, evaluation and management were developed based on the GIN 2015 recommendations (see Additional file 1: Appendix 1) [12]. Each potential participant was required to complete a conflict of interest declaration, which was then analyzed by an ethicist. Participants who were deemed at risk of bias on a particular subject were excluded from the room during the deliberation meeting for the time during which the related recommendations were being discussed.

Every step was taken to ensure that best practices for respect of persons were carried out, with respectful atmosphere created by chairs for each group discussion and by interviewers for each individual interview; as well documentation was provided ahead of time to give each participant time to prepare. All patients completed an informed consent; they were informed that only their perspective on the topic was solicited, not personal clinical data, and that psychological support was available.

Literature review

Sources of data

An extensive literature review was performed. The primary sources of data were peer-reviewed full-text publications retrieved through searching multiple sources, including PubMed, MEDLINE (Ovid), Embase, EBM Reviews, CINAHL and Web of Science bibliographic databases. Searches for OIT clinical data were performed on April 17–18, 2019, and searches for epidemiological and burden of disease data were performed on May 30–31, 2019. Search terms are presented in Additional file 1: Appendix 2A. These searches aimed to collect data for all dimensions of the framework, including the sociopolitical, populational, clinical, organizational and economic dimensions. Bibliographical database searches were supplemented by individually searching the tables of contents of pertinent recent periodicals (up to September 2019), as well as bibliographies of key publications. Websites from government agencies, HTA bodies, professional associations and patient associations were also consulted. In order to capture the perception of OIT in the media, a review of the Canadian news media of the last 5 years was conducted via the FACTIVA Global News Monitoring & Search Engine using ‘oral immunotherapy’ and ‘immunothérapie orale’ (French) as search terms.

Data selection for each dimension

Retrieved records were screened based on title and abstract to select those for further assessment in full-text format. For the clinical dimension, patient focus groups or surveys providing insight on the importance of the therapy and its outcomes for patients, were considered for inclusion in addition to clinical studies (see details below), qualitative studies, and published CPGs. For the sociopolitical dimension, different types of publications on relevant topics such as the history of OIT and regulatory aspects were reviewed. Stakeholder’s positions were collected and informed by the media press review. For the populational dimension, records such as recent systematic reviews of epidemiological studies, original studies conducted in large, representative populations, preferably in the Canadian context, and recent reviews of qualitative research were considered for inclusion. For the organizational dimension, data concerning the healthcare system’s organization and requirements for the provision of OIT was sought. For the economic dimension, data collected included economic studies, such as cost of illness studies or economic analyses, and cost data (i.e., the costs of products and procedures for OIT). An analysis of associated ethical issues was included for each criterion throughout the framework, as applicable.

Selection of clinical studies and assessment of their methodological quality

Clinical studies were selected using eligibility (inclusion/exclusion) criteria listed in Additional file 1: Appendix 2B. Study designs included experimental, comparative studies, randomized [RCTs], non-randomized controlled clinical trials [CCTs], and observational studies reporting outcomes from clinical practice. RCTs are best suited to answer the questions whether an intervention is efficacious and safe, compared to another intervention or management option. These, however, can be limited in their ability to answer certain specific questions that arise in clinical practice, including how patient characteristics may impact outcomes or how to adapt to what happens over the course of treatment [20]. These limits stem from highly selective patient eligibility criteria, rigid study protocols, non-patient-centered end-points, short follow-up durations or small population sizes. A blinded study design, in particular, is not best suited to study personalized care, nor to investigate the impact of a treatment on patient-reported quality of life outcomes—examples include topics such as food-allergy related anxiety and the burden of allergen avoidance, both of which require awareness of the treatment received and the results achieved (e.g., level of desensitization). Observational studies can complement experimental comparative studies, especially when they involve larger less strictly selected patient populations that are closely followed over a long-term horizon in a real world practice setting.

The methodological quality of RCTs was assessed according to the Cochrane risk of bias approach [21], using published risk of bias assessments from systematic reviews, whenever available [22,23,24]. For case series, the Institute of Health Economics Quality Appraisal of Case Series Studies Checklist was applied [25]. Among its 20 items, 10 core items on study conduct and reporting were selected to operationalize quality assessment. A case series was deemed of high methodological quality (low risk of bias) if all 10 were positive, moderate if 8 to 9 were positive, and low (high risk of bias) if less than 8 were positive. The methodological quality of meta-analyses was determined by checking whether all eligible studies had been included, and by repeating key analyses based on the data reported in the original publications.

Extraction, analysis and synthesis

Clinical study data was extracted and synthesised in evidence tables in a per-criterion report format. The extraction tool was validated by a clinical expert. The report was validated by two assessors and then reviewed and revised by the Working Group. Clinical data was further synthesized and integrated into the multicriteria grid for the deliberation. Data pertaining to the other dimensions were directly synthesized in the multicriteria grid.

Consultations

Objective

The objective of the consultation process was to capture relevant experiential and contextual data from diverse perspectives to develop a comprehensive understanding of the topic. Data for each dimension of the multicriteria grid was collected through discussions with allergists, patients and other healthcare professionals. Healthcare professionals were able to provide insight into two main aspects: first, to validate and enrich the data collected through the literature review, and second, enhance the data with relevant clinical practice experience that might not have been elucidated from published studies. Patients with food allergy contributed their unique perspective of the condition and its impact, as well as their view and experience of food allergy therapy, including its benefits and its constraints.

General approach

In order to ensure diversity of participants, an open call for applications was posted on the website of applicable patient associations and the CSACI, and targeted calls were made by sending e-mail invitations to relevant professional societies and all CSACI members. The calls provided a short text explaining the objectives of the project, as well as the goals and format of the consultation, followed by a link to a small questionnaire covering the necessary information to enable selecting a diversity of participants to be consulted. The recruitment process was completed by applying a chain-referral sampling strategy. For each potential consultant, the geographical location, gender, general opinion of OIT and, when relevant, the type of practice (e.g. academic vs community, pediatric vs adult) were considered for participant diversification.

Selected participants were contacted and, upon confirmation of participation, asked to complete COI and consent forms. Subsequent steps differed depending on the format of the consultation for each specific group. Possible steps were discussion panels, individual interviews and an online questionnaire. Discussion panels served to create a debate around the various aspects of OIT from different contexts and viewpoints. Individual interviews allowed for the collection of in-depth data from a specific context and its implications. The online questionnaire allowed for the collection of a broad range of experience from a greater number of experts than would have been possible through discussion panels and interviews alone. For every consultation, participants received a consultation guide with questions adapted to their contexts and expertise, which was based on the multicriteria grid and grouped by criteria, in order to facilitate data collection and analysis. All participants were provided with the consultation document a week prior to the consultation in order to give time to familiarize themselves with it. During the discussion panels, the Chatham House Rule [26] was applied, which guarantees anonymity of all information provided by participants. These panels were led by co-chairs experienced in data appropriation while fostering an environment of respect, attentiveness and constructive exchanges.

Allergists

Consultations with allergists were twofold, with two different groups. A panel discussion was first conducted with the 15 Working Group members to identify key points on which to focus the CPGs. The group was well diversified in terms of experience with OIT: some participants had never offered it, some had offered it only in research settings, and those offering it in clinical practice reported using different methods and protocols. The discussion followed the multicriteria questionnaire, discussing every issue per criteria. Data was collected through recording and notetaking. Secondly, 42 CSACI members completed an online questionnaire, following the same structure. The data obtained from both the panel discussion and the questionnaire were compiled per criteria. Key themes within each criterion were identified through thematic analysis.

Patients and caregivers

Consultations with patients also occurred in two formats to fit two purposes. A panel discussion was held with 8 participants, diversified according to their individual context pertaining to OIT, which included whether or not they had direct experience with it, what their vision of the therapy was and the type and impact of their food allergy condition. Discussions were guided by the multicriteria grid. Patient perspective narrative data was collected for each of the criteria through recording and notetaking. The second consultation format consisted of individual interviews with 6 participants, following the same diversification criteria and using the same questionnaire as the panel discussion. The data from both sources was then compiled in the same way as the allergist consultations and analyzed thematically to identify key points raised by the patients.

Other healthcare professionals and lay representatives

These consultations served the purpose of obtaining the viewpoints of stakeholders involved in the care of food allergy patients other than allergists, namely family physicians, pediatricians, nurses, registered dieticians, psychologists and caregivers engaged in peer support activities. They were consulted via a discussion panel involving 10 participants diversified by their professions and the differences in their experience with OIT, which followed the same format as the patient panel. Another two healthcare professionals completed an online questionnaire. Representatives from patient associations were also interviewed, via phone, using the same questionnaire as the panel, to provide patient-centered contextual knowledge on OIT. This data was collected through notes and recording, per criteria, compiled in the same way as for the other consulted groups, and thematically analyzed, bringing forward essential notions associated with OIT provision to include in the CPGs.

Data from milieu of care

Data from Canadian clinical practices (milieu of care) was collected in the form of economic data provided by the working group members from their clinical practice offer of OIT, which was extracted and compiled into criteria of the economic dimension.

Deliberation

Data integration

Compiled data from the literature review, consultations and milieu of care were integrated into the deliberation guide in a highly synthesized format in order to facilitate comprehension of the data by all participants during the deliberation. Prior to this integration, the data from the literature review was revised and enriched by the Working Group. Data for each criterion was separated into three sections: literature review data, consultation data and ethical aspects. The deliberation guide consisted of the synthesized data integrated into a multicriteria grid, with an additional column in which participants were invited to add their interpretation of the data provided in order to facilitate discussion. Empty recommendation boxes at the end of each dimension were also included to allow discussion and the formation of informed recommendations for one subject at a time.

Deliberation process

The participants in the deliberation included the allergists of the Working Group along with selected participants from the consultations, in order to provide relevant data from a global patient perspective. These included two patients with different and longstanding experiences, two family physicians, a pediatrician, a nurse, a registered dietician, a pharmacist and a peer supporter. Each participant received a copy of the deliberation guide 1 week prior to the meeting. The deliberation meeting was chaired by three methodologists who were experts in multicriteria deliberation, multicriteria literature review and synthesis and multicriteria consultations, for a total of 22 participants. The deliberation was divided into four sessions according to the dimensions of the multicriteria grid, during which the literature review and consultation data were presented by the co-chairs. Corresponding recommendations were discussed and determined at the end of each session. All recommendations were based on a group consensus—recommendations for which a COI was identified were based on a consensus of the group excluding those with a COI.

Rationales for recommendations

Recommendations were based on a variety of rationales that included evidence from consultations and scientific studies, ethical imperatives, such as promotion of equity or patient autonomy, and other considerations, such as general standards of clinical care, clinical reasoning, and biological plausibility.

For recommendations that included evidence from clinical studies, the strength of the evidence supporting the recommendation was determined using a method that builds on the approach of the Oxford Centre for Evidence-Based Medicine [8, 27]. The risk of bias was assessed using the Cochrane approach [21] and the Institute for Health Economics (IHE) method for case series [25]. These methods, developed for quantitative data, were adapted to include qualitative data from consultations such that the strength of the evidence supporting the recommendation takes into account:

  • risk of bias (study design, including the methodology chose for data collection and analysis can affect the risk of misleading results);

  • type of study (meta-analyses, RCTs, non-randomized controlled trials (CCTs), case series); studies could be carried out in usual clinical practice or in a research context;

  • consistency of evidence across studies; and

  • the level of coherence between evidence from studies and data obtained from the consultation process.

The amount, quality and consistency of the evidence supporting the recommendation is defined at three levels:

  • High Large amount of consistent evidence from RCTs (or meta-analyses) and large studies in clinical practice, ideally at a low risk of bias; AND coherence with data from consultations and/or qualitative studies.

  • Moderate Moderate amount of consistent evidence from RCTs (or meta-analyses) and/or studies in clinical practice, ideally at a moderate or low risk of bias; AND coherence with data from consultations and/or qualitative studies.

  • Low Small amount of evidence OR evidence with some incoherence in data from RCTs (or meta-analyses) and/or studies in clinical practice OR data at moderate to high risk of bias; AND coherence with data from consultations and/or qualitative studies.

Strength of recommendations The strength of recommendations in CPGs is often graded based on the quality of clinical evidence regarding the efficacy and safety of an intervention. However, this approach does not apply to recommendations that do not rest on clinical trial outcomes, but for which the body of evidence from clinical research and practice shows a clear clinical benefit because conducting such trials would neither be reasonable nor ethical [28]. Moreover, grading in such a way does not take into account factors other than clinical outcomes, such as ethical imperatives, social context or economic considerations, which can be key elements of the rationale underlying a recommendation. Therefore, to ensure that all types of recommendations in these CPGs will be regarded on an equal footing, the strength of recommendations was not given a rating. Rather, in the spirit of accountability for reasonableness (A4R) [18], the rationale for each recommendation, the level of supporting evidence, where appropriate, and the necessary contextualization and nuances were all clearly stated.

Results

Multicriteria grid

The multicriteria grid used for this project included five dimensions divided into 22 criteria and is shown in Fig. 1.

Fig. 1
figure 1

Multicriteria grid: dimensions and criteria

Data used as basis for recommendations

The literature review yielded a total of 8157 records; 468 of them were assessed for eligibility in full-text records and 145 were included in the multicriteria grid (Fig. 2). An additional 17 articles were included from the media press review.

Fig. 2
figure 2

PRISMA diagram

A total of 14 patients or caregivers, 13 allergists and 16 other healthcare professionals or patient association representatives were consulted through panel discussions or individual interviews. In addition, 42 CSACI allergists responded to the online consultation survey.

Data on the economic aspects of OIT was available from three Canadian practices, and data on quality of life impact of OIT was collected from one practice.

The synthesis of the data collected through the literature review, consultations and from the milieu of care is presented by criteria along with complete references in the deliberation guide (Table 1). Detailed clinical evidence tables with results of quality assessments are available in Additional file 1: Appendix 3.

Table 1 Multicriteria grid with data from literature review, consultations and milieu of care synthesized by criteria and used for the deliberation

Deliberation guide

The deliberation guide includes the synthesized data for each criterion along with a section prompting participants to interpret and discuss the data. Boxes for the development of recommendations were added at the end of each dimension.

Recommendations

Recommendations are presented in the following format:

  1. 1.

    A lay summary of the data synthesis that led to the recommendation(s) written in a format that is accessible to a non-physician audience (for the clinical dimension, detailed narrative summaries of the data are available in Additional file 1: Appendix 4).

  2. 2.

    Additional key points discussed during deliberation that led to the recommendation(s)’ development.

  3. 3.

    Recommendations and rationales.

To facilitate the understanding of the presented data, key concepts and definitions pertaining to OIT are illustrated in Fig. 3.

Fig. 3
figure 3

Key concepts and definitions pertaining to OIT

Sociopolitical dimension: promotion of the common good

Populational dimension: promotion of equity

Clinical dimension: promotion of health and well-being

Organizational dimension: promotion optimized organization of care

Economic dimension: promotion of sustainability

Discussion

This development process of these CPGs consisted of a comprehensive approach to both the data collection and the collection of perspectives from all stakeholders. This allowed for the collaborative development of 38 recommendations guided by the five principles of the common good, equity, health and wellbeing, optimized organization of care and sustainability of healthcare systems, allowing a 360° view of OIT.

This comprehensive approach included the full extent of a traditional review of the available clinical data. The large number of both RCTs and diverse observational studies provided a rich body of complementary evidence. In addition, the inclusion of patients in both the consultation and the deliberation phases provided key experiential knowledge and contributed to the interpretation of data and the development of recommendations. Similar involvement of first line and allied healthcare professionals ensured a multidisciplinary perspective for optimal organization and provision of care as well as efficient use of healthcare resources. Throughout the aforementioned steps, the multicriteria methodology drove the systematic collection of scientific and experiential knowledge for all dimensions, the sociopolitical, populational, clinical, organizational and economic. This was instrumental in developing balanced recommendations allowing us to address issues beyond what is usually covered by traditional CPGs.

This was critical in the specific context of OIT, which poses challenges beyond its clinical aspects. Offering OIT requires adapting the organization of care in a setting of highly limited resources. It marks a rupture with a culture focusing on avoiding reactions at all cost, which creates confusion and tensions. Competing developments as a drug-based versus a food-based therapy also create a number of ethical issues. The methodology provides an ethics-based framework to identify and address these issues in the elaboration of recommendations.

The 360° approach necessitates a team with diverse expertise in various methodological domains and fields of knowledge to collect and integrate the body of knowledge in the multicriteria grid. Expertise in communicating in lay language and chairing a discussion on all these aspects is essential to ensure an understanding by all. The inclusion of participants with diverging perspectives is important to stimulate individual and group reflection during discussions. A tailored multidisciplinary team and a method that allows for efficient organization of data per criteria are essential to the process and key to promote understanding of numerous complex concepts and fulfill the A4R conditions.

Accountability for Reasonableness (A4R) defines four main conditions that can enhance legitimacy and help stakeholders develop a mutual basis for decision making: publicity of rationales for choices; relevance of criteria agreed to by a broad range of stakeholders; revisability of the decision in light of new evidence or arguments; and enforcement that means the other conditions are met [14]. The multicriteria approach facilitates meting these four conditions. The use and publication of the multicriteria grid completed with the data, from which detailed and lay rationales were developed, addresses the publicity and the relevance conditions. The multicriteria grid also offers a pragmatic tool to update the CPGs as significant advancements in knowledge emerges, which contributes to revisability. The implication of a large number of stakeholders, of methodologists and ethicists, the participation of external observers at the deliberation, as well as oversight by the CSACI board contributed to the enforcement of these three conditions.

Clinical recommendations developed here are generally concordant with those from the European Academy of Allergy and Clinical Immunology (EAACI) [8]. That said, in addition to the recommendations made outside the clinical dimension, there are some noteworthy differences between the CPGs, which likely stem from the difference in methodology. The authors of the EAACI guidelines explicitly stated in their discussion that “there is no evidence that the efficacy and safety are affected by the type and nature of the food allergen used in allergen immunotherapy”. However, their methodology resulted in limiting the indication for OIT to milk, egg and peanut only. The current guidelines diverge from those from EEACI in a few other recommendations, namely regarding the indication for OIT in children less than 4 years of age or in adults and the recognition that OIT can promote sustained unresponsiveness in some patients, over the long term.

There was also a difference in the overall vision surrounding the development of OIT. In the EAACI guidelines, the need for standardized protocols and products was identified as a high priority, while these CPGs emphasize that there is a need for more personalization. Standardization is often seen as a desirable objective in modern medicine because it allows generating quantitative data that can be used as the basis for more recommendations using a traditional CPG methodology. However, the ability to adapt protocols to patients’ needs is essential in OIT. Standardized products and protocols have not been shown to provide clinical benefits over their adaptable alternatives and can actually increase barriers and costs, thus threatening sustainability and access.

Personalized approaches, however, represent a challenge for the transfer of expertise. The development of training initiatives, clinical tools, and organizational models adapted to the delivery of personalized care should therefore be made a priority. Shared decision-making tools should include clear information on food allergy management, consent forms, home dosing instructions, and a reaction management plan. Specific training tools directed toward allergists, primary care physicians and allied healthcare professionals should focus on the biological, psychological and social aspects of the therapy and be included in training curricula and continuing professional development (CPD).

Clinical tools will be developed collaboratively as a second step of these CPGs and made available online at http://www.csaci.ca/OIT-guidelines.

These CPGs also identified a number of data gaps that require further research, including:

  • long-term patient-centered outcomes

  • underrepresented groups, e.g. adults

  • optimal use of biomarkers to guide therapy

  • optimal use of medication to support treatment

  • determinants of reactivity, e.g. cofactors, individual differences in absorption and distribution of allergens

  • benefits and optimal use of nutritional, psychological, and peer support

  • economic and organizational aspects of the treatment.

Conclusion

These guidelines bring to the forefront the critical importance and value of placing patients at the center of the development of clinical practice guidelines. Here, this approach was instrumental to developing recommendations for the responsible implementation of OIT in clinical practice, adapted to individual patient needs. The multicriteria approach offers an alternative to technocentric approaches in CPG development by balancing human, ethical and technical considerations in decision making (Fig. 4).

Fig. 4
figure 4

Considerations for balanced decision-making in medicine

Technocentric approaches tend to create a pressure to standardize patient care in order to generate quantitative data, which may not always be in patients’ best interest. Rather than adapting patient care to meet methodological needs for quantitative data, the methodology should be adapted to patients’ needs for best care. As the saying goes, ‘Not everything that counts can be counted, and not everything that can be counted counts [75].’

Availability of data and materials

Additional data is included in the supplemental repository. Clinical tools for implementation in practice will be made available at http://www.csaci.ca/OIT.

Abbreviations

A4R:

Accountability for reasonableness

CCT:

Controlled clinical trial

CPG:

Clinical practice guidelines

CSACI:

Canadian Society of Allergy and Clinical Immunology

EAACI:

European Academy of Allergy and Clinical Immunology

EoE:

Eosinophilic esophagitis

EPIT:

Epicutaneous Immunotherapy

IHE:

Institute for Health Economics

OIT:

Oral immunotherapy

RCT:

Randomized controlled clinical trial

SLIT:

Sub-lingual Immunotherapy

References

  1. Moen OL, Opheim E, Trollvik A. Parents experiences raising a child with food allergy; a qualitative review. J Pediatr Nurs. 2019;46:e52–63. https://doi.org/10.1016/j.pedn.2019.02.036.

    Article  PubMed  Google Scholar 

  2. Chow C, Pincus DB, Comer JS. Pediatric food allergies and psychosocial functioning: examining the potential moderating roles of maternal distress and overprotection. J Pediatr Psychol. 2015;40(10):1065–74. https://doi.org/10.1093/jpepsy/jsv058.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Cortes A, Castillo A, Sciaraffia A. Food allergy: children’s symptom levels are associated with mothers’ psycho-socio-economic variables. J Psychosom Res. 2018;104:48–54. https://doi.org/10.1016/j.jpsychores.2017.11.009.

    Article  PubMed  Google Scholar 

  4. Johnson SF, Woodgate RL. Qualitative research in teen experiences living with food-induced anaphylaxis: a meta-aggregation. J Adv Nurs. 2017;73(11):2534–46. https://doi.org/10.1111/jan.13325.

    Article  PubMed  Google Scholar 

  5. Protudjer JL, Jansson SA, Middelveld R, et al. Impaired health-related quality of life in adolescents with allergy to staple foods. Clin Transl Allergy. 2016;6:37. https://doi.org/10.1186/s13601-016-0128-5.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Institute for Clinical and Economic Review. Oral immunotherapy and Viaskin® peanut for peanut allergy: effectiveness and value: Institute for Clinical and Economic Review. 2019.

  7. Schofield AT. A case of egg poisoning. Lancet. 1908;171(4410):716. https://doi.org/10.1016/S0140-6736(00)67313-0.

    Article  Google Scholar 

  8. Pajno GB, Fernandez-Rivas M, Arasi S, et al. EAACI Guidelines on allergen immunotherapy: IgE-mediated food allergy. Allergy. 2018;73(4):799–815. https://doi.org/10.1111/all.13319.

    Article  CAS  PubMed  Google Scholar 

  9. Martorell A, Alonso E, Echeverria L, et al. Oral immunotherapy for food allergy: a Spanish guideline. Immunotherapy egg and milk Spanish guide (ITEMS guide). Part I: cow milk and egg oral immunotherapy: introduction, methodology, rationale, current state, indications, contraindications, and oral immunotherapy build-up phase. J Investig Allergol Clin Immunol. 2017;27(4):225–37. https://doi.org/10.18176/jiaci.0177.

    Article  CAS  PubMed  Google Scholar 

  10. Martorell A, Alonso E, Echeverria L, et al. Oral Immunotherapy for food allergy: a Spanish guideline. egg and milk immunotherapy Spanish guide (ITEMS GUIDE). Part II: maintenance phase of cow milk (CM) and egg oral immunotherapy (OIT), special treatment dosing schedules. Models of dosing schedules of OIT with CM and egg. J Investig Allergol Clin Immunol. 2017;27(5):279–90. https://doi.org/10.18176/jiaci.0178.

    Article  CAS  PubMed  Google Scholar 

  11. Pattani R, Straus SE. What is EBM? 2019. https://bestpractice.bmj.com/info/toolkit/learn-ebm/what-is-ebm/. Accessed 22 Nov 2019.

  12. Schunemann HJ, Al-Ansary LA, Forland F, et al. Guidelines International Network: principles for disclosure of interests and management of conflicts in guidelines. Ann Intern Med. 2015;163(7):548–53. https://doi.org/10.7326/M14-1885.

    Article  PubMed  Google Scholar 

  13. Deal CL, Tony M, Hoybye C, et al. GrowthHormone Research Society workshop summary: consensus guidelines for recombinant human growth hormone therapy in Prader-Willi syndrome. J Clin Endocrinol Metab. 2013;98(6):E1072–87. https://doi.org/10.1210/jc.2012-3888.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Goetghebeur M, Castro-Jaramillo H, Baltussen R, et al. The art of priority setting. Lancet. 2017;389(10087):2368–9. https://doi.org/10.1016/S0140-6736(17)31573-8.

    Article  PubMed  Google Scholar 

  15. Goetghebeur M, Wagner M. Identifying value(s): a reflection on the ethical aspects of MCDA in healthcare decisionmaking. In: Marsh K, Goetghebeur M, Thokala P, et al., editors. Multi-criteria decision analysis to support healthcare decisions. Cham: Spinger Nature; 2017. p. 29–46.

    Chapter  Google Scholar 

  16. Goetghebeur MM, Cellier MS. Can reflective multicriteria be the new paradigm for healthcare decision-making? The EVIDEM journey. Cost Eff Resour Alloc. 2018;16(Suppl 1):54. https://doi.org/10.1186/s12962-018-0116-9.

    Article  PubMed  PubMed Central  Google Scholar 

  17. The EVIDEM framework. 2019. http://www.evidem.org/evidem-framework/. Accessed 22 Nov 2019.

  18. Daniels N, Sabin J. Limits to health care: fair procedures, democratic deliberation, and the legitimacy problem for insurers. Philos Public Aff. 1997;26(4):303–50.

    Article  Google Scholar 

  19. Berwick DM, Nolan TW, Whittington J. The triple aim: care, health, and cost. Health Aff. 2008;27(3):759–69. https://doi.org/10.1377/hlthaff.27.3.759.

    Article  Google Scholar 

  20. Horwitz RI, Hayes-Conroy A, Caricchio R, et al. From evidence based medicine to medicine based evidence. Am J Med. 2017;130(11):1246–50. https://doi.org/10.1016/j.amjmed.2017.06.012.

    Article  PubMed  Google Scholar 

  21. Higgins JPT, Green S (eds). Cochrane handbook for systematic reviews of interventions version 5.1.0 [updated March 2011]. The Cochrane Collaboration; 2011. https://handbook-5-1.cochrane.org/.

  22. Nurmatov U, Dhami S, Arasi S, et al. Allergen immunotherapy for IgE-mediated food allergy: a systematic review and meta-analysis. Allergy. 2017;72(8):1133–47. https://doi.org/10.1111/all.13124.

    Article  CAS  PubMed  Google Scholar 

  23. Romantsik O, Tosca MA, Zappettini S, et al. Oral and sublingual immunotherapy for egg allergy. Cochrane Database Syst Rev. 2018;4:CD010638. https://doi.org/10.1002/14651858.CD010638.pub3.

    Article  PubMed  Google Scholar 

  24. Chu DK, Wood RA, French S, et al. Oral immunotherapy for peanut allergy (PACE): a systematic review and meta-analysis of efficacy and safety. Lancet. 2019. https://doi.org/10.1016/S0140-6736(19)30420-9.

    Article  PubMed  Google Scholar 

  25. Institute of Health Economics (IHE). Quality Appraisal of Case Series Studies Checklist, 2014.

  26. Chatham House The Royal Institute of International Affairs. Chatham House Rule: Chatham House; 2019. https://www.chathamhouse.org/chatham-house-rule#. Accessed 13 Dec 2019.

  27. Centre for Evidence-based Medicine. Oxford Centre for evidence-based medicine—Levels of Evidence (March 2009). 2009. https://www.cebm.net/2009/06/oxford-centre-evidence-based-medicine-levels-evidence-march-2009/.

  28. Guyatt G, Oxman AD, Akl EA, et al. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J Clin Epidemiol. 2011;64(4):383–94. https://doi.org/10.1016/j.jclinepi.2010.04.026.

    Article  PubMed  Google Scholar 

  29. Wasserman RL, Factor JM, Baker JW, et al. Oral immunotherapy for peanut allergy: multipractice experience with epinephrine-treated reactions. J Allergy Clin Immunol Pract. 2014;2(1):91–6. https://doi.org/10.1016/j.jaip.2013.10.001.

    Article  PubMed  Google Scholar 

  30. Soller L, Abrams EM, Carr S, et al. First real-world safety analysis of preschool peanut oral immunotherapy. J Allergy Clin Immunol Pract. 2019. https://doi.org/10.1016/j.jaip.2019.04.010.

    Article  PubMed  Google Scholar 

  31. Martin-Munoz MF, Belver MT, Alonso Lebrero E, et al. Egg oral immunotherapy in children (SEICAP I): daily or weekly desensitization pattern. Pediatr Allergy Immunol. 2019;30(1):81–92. https://doi.org/10.1111/pai.12974.

    Article  PubMed  Google Scholar 

  32. Levy MB, Elizur A, Goldberg MR, et al. Clinical predictors for favorable outcomes in an oral immunotherapy program for IgE-mediated cow’s milk allergy. Ann Allergy Asthma Immunol. 2014;112(1):58–63.e1. https://doi.org/10.1016/j.anai.2013.10.001.

    Article  CAS  PubMed  Google Scholar 

  33. Kauppila TK, Paassilta M, Kukkonen AK, et al. Outcome of oral immunotherapy for persistent cow’s milk allergy from 11 years of experience in Finland. Pediatr Allergy Immunol. 2019. https://doi.org/10.1111/pai.13025.

    Article  PubMed  Google Scholar 

  34. Nowak-Wegrzyn A, Wood RA, Nadeau KC, et al. Multicenter, randomized, double-blind, placebo-controlled clinical trial of vital wheat gluten oral immunotherapy. J Allergy Clin Immunol. 2019;143(2):651–661.e9. https://doi.org/10.1016/j.jaci.2018.08.041.

    Article  CAS  PubMed  Google Scholar 

  35. Kulmala P, Pelkonen AS, Kuitunen M, et al. Wheat oral immunotherapy was moderately successful but was associated with very frequent adverse events in children aged 6–18 years. Acta Paediatr. 2018;107(5):861–70. https://doi.org/10.1111/apa.14226.

    Article  CAS  PubMed  Google Scholar 

  36. Elizur A, Appel MY, Nachshon L, et al. Walnut oral immunotherapy for desensitisation of walnut and additional tree nut allergies (Nut CRACKER): a single-centre, prospective cohort study. Lancet Child Adolesc Health. 2019;3(5):312–21. https://doi.org/10.1016/s2352-4642(19)30029-x.

    Article  PubMed  Google Scholar 

  37. Barni S, Mori F, Piccorossi A, et al. Low-dose oral food challenge with hazelnut: efficacy and tolerability in children. Int Arch Allergy Immunol. 2019;178(1):97–100. https://doi.org/10.1159/000493019.

    Article  CAS  PubMed  Google Scholar 

  38. Nachshon L, Goldberg MR, Levy MB, et al. Efficacy and safety of sesame oral immunotherapy—a real-world, single-center study. J Allergy Clin Immunol Pract. 2019. https://doi.org/10.1016/j.jaip.2019.05.031.

    Article  PubMed  Google Scholar 

  39. De Schryver S, Mazer B, Clarke AE, et al. Adverse events in oral immunotherapy for the desensitization of cow’s milk allergy in children: a randomized controlled trial. J Allergy Clin Immunol Pract. 2019. https://doi.org/10.1016/j.jaip.2019.02.007.

    Article  PubMed  Google Scholar 

  40. Wasserman RL, Hague AR, Pence DM, et al. Real-world experience with peanut oral immunotherapy: lessons learned from 270 patients. J Allergy Clin Immunol Pract. 2019;7(2):418–426.e4. https://doi.org/10.1016/j.jaip.2018.05.023.

    Article  PubMed  Google Scholar 

  41. Longo G, Barbi E, Berti I, et al. Specific oral tolerance induction in children with very severe cow’s milk-induced reactions. J Allergy Clin Immunol. 2008;121(2):343–7. https://doi.org/10.1016/j.jaci.2007.10.029.

    Article  CAS  PubMed  Google Scholar 

  42. Pajno GB, Caminiti L, Ruggeri P, et al. Oral immunotherapy for cow’s milk allergy with a weekly up-dosing regimen: a randomized single-blind controlled study. Ann Allergy Asthma Immunol. 2010;105(5):376–81. https://doi.org/10.1016/j.anai.2010.03.015.

    Article  CAS  PubMed  Google Scholar 

  43. Bird JA, Spergel JM, Jones SM, et al. Efficacy and safety of AR101 in oral immunotherapy for peanut allergy: results of ARC001, a randomized, double-blind, placebo-controlled phase 2 clinical trial. J Allergy Clin Immunol Pract. 2018;6(2):476–85.e3. https://doi.org/10.1016/j.jaip.2017.09.016.

    Article  PubMed  Google Scholar 

  44. Fauquert JL, Michaud E, Pereira B, et al. Peanut gastrointestinal delivery oral immunotherapy in adolescents: results of the build-up phase of a randomized, double-blind, placebo-controlled trial (PITA study). Clin Exp Allergy. 2018;48(7):862–74. https://doi.org/10.1111/cea.13148.

    Article  CAS  PubMed  Google Scholar 

  45. Narisety SD, Frischmeyer-Guerrerio PA, Keet CA, et al. A randomized, double-blind, placebo-controlled pilot study of sublingual versus oral immunotherapy for the treatment of peanut allergy. J Allergy Clin Immunol. 2015;135(5):1275–1282.e1–6. https://doi.org/10.1016/j.jaci.2014.11.005.

    Article  CAS  PubMed  Google Scholar 

  46. Tang ML, Ponsonby AL, Orsini F, et al. Administration of a probiotic with peanut oral immunotherapy: a randomized trial. J Allergy Clin Immunol. 2015;135(3):737–744.e8. https://doi.org/10.1016/j.jaci.2014.11.034.

    Article  CAS  PubMed  Google Scholar 

  47. Vickery BP, Vereda A, Casale TB, et al. AR101 oral immunotherapy for peanut allergy. N Engl J Med. 2018;379(21):1991–2001. https://doi.org/10.1056/NEJMoa1812856.

    Article  PubMed  Google Scholar 

  48. Varshney P, Jones SM, Scurlock AM, et al. A randomized controlled study of peanut oral immunotherapy: clinical desensitization and modulation of the allergic response. J Allergy Clin Immunol. 2011;127(3):654–60. https://doi.org/10.1016/j.jaci.2010.12.1111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Burks AW, Jones SM, Wood RA, et al. Oral immunotherapy for treatment of egg allergy in children. N Engl J Med. 2012;367(3):233–43. https://doi.org/10.1056/NEJMoa1200435.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Jones SM, Burks AW, Keet C, et al. Long-term treatment with egg oral immunotherapy enhances sustained unresponsiveness that persists after cessation of therapy. J Allergy Clin Immunol. 2016;137(4):1117–27.e10. https://doi.org/10.1016/j.jaci.2015.12.1316.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hsiao KC, Ponsonby AL, Axelrad C, et al. Long-term clinical and immunological effects of probiotic and peanut oral immunotherapy after treatment cessation: 4-year follow-up of a randomised, double-blind, placebo-controlled trial. Lancet Child Adolesc Health. 2017;1(2):97–105. https://doi.org/10.1016/s2352-4642(17)30041-x.

    Article  PubMed  Google Scholar 

  52. Meglio P, Giampietro PG, Carello R, et al. Oral immunotherapy in children with IgE-mediated hen’s egg allergy: follow-ups at 25 and 7 years. Allergy Rhinol. 2017;8(3):157–69. https://doi.org/10.2500/ar.2017.8.0211.

    Article  Google Scholar 

  53. Elizur A, Appel MY, Goldberg MR, et al. Clinical and laboratory 2-year outcome of oral immunotherapy in patients with cow’s milk allergy. Allergy. 2016;71(2):275–8. https://doi.org/10.1111/all.12794.

    Article  CAS  PubMed  Google Scholar 

  54. Nachshon L, Goldberg MR, Katz Y, et al. Long-term outcome of peanut oral immunotherapy-real-life experience. Pediatr Allergy Immunol. 2018;29(5):519–26. https://doi.org/10.1111/pai.12914.

    Article  PubMed  Google Scholar 

  55. Martin-Munoz MF, Alonso Lebrero E, Zapatero L, et al. Egg OIT in clinical practice (SEICAP II): maintenance patterns and desensitization state after normalizing the diet. Pediatr Allergy Immunol. 2019;30(2):214–24. https://doi.org/10.1111/pai.13002.

    Article  PubMed  Google Scholar 

  56. Sampson HA, Leung DYM, Jones SM, et al. Egg oral immunotherapy (OIT) induces more rapid desensitization and sustained unresponsiveness (SU) in egg-allergic, baked-egg tolerant children than the addition of daily baked-egg products. J Allergy Clin Immunol. 2019;143(2):AB255. https://doi.org/10.1016/j.jaci.2018.12.779.

    Article  Google Scholar 

  57. Takahashi M, Taniuchi S, Soejima K, et al. Two-weeks-sustained unresponsiveness by oral immunotherapy using microwave heated cow’s milk for children with cow’s milk allergy. Allergy Asthma Clin Immunol. 2016;12(1):44. https://doi.org/10.1186/s13223-016-0150-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Vickery BP, Berglund JP, Burk CM, et al. Early oral immunotherapy in peanut-allergic preschool children is safe and highly effective. J Allergy Clin Immunol. 2017;139(1):173–81.e8. https://doi.org/10.1016/j.jaci.2016.05.027.

    Article  CAS  PubMed  Google Scholar 

  59. Escudero C, Rodriguez Del Rio P, Sanchez-Garcia S, et al. Early sustained unresponsiveness after short-course egg oral immunotherapy: a randomized controlled study in egg-allergic children. Clin Exp Allergy. 2015;45(12):1833–43. https://doi.org/10.1111/cea.12604.

    Article  CAS  PubMed  Google Scholar 

  60. Reier-Nilsen T, Michelsen MM, Lodrup Carlsen KC, et al. Feasibility of desensitizing children highly allergic to peanut by high-dose oral immunotherapy. Allergy. 2019;74(2):337–48. https://doi.org/10.1111/all.13604.

    Article  CAS  PubMed  Google Scholar 

  61. Blumchen K, Trendelenburg V, Ahrens F, et al. Efficacy, safety, and quality of life in a multicenter, randomized, placebo-controlled trial of low-dose peanut oral immunotherapy in children with peanut allergy. J Allergy Clin Immunol Pract. 2019;7(2):479–91.e10. https://doi.org/10.1016/j.jaip.2018.10.048.

    Article  PubMed  Google Scholar 

  62. Morisset M, Moneret-Vautrin DA, Guenard L, et al. Oral desensitization in children with milk and egg allergies obtains recovery in a significant proportion of cases A randomized study in 60 children with cow’s milk allergy and 90 children with egg allergy. Eur Ann Allergy Clin Immunol. 2007;39(1):12–9.

    CAS  PubMed  Google Scholar 

  63. Martorell A, De la Hoz B, Ibanez MD, et al. Oral desensitization as a useful treatment in 2-year-old children with cow’s milk allergy. Clin Exp Allergy. 2011;41(9):1297–304. https://doi.org/10.1111/j.1365-2222.2011.03749.x.

    Article  CAS  PubMed  Google Scholar 

  64. Anagnostou K, Islam S, King Y, et al. Assessing the efficacy of oral immunotherapy for the desensitisation of peanut allergy in children (STOP II): a phase 2 randomised controlled trial. Lancet. 2014;383(9925):1297–304. https://doi.org/10.1016/s0140-6736(13)62301-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Fuentes-Aparicio V, Alvarez-Perea A, Infante S, et al. Specific oral tolerance induction in paediatric patients with persistent egg allergy. Allergol Immunopathol. 2013;41(3):143–50. https://doi.org/10.1016/j.aller.2012.02.007.

    Article  CAS  Google Scholar 

  66. Caminiti L, Pajno GB, Crisafulli G, et al. Oral immunotherapy for egg allergy: a double-blind placebo-controlled study, with postdesensitization follow-up. J Allergy Clin Immunol Pract. 2015;3(4):532–9. https://doi.org/10.1016/j.jaip.2015.01.017.

    Article  PubMed  Google Scholar 

  67. Burks A, Casale T, Beyer K, et al. Age-related findings from the peanut allergy oral immunotherapy study of Ar101 for desensitization (Palisade) study. Ann Allergy Asthma Immunol. 2018;121(5 Supplement):S4.

    Article  Google Scholar 

  68. Begin P, Winterroth LC, Dominguez T, et al. Safety and feasibility of oral immunotherapy to multiple allergens for food allergy. Allergy Asthma Clin Immunol. 2014;10(1):1. https://doi.org/10.1186/1710-1492-10-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Begin P, Dominguez T, Wilson SP, et al. Phase 1 results of safety and tolerability in a rush oral immunotherapy protocol to multiple foods using Omalizumab. Allergy Asthma Clin Immunol. 2014;10(1):7. https://doi.org/10.1186/1710-1492-10-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Eapen AA, Lavery WJ, Siddiqui JS, et al. Oral Immunotherapy for multiple foods in a pediatric allergy clinic setting. Ann Allergy Asthma Immunol. 2019. https://doi.org/10.1016/j.anai.2019.08.463.

    Article  PubMed  Google Scholar 

  71. Andorf S, Purington N, Block WM, et al. Anti-IgE treatment with oral immunotherapy in multifood allergic participants: a double-blind, randomised, controlled trial. Lancet Gastroenterol Hepatol. 2018;3(2):85–94. https://doi.org/10.1016/S2468-1253(17)30392-8.

    Article  PubMed  Google Scholar 

  72. Elizur A, Goldberg MR, Levy MB, et al. Oral immunotherapy in cow’s milk allergic patients: course and long-term outcome according to asthma status. Ann Allergy Asthma Immunol. 2015;114(3):240–44.e1. https://doi.org/10.1016/j.anai.2014.12.006.

    Article  CAS  PubMed  Google Scholar 

  73. MacGinnitie AJ, Rachid R, Gragg H, et al. Omalizumab facilitates rapid oral desensitization for peanut allergy. J Allergy Clin Immunol. 2017;139(3):873–881.e8. https://doi.org/10.1016/j.jaci.2016.08.010.

    Article  CAS  PubMed  Google Scholar 

  74. Howe LC, Leibowitz KA, Perry MA, et al. Changing patient mindsets about non-life-threatening symptoms during oral immunotherapy: a randomized clinical trial. J Allergy Clin Immunol Pract. 2019. https://doi.org/10.1016/j.jaip.2019.01.022.

    Article  PubMed  Google Scholar 

  75. Wb C. Informal sociology, a casual introduction to sociological thinking. New York: Random House; 1963. p. 13.

    Google Scholar 

  76. HealthLink BC. Reducing risk of food allergy in your baby British Columbia: HealthLink BC; 2018. https://www.healthlinkbc.ca/healthy-eating/reducing-baby-food-allergy-risk. Accessed 10 Oct 2019.

  77. Haeusermann T. I can’t eat that: the sociology behind the rise in food allergies and intolerances. Curr Sociol. 2015;63(3):369–86. https://doi.org/10.1177/0011392114559847.

    Article  Google Scholar 

  78. Wasserman RL, Jones DH, Windom HH. Oral immunotherapy for food allergy: the FAST perspective. Ann Allergy Asthma Immunol. 2018;121(3):272–5. https://doi.org/10.1016/j.anai.2018.06.011.

    Article  PubMed  Google Scholar 

  79. Perkin MR. Oral desensitization to peanuts. N Engl J Med. 2018;379(21):2074–5. https://doi.org/10.1056/NEJMe1813314.

    Article  PubMed  Google Scholar 

  80. Hamblin J. The U.S. health-care system found a way to make peanuts cost $4,200. The Atlantic, 2019.

  81. Blaiss MS, Tilles S, Petroni D, et al. Current management and use of oral immunotherapy in the United States for patients with peanut allergy. Allergy Asthma Proc. 2019. https://doi.org/10.2500/aap.2019.40.4228.

    Article  PubMed  Google Scholar 

  82. Arasi S, Corsello G, Villani A, et al. The future outlook on allergen immunotherapy in children: 2018 and beyond. Ital J Pediatr. 2018;44(1):80. https://doi.org/10.1186/s13052-018-0519-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Greenhawt MJ, Vickery BP. Allergist-reported trends in the practice of food allergen oral immunotherapy. J Allergy Clin Immunol Pract. 2015;3(1):33–8. https://doi.org/10.1016/j.jaip.2014.06.023.

    Article  PubMed  Google Scholar 

  84. Soller L, Abrams EM, Chan ES. An update on the controversy around offering oral immunotherapy to peanut-allergic children outside of research. Ann Allergy Asthma Immunol. 2019. https://doi.org/10.1016/j.anai.2019.02.011.

    Article  PubMed  Google Scholar 

  85. Greenhawt M, Hourihane JO. Looking into the seeds of time: are home-based OIT programs the shape of things to come for selected patients? Ann Allergy Asthma Immunol. 2018;120(5):453–4. https://doi.org/10.1016/j.anai.2017.11.004.

    Article  PubMed  Google Scholar 

  86. Mansfield LE. Oral immunotherapy for peanut allergy in clinical practice is ready. Allergy Asthma Proc. 2013;34(3):205–9. https://doi.org/10.2500/aap.2013.34.3666.

    Article  PubMed  Google Scholar 

  87. Umasunthar T, Leonardi-Bee J, Hodes M, et al. Incidence of fatal food anaphylaxis in people with food allergy: a systematic review and meta-analysis. Clin Exp Allergy. 2013;43(12):1333–41. https://doi.org/10.1111/cea.12211.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Shaker MS, Schwartz J, Ferguson M. An update on the impact of food allergy on anxiety and quality of life. Curr Opin Pediatr. 2017;29(4):497–502. https://doi.org/10.1097/mop.0000000000000509.

    Article  PubMed  Google Scholar 

  89. Xu YS, Kastner M, Harada L, et al. Anaphylaxis-related deaths in Ontario: a retrospective review of cases from 1986 to 2011. Allergy Asthma Clin Immunol. 2014;10(1):38. https://doi.org/10.1186/1710-1492-10-38.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Versluis A, Knulst AC, Kruizinga AG, et al. Frequency, severity and causes of unexpected allergic reactions to food: a systematic literature review. Clin Exp Allergy. 2015;45(2):347–67. https://doi.org/10.1111/cea.12328.

    Article  CAS  PubMed  Google Scholar 

  91. Cherkaoui S, Ben-Shoshan M, Alizadehfar R, et al. Accidental exposures to peanut in a large cohort of Canadian children with peanut allergy. Clin Transl Allergy. 2015;5:16. https://doi.org/10.1186/s13601-015-0055-x.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Umasunthar T, Leonardi-Bee J, Turner PJ, et al. Incidence of food anaphylaxis in people with food allergy: a systematic review and meta-analysis. Clin Exp Allergy. 2015;45(11):1621–36. https://doi.org/10.1111/cea.12477.

    Article  CAS  PubMed  Google Scholar 

  93. O’Keefe A, Clarke A, St Pierre Y, et al. The risk of recurrent anaphylaxis. J Pediatr. 2017;180:217–21. https://doi.org/10.1016/j.jpeds.2016.09.028.

    Article  PubMed  Google Scholar 

  94. Kimchi N, Clarke A, Moisan J, et al. Anaphylaxis cases presenting to primary care paramedics in Quebec. Immun Inflamm Dis. 2015;3(4):406–10. https://doi.org/10.1002/iid3.78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Hochstadter E, Clarke A, De Schryver S, et al. Increasing visits for anaphylaxis and the benefits of early epinephrine administration: a 4-year study at a pediatric emergency department in Montreal, Canada. J Allergy Clin Immunol. 2016;137(6):1888–1890.e4. https://doi.org/10.1016/j.jaci.2016.02.016.

    Article  PubMed  Google Scholar 

  96. Ben-Shoshan M, La Vieille S, Eisman H, et al. Anaphylaxis treated in a Canadian pediatric hospital: incidence, clinical characteristics, triggers, and management. J Allergy Clin Immunol. 2013;132(3):739–741.e3. https://doi.org/10.1016/j.jaci.2013.06.016.

    Article  PubMed  Google Scholar 

  97. Asai Y, Yanishevsky Y, Clarke A, et al. Rate, triggers, severity and management of anaphylaxis in adults treated in a Canadian emergency department. Int Arch Allergy Immunol. 2014;164(3):246–52. https://doi.org/10.1159/000365631.

    Article  PubMed  Google Scholar 

  98. Chooniedass R, Temple B, Martin D, et al. A qualitative study exploring parents’ experiences with epinephrine use for their child’s anaphylactic reaction 11 Medical and Health Sciences 1117 Public Health and Health Services. Clin Transl Allergy. 2018;8(1):1–9. https://doi.org/10.1186/s13601-018-0230-y.

    Article  Google Scholar 

  99. Patel N, Herbert L, Green TD. The emotional, social, and financial burden of food allergies on children and their families. Allergy Asthma Proc. 2017;38(2):88–91. https://doi.org/10.2500/aap.2017.38.4028.

    Article  PubMed  Google Scholar 

  100. Fong AT, Katelaris CH, Wainstein B. Bullying and quality of life in children and adolescents with food allergy. J Paediatr Child Health. 2017;53(7):630–5. https://doi.org/10.1111/jpc.13570.

    Article  PubMed  Google Scholar 

  101. Dean J, Fenton NE, Shannon S, et al. Disclosing food allergy status in schools: health-related stigma among school children in Ontario. Health Soc Care Community. 2016;24(5):e43–52. https://doi.org/10.1111/hsc.12244.

    Article  PubMed  Google Scholar 

  102. Protudjer JL, Jansson SA, Heibert Arnlind M, et al. Household costs associated with objectively diagnosed allergy to staple foods in children and adolescents. J Allergy Clin Immunol Pract. 2015;3(1):68–75. https://doi.org/10.1016/j.jaip.2014.09.021.

    Article  PubMed  Google Scholar 

  103. Protudjer JL, Jansson SA, Ostblom E, et al. Health-related quality of life in children with objectively diagnosed staple food allergy assessed with a disease-specific questionnaire. Acta Paediatr. 2015;104(10):1047–54. https://doi.org/10.1111/apa.13044.

    Article  CAS  PubMed  Google Scholar 

  104. Indinnimeo L, Baldini L, De Vittori V, et al. Duration of a cow-milk exclusion diet worsens parents’ perception of quality of life in children with food allergies. BMC Pediatr. 2013;13:7. https://doi.org/10.1186/1471-2431-13-203.

    Article  Google Scholar 

  105. Soller L, Clarke AE, Lyttle A, et al. Comparing quality of life in Canadian children with peanut, sesame, and seafood allergy. J Allergy Clin Immunol Pract. 2019. https://doi.org/10.1016/j.jaip.2019.07.006.

    Article  PubMed  Google Scholar 

  106. Thornqvist V, Middelveld R, Wai HM, et al. Health-related quality of life worsens by school age amongst children with food allergy. Clin Transl Allergy. 2019;9:10. https://doi.org/10.1186/s13601-019-0244-0.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Soller L, Ben-Shoshan M, Harrington DW, et al. Adjusting for nonresponse bias corrects overestimates of food allergy prevalence. J Allergy Clin Immunol Pract. 2015;3(2):291–293.e2. https://doi.org/10.1016/j.jaip.2014.11.006.

    Article  PubMed  Google Scholar 

  108. CHU St-Justine, Université de Montréal. Clinique d’immunothérapie orale (CITO) - Coûts des phases 1 à 3 : du pilote au transfert de connaissances et à une offre de soins et services de réseau provincial. Montreal, 2016.

  109. DunnGalvin A, Chang WC, Laubach S, et al. Profiling families enrolled in food allergy immunotherapy studies. Pediatrics. 2009;124(3):e503–9. https://doi.org/10.1542/peds.2008-3642.

    Article  PubMed  Google Scholar 

  110. Tackett AP, Farrow ML, McQuaid EL. Food security, utilization of food assistance programs, and caregiver perceptions of food-induced anaphylaxis risk in children with food allergies. Pediatr Allergy Immunol Pulmonol. 2018;31(2):91–6. https://doi.org/10.1089/ped.2017.0857.

    Article  Google Scholar 

  111. Sharma HP, Herbert LJ. Food allergy: psychosocial impact and public policy implications. Chem Immunol Allergy. 2015;101:221–6. https://doi.org/10.1159/000373907.

    Article  PubMed  Google Scholar 

  112. Harrington DW, Elliott SJ, Clarke AE, et al. Exploring the determinants of the perceived risk of food allergies in Canada. Hum Ecol Risk Assess. 2012;18(6):1338–58. https://doi.org/10.1080/10807039.2012.722857.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Graham F, Begin P. L’immunothérapie orale pour le traitement des allergies alimentaires: L’Association des Allergologues et Immunologues du Québec, 2016.

  114. Begin P, Paradis L, Paradis J, et al. Natural resolution of peanut allergy: a 12-year longitudinal follow-up study. J Allergy Clin Immunol Pract. 2013;1(5):528–530.e1–4. https://doi.org/10.1016/j.jaip.2013.05.008.

    Article  PubMed  Google Scholar 

  115. Abrams EM, Sicherer SH. Diagnosis and management of food allergy. Can Med Assoc J. 2016;188(15):1087–93. https://doi.org/10.1503/cmaj.160124.

    Article  Google Scholar 

  116. Arasi S, Caminiti L, Crisafulli G, et al. A general strategy for de novo immunotherapy design: the active treatment of food allergy. Expert Rev Clin Immunol. 2018;14(8):665–71. https://doi.org/10.1080/1744666x.2018.1498784.

    Article  CAS  PubMed  Google Scholar 

  117. Remington BC, Baumert JL, Blom WM, et al. Unintended allergens in precautionary labelled and unlabelled products pose significant risks to UK allergic consumers. Allergy. 2015;70(7):813–9. https://doi.org/10.1111/all.12625.

    Article  CAS  PubMed  Google Scholar 

  118. Blom WM, Michelsen-Huisman AD, van Os-Medendorp H, et al. Accidental food allergy reactions: products and undeclared ingredients. J Allergy Clin Immunol. 2018;142(3):865–75. https://doi.org/10.1016/j.jaci.2018.04.041.

    Article  PubMed  Google Scholar 

  119. Prell C, Koletzko B. Restrictive diets risk of malnutrition and preventive options. Monschr Kinderheilkd. 2014;162(6):503–10. https://doi.org/10.1007/s00112-013-3070-9.

    Article  Google Scholar 

  120. Lavine E, Clarke A, Joseph L, et al. Peanut avoidance and peanut allergy diagnosis in siblings of peanut allergic children. Clin Exp Allergy. 2015;45(1):249–54. https://doi.org/10.1111/cea.12403.

    Article  CAS  PubMed  Google Scholar 

  121. Venter C, Mazzocchi A, Maslin K, et al. Impact of elimination diets on nutrition and growth in children with multiple food allergies. Curr Opin Allergy Clin Immunol. 2017;17(3):220–6. https://doi.org/10.1097/ACI.0000000000000358.

    Article  PubMed  Google Scholar 

  122. Nachshon L, Goldberg MR, Schwartz N, et al. Decreased bone mineral density in young adult IgE-mediated cow’s milk-allergic patients. J Allergy Clin Immunol. 2014;134(5):1108–1113.e3. https://doi.org/10.1016/j.jaci.2014.06.026.

    Article  CAS  PubMed  Google Scholar 

  123. Shanahan L, Zucker N, Copeland WE, et al. Are children and adolescents with food allergies at increased risk for psychopathology? J Psychosom Res. 2014;77(6):468–73. https://doi.org/10.1016/j.jpsychores.2014.10.005.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Cannon HE. The economic impact of peanut allergies. Am J Manag Care. 2018;24(19 Suppl):S428–33.

    PubMed  Google Scholar 

  125. Marchisotto MJ, Harada L, Kamdar O, et al. Food allergen labeling and purchasing habits in the United States and Canada. J Allergy Clin Immunol Pract. 2017;5(2):345–351.e2. https://doi.org/10.1016/j.jaip.2016.09.020.

    Article  PubMed  Google Scholar 

  126. Tilles SA, Petroni D. FDA-approved peanut allergy treatment: the first wave is about to crest. Ann Allergy Asthma Immunol. 2018;121(2):145–9. https://doi.org/10.1016/j.anai.2018.06.005.

    Article  PubMed  Google Scholar 

  127. Fleischer DM, Greenhawt M, Sussman G, et al. Effect of epicutaneous immunotherapy vs placebo on reaction to peanut protein ingestion among children with peanut allergy: the PEPITES randomized clinical trial. JAMA. 2019. https://doi.org/10.1001/jama.2019.1113.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Scurlock AM. Oral and sublingual immunotherapy for treatment of IgE-mediated food allergy. Clin Rev Allergy Immunol. 2018;55(2):139–52. https://doi.org/10.1007/s12016-018-8677-0.

    Article  CAS  PubMed  Google Scholar 

  129. Kim EH, Yang L, Ye P, et al. Long-term sublingual immunotherapy for peanut allergy in children: clinical and immunologic evidence of desensitization. J Allergy Clin Immunol. 2019. https://doi.org/10.1016/j.jaci.2019.07.030.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Upton J, Nowak-Wegrzyn A. The impact of baked egg and baked milk diets on IgE- and non-IgE-mediated allergy. Clin Rev Allergy Immunol. 2018;55(2):118–38. https://doi.org/10.1007/s12016-018-8669-0.

    Article  CAS  PubMed  Google Scholar 

  131. Amat F, Kouche C, Gaspard W, et al. Is a slow-progression baked milk protocol of oral immunotherapy always a safe option for children with cow’s milk allergy? A randomized controlled trial. Clin Exp Allergy. 2017;47(11):1491–6. https://doi.org/10.1111/cea.13022.

    Article  CAS  PubMed  Google Scholar 

  132. Murdoch B, Adams EM, Caulfield T. The law of food allergy and accommodation in Canadian schools. Allerg Asthma Clin Immunol. 2018;14:8. https://doi.org/10.1186/s13223-018-0273-6.

    Article  Google Scholar 

  133. Wood RA, Kim JS, Lindblad R, et al. A randomized, double-blind, placebo-controlled study of omalizumab combined with oral immunotherapy for the treatment of cow’s milk allergy. J Allergy Clin Immunol. 2016;137(4):1103–1110.e11. https://doi.org/10.1016/j.jaci.2015.10.005.

    Article  CAS  PubMed  Google Scholar 

  134. Aimmune Therapeutics. Allergenic Products Advisory Committee Meeting (APAC) briefing document—peanut, arachis hypogaea, allergen powder for oral administration (Proposed Trade Name: Palforzia), 2019.

  135. Cox L, Larenas-Linnemann D, Lockey RF, et al. Speaking the same language: the world allergy organization subcutaneous immunotherapy systemic reaction grading system. J Allergy Clin Immunol. 2010;125(3):569–574.e1–e7. https://doi.org/10.1016/j.jaci.2009.10.060.

    Article  Google Scholar 

  136. Sampson HA. Anaphylaxis and emergency treatment. Pediatrics. 2003;111(6 Pt 3):1601–8.

    PubMed  Google Scholar 

  137. Burks AW, Jones SM, Plaut M. Oral immunotherapy for egg allergy in children REPLY. N Engl J Med. 2012;367(15):1472–3.

    CAS  Google Scholar 

  138. Lucendo AJ, Arias A, Tenias JM. Relation between eosinophilic esophagitis and oral immunotherapy for food allergy: a systematic review with meta-analysis. Ann Allergy Asthma Immunol. 2014;113(6):624–9. https://doi.org/10.1016/j.anai.2014.08.004.

    Article  PubMed  Google Scholar 

  139. Echeverria-Zudaire LA, Fernandez-Fernandez S, Rayo-Fernandez A, et al. Primary eosinophilic gastrointestinal disorders in children who have received food oral immunotherapy. Allergol Immunopathol. 2016;44(6):531–6. https://doi.org/10.1016/j.aller.2016.05.002.

    Article  CAS  Google Scholar 

  140. Gomez Torrijos E, Mendez Diaz Y, Moreno Lozano L, et al. Frequency and course of eosinophilic esophagitis during oral immunotherapy for cow’s milk allergy in a series of 57 children. J Investig Allergol Clin Immunol. 2017;27(2):132–3. https://doi.org/10.18176/jiaci.0130.

    Article  CAS  PubMed  Google Scholar 

  141. Rodriguez CG, Torrijos EG, De la Pinzon FR, et al. Dysphagia in a boy treated with oral immunotherapy for cow’s milk allergy. J Investig Allergol Clin Immunol. 2014;24(5):363–5.

    PubMed  Google Scholar 

  142. Hill DA, Dudley JW, Spergel JM. The prevalence of eosinophilic esophagitis in pediatric patients with IgE-mediated food allergy. J Allergy Clin Immunol Pract. 2017;5(2):369–75. https://doi.org/10.1016/j.jaip.2016.11.020.

    Article  PubMed  Google Scholar 

  143. Goldberg MR, Elizur A, Nachshon L, et al. Oral immunotherapy-induced gastrointestinal symptoms and peripheral blood eosinophil responses. J Allergy Clin Immunol. 2017;139(4):1388–1390.e4. https://doi.org/10.1016/j.jaci.2016.09.053.

    Article  PubMed  Google Scholar 

  144. Goldberg MR, Nachshon L, Levy MB, et al. Risk factors and treatment outcomes for oral immunotherapy-induced gastrointestinal symptoms and eosinophilic responses (OITIGER). J Allergy Clin Immunol Pract. 2019. https://doi.org/10.1016/j.jaip.2019.07.034.

    Article  PubMed  Google Scholar 

  145. Manabe T, Sato S, Yanagida N, et al. Long-term outcomes after sustained unresponsiveness in patients who underwent oral immunotherapy for egg, cow’s milk, or wheat allergy. Allergol Int. 2019. https://doi.org/10.1016/j.alit.2019.02.012.

    Article  PubMed  Google Scholar 

  146. Barbi E, Longo G, Berti I, et al. Adverse effects during specific oral tolerance induction: in-hospital “rush” phase. Eur Ann Allergy Clin Immunol. 2012;44(1):18–25.

    CAS  PubMed  Google Scholar 

  147. Virkud YV, Burks AW, Steele PH, et al. Novel baseline predictors of adverse events during oral immunotherapy in children with peanut allergy. J Allergy Clin Immunol. 2017;139(3):882–88.e5. https://doi.org/10.1016/j.jaci.2016.07.030.

    Article  PubMed  Google Scholar 

  148. Barbi E, Longo G, Berti I, et al. Adverse effects during specific oral tolerance induction: in home phase. Allergol Immunopathol. 2012;40(1):41–50. https://doi.org/10.1016/j.aller.2011.05.004.

    Article  CAS  Google Scholar 

  149. Vazquez-Ortiz M, Alvaro M, Piquer M, et al. Life-threatening anaphylaxis to egg and milk oral immunotherapy in asthmatic teenagers. Ann Allergy Asthma Immunol. 2014;113(4):482–4. https://doi.org/10.1016/j.anai.2014.07.010.

    Article  PubMed  Google Scholar 

  150. Pajno GB, Caminiti L, Salzano G, et al. Comparison between two maintenance feeding regimens after successful cow’s milk oral desensitization. Pediatr Allergy Immunol. 2013;24(4):376–81. https://doi.org/10.1111/pai.12077.

    Article  PubMed  Google Scholar 

  151. Sampson HA. Peanut oral immunotherapy: is it ready for clinical practice? J Allergy Clin Immunol Pract. 2013;1(1):15–21. https://doi.org/10.1016/j.jaip.2012.10.009.

    Article  PubMed  Google Scholar 

  152. Sato S, Yanagida N, Ebisawa M. Recent trends in the management of food allergy. Curr Allergy Clin Immunol. 2018;31(3):138–41.

    Google Scholar 

  153. Remington BC, Krone T, Koppelman SJ. Quantitative risk reduction through peanut immunotherapy: safety benefits of an increased threshold in Europe. Pediatr Allergy Immunol. 2018;29(7):762–72. https://doi.org/10.1111/pai.12961.

    Article  PubMed  Google Scholar 

  154. Baumert JL, Taylor SL, Koppelman SJ. Quantitative assessment of the safety benefits associated with increasing clinical peanut thresholds through immunotherapy. J Allergy Clin Immunol Pract. 2018;6(2):457–65.e4. https://doi.org/10.1016/j.jaip.2017.05.006.

    Article  PubMed  Google Scholar 

  155. Kao LM, Greenhawt MJ, Warren CM, et al. Parental and parent-perceived child interest in clinical trials for food allergen immunotherapy. Ann Allergy Asthma Immunol. 2018;120(3):331–33.e1. https://doi.org/10.1016/j.anai.2017.12.012.

    Article  PubMed  Google Scholar 

  156. Greenhawt M, Marsh R, Gilbert H, et al. Understanding caregiver goals, benefits, and acceptable risks of peanut allergy therapies. Ann Allergy Asthma Immunol. 2018;121(5):575–9. https://doi.org/10.1016/j.anai.2018.06.018.

    Article  PubMed  Google Scholar 

  157. Dunlop JH, Keet CA. Goals and motivations of families pursuing oral immunotherapy for food allergy. J Allergy Clin Immunol Pract. 2019;7(2):662–63.e18. https://doi.org/10.1016/j.jaip.2018.05.035.

    Article  PubMed  Google Scholar 

  158. Sato S, Sugizaki C, Yanagida N, et al. Nationwide questionnaire-based survey of oral immunotherapy in Japan. Allergol Int. 2018;67(3):399–404. https://doi.org/10.1016/j.alit.2018.02.006.

    Article  PubMed  Google Scholar 

  159. Traister RS, Green TD, Mitchell L, et al. Community opinions regarding oral immunotherapy for food allergies. Ann Allergy Asthma Immunol. 2012;109(5):319–23. https://doi.org/10.1016/j.anai.2012.08.012.

    Article  PubMed  Google Scholar 

  160. Dunn Galvin A, McMahon S, Ponsonby AL, et al. The longitudinal impact of probiotic and peanut oral immunotherapy on health-related quality of life. Allergy. 2018;73(3):560–8. https://doi.org/10.1111/all.13330.

    Article  CAS  PubMed  Google Scholar 

  161. Epstein-Rigbi N, Goldberg MR, Levy MB, et al. Quality of life of food-allergic patients before, during, and after oral immunotherapy. J Allergy Clin Immunol Pract. 2019;7(2):429–436.e2. https://doi.org/10.1016/j.jaip.2018.06.016.

    Article  PubMed  Google Scholar 

  162. Factor JM, Mendelson L, Lee J, et al. Effect of oral immunotherapy to peanut on food-specific quality of life. Ann Allergy Asthma Immunol. 2012;109(5):348–52.e2. https://doi.org/10.1016/j.anai.2012.08.015.

    Article  CAS  PubMed  Google Scholar 

  163. Dufresne E, Poder TG, Lacombe-Barrios J, et al. Quality-adjusted life-year gain with oral immunotherapy: preliminary results from a real-life cohort. Montreal: CSACI Annual Scientific Meeting; 2019.

    Google Scholar 

  164. Reier-Nilsen T, Carlsen KCL, Michelsen MM, et al. Parent and child perception of quality of life in a randomized controlled peanut oral immunotherapy trial. Pediatr Allergy Immunol. 2019. https://doi.org/10.1111/pai.13066.

    Article  PubMed  Google Scholar 

  165. Danchin M, De Bono N, Allen K, et al. Managing simple food allergy in community settings: a pilot study investigating a new model of care. J Paediatr Child Health. 2016;52(3):315–20. https://doi.org/10.1111/jpc.13026.

    Article  PubMed  Google Scholar 

  166. Nachshon L, Goldberg MR, Elizur A, et al. A Web site-based reporting system for monitoring home treatment during oral immunotherapy for food allergy. Ann Allergy Asthma Immunol. 2015;114(6):510–5. https://doi.org/10.1016/j.anai.2015.04.007.

    Article  PubMed  Google Scholar 

  167. Leroux H, Langlois A, Dion C, et al. Efficiency of peanut suspension with simple syrup compared to individual pre-weighted flour doses for oral immunotherapy. Montreal: CSACI Annual Scientific Meeting; 2019.

    Google Scholar 

  168. Minaker LM, Elliott SJ, Clarke A. Exploring low-income families’ financial barriers to food allergy management and treatment. J Allergy. 2014;2014:160363. https://doi.org/10.1155/2014/160363.

    Article  Google Scholar 

  169. Gupta R, Holdford D, Bilaver L, et al. The economic impact of childhood food allergy in the United States. JAMA Pediatr. 2013;167(11):1026–31. https://doi.org/10.1001/jamapediatrics.2013.2376.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge the other members of the deliberative committee. The deliberative committee was composed of members of the working group as well as A Boisvert, Peer supporter, Montréal, QC; MJ Cadieux, BPharm, Pharmacist and Peer supporter, University of Montreal, QC; B Francoeur, MD Family physician, Trois-Rivières, QC; J Garnavos; L Lavallée, RN, Allergy nurse, CHU Ste-Justine, Montréal, QC; G Parizeault MD, Pediatrician, Chicoutimi, QC; S Pernice, MSc, Nutritionist, CHU Ste-Justine, Montréal, QC; M Stasiuk; C Vaillancourt, MD, Family physician, St-John, NB.

Regarding methodological contributions, we are indebted to the ethics support of I Ganache, PhD, ethicist, Unit Methods, ethics and participation, INESSS, National Institute for excellence in health and social services, Montreal, QC, as well as to the contribution of S Nairn, PhD Department of Sociology, McGill University, Montreal QC, to patient consultation and literature review. We also would like to acknowledge V Berretta, PhD, Pavia University, Italy for setting up the multicriteria software interface (CITAVI), P Bush, PhD, INESSS, for clinical data extraction, P Dodin, librarian, Information Unit services, CHU Sainte-Justine, Montreal, QC, for the search strategy, A Maurice, Research center of the university hospital center Sainte-Justine, Montréal QC for IT support and L Tremblay, CSACI Executive Director, Canadian Society of Allergy and Clinical Immunology, Ottawa, ON, for administrative support.

Finally, we wish to acknowledge all the other participants to the consultations including C Avril; MJ Bettez, Peer supporter at Déjouez les allergies, Quebec, QC; G Briand; S Beugnot, PhD Psychologist, Montreal, QC; A Boisvert, Peer supporter, Montréal, QC; V Brousseau; P Chainé; L Couture; N Ford; J Gerdts, Food Allergy Canada; J Gomis, RN, Nurse, CHU Sherbrooke, QC; J Huang, Food Allergy Canada; L Lavallée, RN, Nurse, CHU Ste Justine, Montréal, QC; C Marcoux; H Matharu, MD, Family Physician, Toronto, ON; O Nazarko; S Pietrzykowski; S Sissons; MA Therrien; J Trudel MD, Pediatrician, Shawiningan, QC; W Toma, MD, Pediatrician Vancouver, BC; G Van Herck, Social worker & psychotherapist, Montreal, QC; G Zheng.

Funding

CSACI.

Author information

Authors and Affiliations

Authors

Contributions

P. Bégin, E. S. Chan, H. Kim, E. M. Abrams, M. Ben-Shoshan, S. B. Cameron, S. Carr, D. Fischer, A. Haynes, S. Kapur, M. N. Primeau, J. Upton, T. K. Vander Leek are members of the OIT CPG working group, in alphabetical order except for the executive team. MSC adapted the EVIDEM framework to transform it into a patient-centered ethical framework. MMG reorganized the criteria of the EVIDEM framework into five dimensions and adapted the framework to a qualitative MCDA approach (no weighting and no scoring) to deepen the reflective aspects of the framework. HK, ESC, PB, MW, MMG were responsible for executive decisions throughout the development of the clinical practice guidelines. MW performed the review of the literature to which all authors contributed, except for the ethical aspects, which was performed by CFG. MSC, MW, MMG and PB performed consultations and chaired consultations panels. MSC organized and analyzed the consultations. MMG performed the data integration. All authors had full access to all of the data collected through the literature review and consultations. MSC, MW, MMG and PB take responsibility for the accuracy of the data synthesis. All authors contributed to the analysis and interpretation of data. MSC, MW, MMG and PB drafted the deliberation guide. MSC, MW and MMG chaired the deliberation committee. All authors drafted the recommendations during the deliberation. MSC, MMG, MW, and PB drafted the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to P. Bégin.

Ethics declarations

Ethics approval and consent to participate

The study was performed according to the current practices at INESSS for clinical practice guidelines; patients consulted on their perspectives of the different dimensions of OIT completed an informed consent.

Consent for publication

Not applicable.

Competing interests

Direct competing interests related to the topic of the guidelines P. Bégin and J. Upton are non-remunerated investigators on an investigator-instigated trial sponsored by the Canadian Institutes of Health on the use of omalizumab in oral immunotherapy (in-kind contribution of investigative drug product by Novartis worth > $100,000 CAD). They were excluded from the room for the discussion and deliberations on recommendations related to the use of omalizumab in oral immunotherapy. M. Ben-Shoshan was a principal investigator on a trial on the use of pharmaceutical peanut flour preparations in OIT sponsored by Aimmune Therapeutics (> $100,000 CAD in grant support). J. Upton was a non-remunerated sub-investigator on clinical trial sponsored by Aimmune Therapeutics on the use of pharmaceutical peanut flour preparations in OIT. They were excluded from the room for the discussion and deliberations on recommendations related to the use of pharmaceutical food products in OIT.

Direct competing interests unrelated to the topic of the guidelines P. Begin reports speaker and/or advisor fees from Food Allergy Canada, Novartis, Pfizer, Sanofi, ALK and Aralez Pharmaceuticals, as well as research grant support from Canadian Institutes for Health Research, Fonds de Recherche en Santé du Québec, Canadian Allergy, Asthma and Immunology Foundation, DBV technologies, CHU Ste-Justine Foundation, Regeneron and Sanofi outside the submitted work. M. Ben-Shoshan reports advisor fees from Food Allergy Canada, as well as research grant support from Canadian Institutes for Health Research, Fonds de recherché en Santé du Québec and Canadian Foundation of Allergy and Clinical Immunology outside the submitted work. S. B. Cameron reports an unrestricted educational grant from Pfizer outside the submitted work. S. Carr reports speaker and/or advisor fees from Aralez, Pfizer, Nutricia, Meda (Mylan), Sanofi, GSK, ALK, and Pediapharm. E. S. Chan reports speaker and/or advisor fees from Pfizer, Kaleo, Food Allergy Canada, Pediapharm, Leo and DBV technologies as well as research grant support from DBV Technologies outside the submitted work. D. Fischer reports speaker and/or advisor fees from ALK, AstraZeneca, Aralez, Bausch Health, Merck, Mylan, Pfizer, Novartis, Pediapharm, Sanofi and Teva outside the submitted work. A. Haynes reports advisor fees from Sanofi outside the submitted work. S. Kapur reports speaker and/or advisor fees from Pediapharm and has been a member of advisory boards for, Bausch, Kaleo, Mylan, Novartis, Pediapharm, Pfizer, and Sanofi outside the submitted work. He also holds shares in ABK Biomedical, who are not involved in the field of allergy. H. Kim reports speaker and/or advisor fees for ALK-Abelló, Aralez, Astra Zeneca, CSL Behring, Shire, Novartis, Pediapharm, Stallergenes, Kaleo, Sanofi, Pfizer, and Mylan outside the submitted work. M. N. Primeau reports speaker and/or advisor fees from ALK, Allergie Québec, Aralez, Bausch Health, Food Allergy Canada, Mead Johnson, Mylan, Novartis, Nutricia, Pediapharm and Pfizer outside the submitted work. J. Upton reports advisor fees from Food Allergy Canada, ALK-Abelló, Kaleo, as well as research grant support from Toronto SickKids Foundation, DBV technologies and Regeneron outside the submitted work. T. K. Vander Leek reports speaker and/or advisor fees from Pediapharm, Pfizer and Aralez outside the submitted work. Other authors and members of the deliberative committee declared no financial conflicts of interest.

Indirect competing interests related to the topic of the guidelines E. M. Abrams is a pediatric allergist currently offering OIT in clinic. She has contributed to a national cohort study published on the topic. She is on the national advisory board of Food Allergy Canada. P. Begin is an adult-trained allergist currently offering OIT in the academic setting. He pursues clinical research projects on OIT. He is the director of a pilot public OIT program funded through philanthropy and government support. He collaborates with a parent committee doing fundraising for a public-funded OIT program in the province of Quebec. He is a clinician-scientist with a research program including clinical trials, epidemiologic and health technology assessment studies on food allergy and OIT. He has published original research as well as editorials and review articles on the topic of OIT. He is a member of the advisory board for Food Allergy Canada. He has given public and scientific lectures on the topic of OIT. M. Ben-Shoshan is a pediatric allergist currently not offering OIT outside research. He is a clinician-scientist with a research program including clinical trials OIT. He has previously published on the topic of OIT. He is on the national advisory board of Food Allergy Canada. A. Boisvert is a peer-supporter at byebyeallergies.ca and Food Allergy Canada. M. J. Cadieux, is a peer-supporter at Déjouer-les-allergies. S. B. Cameron is a pediatric allergist currently offering OIT in clinic. He has contributed to a national cohort study and a review published and has given scientific lectures on the topic. S. Carr is a pediatric allergist currently offering OIT in clinic. He has contributed to a national cohort study and a review published and has given scientific lectures on the topic. E. S. Chan is a pediatric allergist currently offering OIT in the academic setting. He collaborates with a parent committee doing fundraising for a public-funded OIT program in Vancouver. He as contributed to a national cohort study published on the topic. He is a member of the advisory board for Food Allergy Canada, is an eosinophilic esophagitis guideline member for the Joint Task Force/American Gastroenterological Association, and was an expert panel and coordinating committee member of the National Institute of Allergy and Infectious Diseases–sponsored Guidelines for Peanut Allergy Prevention. D. Fischer is an adult-trained allergist currently not offering OIT in clinic. He has given scientific lectures on the topic. B. Francoeur is a family physician from a region without allergist with a practice focus on allergy. He currently follows patients on OIT but does not initiate treatment himself. H. Kim is an adult-trained allergist currently offering OIT in clinic. He has published an editorial on the topic of OIT. He is the president of Canadian Society of Allergy and Clinical Immunology. A. Haynes is a pediatric allergist currently not offering OIT in clinic. S. Kapur is a pediatric allergist currently offering OIT in clinic. He has previously contributed to a national cohort study published on the topic. G. Parizeault is a pediatrician with a practice focus on allergy in a region without allergist. He is currently offering OIT in clinic. S. Pernice, is a registered dietician practicing in an academic allergy clinic offering OIT. M.N. Primeau is a pediatric allergist currently not offering OIT in clinic. J. Upton is an adult-trained allergists currently not offering OIT outside research. She is a clinician with a research program including clinical trials OIT. She is on the national advisory board of Food Allergy Canada. She has given public and scientific lectures on the topic of OIT. C. Vaillancourt is a family physician with a practice focus on allergy in a region without allergist. He is currently offering OIT in clinic. T. K. Vander Leek is a pediatric allergists currently offering OIT in clinic. He has previously contributed to a national cohort study published on the topic. Other authors and members of the deliberative committee declared no indirect conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Additional file 1.

Supplementary appendices.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bégin, P., Chan, E.S., Kim, H. et al. CSACI guidelines for the ethical, evidence-based and patient-oriented clinical practice of oral immunotherapy in IgE-mediated food allergy. Allergy Asthma Clin Immunol 16, 20 (2020). https://doi.org/10.1186/s13223-020-0413-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13223-020-0413-7

Keywords