Allergy (and clinical immunology)
16.7K views | +5 today
Follow
Allergy (and clinical immunology)
Ressources et Actualités pour la spécialité Allergie
Your new post is loading...
Your new post is loading...
Scooped by Gilbert C FAURE
April 10, 2019 5:05 AM
Scoop.it!

Peanut Allergen Inhibitor Developed Using Nanoparticles

Peanut Allergen Inhibitor Developed Using Nanoparticles | Allergy (and clinical immunology) | Scoop.it
Researchershave prevented the binding of peanut allergens with IgE to suppress the allergic reaction to peanuts using novel allergen-specific inhibitors.
No comment yet.
Rescooped by Gilbert C FAURE from Immunology and Biotherapies
February 18, 2019 2:03 AM
Scoop.it!

Allergen Immunotherapy for IgE‐Mediated Food Allergy: there is a measure in everything to a proper proportion of therapy - Pajno - - Pediatric Allergy and Immunology - Wiley Online Library

IgE‐mediated food allergy (FA) is a potentially life‐threatening condition with a negative impact on quality of life and an increasing prevalence in westernized countries in the recent two decades. A strict avoidance of the triggering food(s) represents the current standard approach. However, an elimination diet may be difficult and frustrating, in particular for common foods, (e.g. milk, egg, and peanut). Food allergy immunotherapy (FA‐AIT) may provide an active treatment that enables to increase the amount of food that the patient can intake without reaction during treatment (i.e. desensitization), and reduces the risk of potential life‐threatening allergic reaction in the event of accidental ingestion. However, several gaps need still to be filled. A memorable Latin orator stated: “Est modus in rebus” (Horace, Sermones I, 1, 106‐07). This sentence remembers that there is a measure in everything to a proper proportion of therapy. The common sense of measure should find application in each stage of treatment. A personalized approaching should consider the specific willing and features of each patient. Efforts are devoted to improve the efficacy, the safety but also the quality of life of patients suffering from FA. In the near future it will be important to clarify immunological pathways of FA‐AIT, and to identify reliable biomarkers in order to recognize the most suitable candidates to FA‐AIT and algorithms for treatments tailored on well‐characterized subpopulations of patients. This article is protected by copyright. All rights reserved.

Via Krishan Maggon
No comment yet.
Scooped by Gilbert C FAURE
February 6, 2019 9:09 AM
Scoop.it!

Gender, prick test size and rAra h 2 sIgE level may predict the eliciting dose in patients with peanut allergy: evidence from the Mirabel survey. - PubMed - NCBI

Gender, prick test size and rAra h 2 sIgE level may predict the eliciting dose in patients with peanut allergy: evidence from the Mirabel survey. - PubMed - NCBI | Allergy (and clinical immunology) | Scoop.it
Clin Exp Allergy. 2019 Jan 28. doi: 10.1111/cea.13348.[Epub ahead of print]...
No comment yet.
Scooped by Gilbert C FAURE
January 25, 2019 2:29 AM
Scoop.it!

Peanut Allergy in Spanish Children: Comparative Profile of Peanut Allergy versus Tolerance - Abstract - International Archives of Allergy and Immunology - Karger Publishers

Peanut Allergy in Spanish Children: Comparative Profile of Peanut Allergy versus Tolerance - Abstract - International Archives of Allergy and Immunology - Karger Publishers | Allergy (and clinical immunology) | Scoop.it
<b><i>Background:</i></b> Peanut storage proteins (Ara h 1, Ara h 2, and Ara h 3) have been described as the major peanut allergens in children, although not all peanut-sensitized individuals have cli...
No comment yet.
Scooped by Gilbert C FAURE
December 30, 2018 1:54 PM
Scoop.it!

Peanut allergen–specific antibodies go public

Peanut allergen–specific antibodies go public | Allergy (and clinical immunology) | Scoop.it

Changes in the human environment and activities over the past few decades have caused an epidemic of food allergies ([ 1 ][1]). People suffering from allergies often feel that they live on a cliff edge, as the allergens to which they react are potentially fatal ([ 2 ][2]). For example, tiny amounts of peanut picked up on skin or contaminating other foods can be dangerous to peanut-sensitized individuals ([ 2 ][2]–[ 4 ][3]). Immunoglobulin E (IgE) antibodies mediate the allergic response. They bind to specific receptors on inflammatory immune cells: mast cells in mucosal tissues lining body surfaces and cavities, and basophils in the circulation. These cells mediate allergic responses triggered by specific antigens (allergens) that are recognized by IgE. B cells expressing IgG antibodies have long served as the paradigm for the development of B cells into antibody-secreting plasma cells in the immune response. Until recently, the far less abundant IgE-expressing B cells have proved to be elusive. On page 1306 of this issue, Croote et al. ([ 5 ][4]) have analyzed single B cells from six individuals with peanut allergy, which enabled the identification of the natural Ig heavy- and light-chain pairs from IgE-expressing B cells that are responsible for peanut allergy. With this information they produced recombinant antibodies, identified the peanut allergen–specific antibodies, and used site-directed mutagenesis to suppress their activity. The mutated antibodies could be used to treat peanut allergy.

![Figure][5]

From sensitization to peanut allergy
Dendritic cells in the skin pick up peanut allergens and present them to peanut allergen–specific T helper 2 (TH2) cells, which in turn present them to B cells. Interaction between peanut allergen–specific TH2 cells and B cells solicits help from TH2 cells for B cell proliferation, somatic hypermutation and affinity maturation, class switching to IgE, and plasma cell differentiation. Allergen-specific IgE secreted by plasma cells binds to resident mast cells in the gut, so the ingestion of peanuts triggers an allergic reaction.

GRAPHIC: N. DESAI/ SCIENCE

Whole-exome sequencing of single B cells from peanut-allergic individuals yielded two principal components of gene expression, representing naïve or memory B cells and plasmablasts (the circulating precursors of plasma cells). The majority of Ig-Eexpressing cells were plasmablasts, whereas the majority of cells expressing IgG or IgA (the more abundant antibody classes) were naïve or memory B cells. It has previously been observed that IgE-expressing B cells tend to develop into the plasma cell lineage as opposed to the memory cell lineage. The IgE plasma cells inherit their antigen specificity from B cells of other antibody classes, which have undergone affinity maturation. This is advantageous for their biological function in immediate hypersensitivity to antigens as it cuts out the time that would be required for affinity maturation of IgE memory B cells ([ 6 ][6], [ 7 ][7]).

In immune responses, antigens bind to specific B cells expressing a membrane-bound form of the antibody [the B cell receptor (BCR)], which stimulates B cell maturation through the processes of somatic hypermutation (mutations affecting the antibody affinity for antigen) and affinity maturation (the selection of cells expressing BCRs with the highest affinity for antigen). The cells may also undergo class switching (from IgM to IgG, IgA, or IgE) to the most effective antibody class for a particular location in the body. IgE expression is needed for protection from parasites at barriers to the environment (airways, gut, skin). The cost of this elaborate immune mechanism is frequently the lack of normal tolerance to harmless allergens, causing allergy.

There is compelling clinical and experimental evidence that both IgE class switching and somatic hypermutation in humans occur transiently in the respiratory tract upon allergen stimulation ([ 8 ][8]–[ 10 ][9]). Whether primary contact with peanuts through the skin ([ 3 ][10], [ 4 ][3]) is followed by local class switching to IgE in the aerodigestive tract in food allergy remains to be investigated. Immediate hypersensitivity that is characteristic of allergic reactions mediated by IgE occurs in the gut as it does in the airways (see the figure). The IgE-expressing B cells isolated from blood by Croote et al. may represent peanut-specific cells that have migrated out of the tissue to other sites in the body where they continue to function ([ 10 ][9], [ 11 ][11]).

The authors focused on B cells that were of interest because the variable region sequences in six B cells from two of the six individuals studied were similar. Such similarity between individuals is highly improbable (one in 1014 potential sequences in the far fewer number of B cells that occur in each individual). The similarities suggest that the antigen-binding sequences are convergent or “public” sequences (inherited sequences that are conserved in evolution). Convergent sequences have been observed in infectious disease and in vaccination studies. A rationale is to hand: The relatively small germline gene repertoire encoding the Ig variable region sequences, compared to the repertoire resulting from somatic hypermutation and affinity maturation of the B cells, may have evolved in our ancestors to protect them against commonly encountered pathogens. Whether the conserved sequences serve the same purpose now or allergens are mistaken for the pathogens that affected our ancestors is unclear ([ 12 ][12]).

The six convergent clones were expressed as recombinant antibodies. This revealed high levels of somatic hypermutation, reflecting affinity maturation in the B cells specific for the three most common and clinically relevant peanut allergens, Ara h 1, Ara h 2, and Ara h 3. The coincidence of convergence and peanut specificity here is remarkable. Genetic mutagenesis gave insight into the crucial residues for activity, and this could be further understood through high-resolution crystal structure determination of the allergen-antibody complexes ([ 13 ][13]). One other B cell was shown to express an Ara h 3–specific IgE antibody. This cell was especially interesting because the IgE was related to an IgG4 (an IgG subclass) in the same cell. This confirms previous reports of related IgG4 and IgEs in allergy ([ 10 ][9]). IgG4 is an antibody class that confers tolerance to allergens by competing with IgE for specific antigens ([ 14 ][14], [ 15 ][15]) and is dramatically increased in specific allergen immunotherapy. It is reassuring that the immune system itself can operate a mechanism to prevent or ameliorate allergy, which can be exploited in the clinic.

Further research on these antibodies could lead to modified antibodies or antibody fragments that compete with IgE for allergen binding and prevent the allergic response. Future use of whole-exome sequencing, perhaps comparing the development of IgE-expressing plasma cells with those expressing other antibody classes, may identify genes that regulate IgE plasma cell development and survival that could be counteracted. The work of Croote et al. exemplifies a concerted approach to understanding and potentially intervening in allergic disease.


No comment yet.
Rescooped by Gilbert C FAURE from Immunology and Biotherapies
November 19, 2018 3:47 AM
Scoop.it!

AR101 Oral Immunotherapy for Peanut Allergy | NEJM

AR101 Oral Immunotherapy for Peanut Allergy | NEJM | Allergy (and clinical immunology) | Scoop.it
Original Article from The New England Journal of Medicine — AR101 Oral Immunotherapy for Peanut Allergy...

 

In this phase 3 trial of oral immunotherapy in children and adolescents who were highly allergic to peanut, treatment with AR101 resulted in higher doses of peanut protein that could be ingested without dose-limiting symptoms and in lower symptom severity during peanut exposure at the exit food challenge than placebo. (Funded by Aimmune Therapeutics; PALISADE ClinicalTrials.gov number, NCT02635776.)


Via Krishan Maggon
No comment yet.
Rescooped by Gilbert C FAURE from Immunology and Biotherapies
October 23, 2018 2:56 AM
Scoop.it!

Toxin or treatment? Peanut allergy

Toxin or treatment? Peanut allergy | Allergy (and clinical immunology) | Scoop.it
Ingesting small doses of peanut products guards against allergic reactions—but an undercurrent of anxiety persists.

![Figure][1]

Jacob Kingsley, 12, visits a bakery that was off-limits before he began oral immunotherapy for a peanut allergy.

PHOTO: MADDIE MCGARVEY

Jacob Kingsley was 9 years old when he was handed the poison he'd shunned since before he could walk and told to swallow it as medicine. Obediently, he gulped down a few micrograms of peanut flour—less than 1/1000 of a peanut—diluted in grape Kool-Aid. His mother and a nurse hovered, ready to inject him with epinephrine if an itchy throat and wheezing struck.

Jacob's mother, Jennifer Kingsley, had driven him 2 hours from their home in Columbus to this doctor's office in Cincinnati, Ohio, for the first of dozens of sessions of peanut immunotherapy. Giving Jacob gradually increasing doses of peanuts, she hoped, would desensitize his immune system.

It's a strategy Kingsley hadn't pursued until she reached her breaking point. A year earlier, Jacob had swallowed a handful of popcorn that, unbeknownst to him, was laced with peanut product. He suffered a particularly frightening reaction: two bouts of intense symptoms about 6 hours apart. The incident marked his second peanut-related trip to the emergency room, and Kingsley was terrified that the next encounter could be fatal. “I decided, ‘I can't live like this,’” she says. “I was desperate.”

As Jacob sat through the hourslong appointment in Cincinnati, playing video games and swigging increasing doses of peanut-spiked Kool-Aid, he joined legions of children writing food allergy's next chapter. Today, more than 3000 people worldwide, most of them children, have undergone peanut immunotherapy, with the goal of protecting them if they accidentally encounter the food. Other children are trying immunotherapy for allergies to milk, eggs, and tree nuts. Some, like Jacob, get treatment in allergists' offices, where doctors share protocols informally and in published papers. Other children have enrolled in clinical trials, including those run by two companies racing to introduce a peanut-based capsule or skin patch. Both plan to apply for approval from the Food and Drug Administration (FDA) this year. The agency's blessing would dramatically boost immunotherapy's credibility and reach.

In a field that for decades has had nothing to offer patients beyond avoidance, immunotherapy marks a seismic shift. As it edges closer to mainstream, “There's mixed feelings, with a whole range of enthusiasm,” says Corinne Keet, a pediatric allergist-immunologist at Johns Hopkins Medicine in Baltimore, Maryland. Fear that it might cause harm is mingling with euphoria that children living constrained lives could be set free. Doctors who offer immunotherapy describe families eating in Chinese restaurants for the first time and home-schooled children rejoining their peers.

Like many medical firsts, the therapy is not perfect. “This is version 1.0,” says Brian Vickery, a pediatric allergist-immunologist at Emory University in Atlanta. He has conducted peanut immunotherapy trials and worked for 2 years at Aimmune Therapeutics, headquartered in Brisbane, California, one of the companies whose products are nearing approval. Physicians fret about oral immunotherapy's rigors—treatment must continue indefinitely—and its risks, which include the same allergic reactions it aims to prevent. Last year in Japan, a child suffered brain damage during a trial of immunotherapy for milk allergies.

Meanwhile, physicians on the front lines are navigating hazy science. No one knows exactly how immunotherapy works or who's most likely to be helped or hurt by it. “For me,” Keet says, “it's really not clear for an average child with peanut allergy whether it will make sense to do oral immunotherapy or not.”

LIKE MANY WHO STUDY food allergies, Keet was enticed by their mystery. Animal models are poor. The intensity of allergic reactions varies unpredictably, even in the same person over time. Why one child outgrows an allergy and another doesn't is unknown.

“This was something we didn't cover much in medical school” in the 1990s, says Matthew Greenhawt, a pediatric allergist-immunologist at Children's Hospital Colorado in Denver. Greenhawt's career trajectory tracks with a surge in food allergies, and these days, he can barely keep up with the stream of affected children who visit his hospital. Today, between 1% and 2% of people in the United States, the United Kingdom, and several other countries are allergic to peanuts—a rate that has roughly tripled since the mid-1990s. Other food allergies, such as those to tree nuts, are also on the rise. What's causing the increase is not well understood.

Despite rising caseloads, deaths from food allergies remain rare. Precise numbers are hard to come by, and estimates range from fewer than 10 to more than 150 a year in the United States. But even though an affected child is more likely to be struck by lightning than to die of a food allergy, the risk can feel ever-present. Parents never know when their children will happen upon culprit foods and how they'll be affected if they do. “We live in a complex world—people move food all over the place,” says David Bunning, a businessman whose two sons, now adults, have multiple food allergies. “The impact on children in terms of their confidence to explore their environment can be extreme.” Bunning's family almost never traveled or ate out. At their grandparents' house, the boys were usually confined to one room where food wasn't allowed.

Bunning now chairs the board of directors at Food Allergy Research & Education (FARE), an advocacy group in McLean, Virginia. Families like his, and the doctors who cared for their children, began to agitate for new treatments about a decade ago. Immunotherapy was the obvious candidate: Injections that desensitize the immune system to pollen, grass, pet dander, and bee venom have been around for decades.

Whether for an allergy to cats or pistachios, immunotherapy aims to disrupt the cells that swing out of control when faced with an allergen. When a child who is allergic to a food eats it, food proteins cross from the digestive tract into the bloodstream. An antibody called immunoglobulin E (IgE), which is bound to white blood cells called mast cells in tissues, recognizes the culprits. IgE activates the mast cells, which release histamine and other chemicals. In the skin, that response can lead to hives; in the respiratory tract, wheezing; and in the gut, vomiting. The most serious symptoms, such as a swollen throat or a reaction throughout the body, mark anaphylaxis, which is what families fear the most. Allergy shots blunt production of IgE, in part, researchers believe, by boosting levels of certain T cells that prompt a cascade of immune changes.

![Figure][1]

Fighting fire with fire
Eating gradually increasing doses of a food allergen seems to desensitize the immune system over time. Thousands of children have tried oral immunotherapy, and a capsule to treat peanut allergies might be approved by regulators next year. But there's anxiety about the strategy's risks and unknowns.

GRAPHIC: C. BICKEL/ SCIENCE

Brief testing decades ago indicated that shots for food allergies weren't safe. So around the mid-2000s, scientists began to feed children the allergen instead. One watershed moment came in 2005, when the National Institutes of Health formed a consortium for food allergy clinical trials. A second was in 2011, when advocates sponsored a symposium at Harvard Medical School in Boston to standardize goals and strategy for the pioneering immunotherapy efforts. About 60 people attended. “The patients were very clear,” says Carla McGuire Davis, a pediatric allergist-immunologist at Texas Children's Hospital in Houston. They didn't care about eating a peanut butter sandwich; they wanted protection if they accidentally encountered one. Trialists set their end dose at a couple of peanuts and pressed ahead.

The results of early clinical trials were promising, says Hugh Sampson, a pediatric allergist-immunologist at the Icahn School of Medicine at Mount Sinai in New York City, who has studied immunotherapy in food allergies for many years. After 6 to 12 months of treatment, he says, about 70% to 80% of patients could handle higher doses of the food than before. Lab data were encouraging, too: Ingesting allergens over time seems to make mast cells less reactive, inhibiting their release of harmful chemicals. The therapy also produces other immunoglobulins: IgG4, which further inhibits mast cell activity, and IgA, which helps keep food allergens from escaping the gut (see graphic, p. 280).

The 2011 conference inspired the founding of the company now called Aimmune, fueled by more than $3.5 million from FARE. A second company, DBV Technologies, based in Montrouge, France, and New York City, expanded a few years later. Aimmune began to develop an oral product, essentially a capsule of powder derived from peanut flour with proteins held to consistent levels. In February, the company announced in a press release the results of a phase III trial involving 496 children and teenagers, with a regimen stepping up every 2 weeks through 11 dose levels. Among the 372 people in the treatment group, about 20% dropped out for various reasons, including side effects. After about a year, 96% of people who completed treatment could consume one peanut with no more than mild symptoms, 84% could tolerate two, and 63% could tolerate at least three.

DBV's skin patch represents a more conservative strategy: It delivers tiny amounts of peanut protein, the equivalent of one peanut over 3 years. Last year, DBV announced that in its phase III trial of almost 400 patients, after a year, those using the patch could, on average, eat three peanuts over the course of several hours before experiencing clinical symptoms such as vomiting or hives; before the trial, the average was just under one peanut. Outcomes varied substantially from person to person.

If one or both products are approved by FDA in the coming months, expectations are high that they'll be welcomed: Aimmune is now worth about $1.5 billion on the U.S. stock exchange. In 2016, FARE sold its share in Aimmune for $47 million.

MEANWHILE, SOME DOCTORS embrace another route: offering peanut immunotherapy in their practices. “I can treat 20 patients with $5.95 of peanut flour,” says Richard L. Wasserman, a pediatric allergist-immunologist in Dallas, Texas.

Wasserman ventured into food allergy immunotherapy 11 years ago. He developed a protocol based partly on published case reports and protocols for allergy shots, and he put IVs into his first five peanut allergy patients in case he had only seconds to rescue them from severe anaphylaxis. “When they all sailed through the first day, we stopped doing IVs,” he says. “But that's a measure of how concerned I was.”

Wasserman has since treated more than 300 children with peanut allergies and more than 400 with other food allergies. Other practitioners are joining in, among them the Cincinnati allergist whom the Kingsley family sought out: Justin Greiwe at Bernstein Allergy Group. Greiwe joined the practice in 2014, straight out of medical training. “It was a little nerve-wracking at the beginning,” he says, because no officially sanctioned oral immunotherapy protocol existed. He took precautionary measures, such as lung testing before every treatment, to help ensure patient safety.

Some clinicians—and executives at the companies developing products—aren't happy about the doctor's office treatments. “That gives a lot of us pause,” says Sampson, who in addition to his academic post is chief scientific officer of DBV. “We're very afraid that if this goes on enough, somebody is going to have an accident or a fatal reaction, and that's really going to change the FDA's viewpoint” about the products in development, he says.

Wasserman agrees about the need for caution. “Not every practicing allergist should be doing oral immunotherapy,” he says. Greiwe suggests the treatment requires a dedicated staff, and he gives every immunotherapy family his cellphone number.

Jacob was one of Greiwe's first immunotherapy patients. His mother remembers Jacob's ears burning—a minor reaction that subsided on its own. “Or he said he hated peanuts and wanted to quit,” she says. Worst was about 6 months in, when Kingsley discovered that for 2 weeks, Jacob had hidden his dose to avoid eating it. That was “the only time we ever felt danger,” she says. Stopping treatment can quickly alter the immune system, says Cecilia Berin, an immunologist at Mount Sinai, because immunotherapy requires constant exposure. When Jacob squirreled away his daily dose, the changes induced in his immune system almost certainly started to fade out, putting him at risk. Greiwe restarted him on a lower dose and, his mother says, “We got through it.”

EVEN CHILDREN WHO faithfully follow instructions face risks. The immune system can react to even subtle pressures, and the list of what can provoke a reaction to treatment is long. Exercising within a couple of hours of the dose can do it; so can a cold, a stomach virus, menstruation, or a hot shower. An asthma attack can trigger a reaction—many children with allergies have asthma as well—and so can stress. “We had a patient who had just played the violin on a stage, came down, and about 15 minutes later … took the dose and had a reaction,” Davis says.

Berin posits that external pressures such as physical activity or illness make the gut more permeable, pushing more of the immunotherapy dose into the bloodstream. But that remains hypothesis. Regardless, it's becoming clear that “there are people who react years down the road to a maintenance dose,” Keet says. For Jacob, such a moment came 9 months in. One evening while watching a movie, he downed his peanut M&M's and later ran outside with his cousins to dance in a rainstorm. He broke out in hives head to toe. Kingsley dialed Greiwe's number, and Jacob got a double dose of an allergy medication.

![Figure][1]

Food allergies are becoming more common, and a handful of foods accounts for the vast majority of allergies. But small doses of the foods can blunt allergic reactions.

PHOTO: SCIENCE PHOTO LIBRARY/SCIENCE SOURCE

The most tragic data point to date is the case in Japan. A child had enrolled in a trial of immunotherapy for milk allergies at the Kanagawa Children's Medical Center in Yokohama. He'd raised what he could ingest from less than 8 milliliters to 135 milliliters—about half a glass of milk. After 3 months on that maintenance dose, he swallowed it and soon complained of pain. Within minutes, he had stopped breathing. His heartbeat was later restored in the emergency room, but he'd gone too long without it and sustained severe brain damage, according to a statement from the hospital's president, Sumimasa Yamashita, in November 2017. Kanagawa Children's Medical Center declined to comment, saying only that the incident remains under investigation.

In its statement, the hospital noted the boy had suffered an asthma attack the day before the catastrophic dose. He also was on a protocol that aimed to rapidly escalate the volume of milk he could drink over less than 3 weeks. But why the child reacted so disastrously to that glass of milk is unknown.

“What people don't understand is this level of protection fluctuates,” says Mimi Tang, a pediatric allergist-immunologist at Murdoch Children's Research Institute in Melbourne, Australia. “It is not guaranteed, nor is it constant.”

One of the few long-term analyses was published in 2013 in The Journal of Allergy and Clinical Immunology . Keet, pediatric allergist-immunologist Robert Wood at Johns Hopkins Medicine, and their colleagues sought out 32 children who'd been in a milk immunotherapy trial. Three to 5 years later, “The results were surprising in a sobering kind of way,” Wood says. Only about a quarter “were doing great … tolerating unlimited quantities of milk without side effects.” Another quarter had abandoned the protocol and returned to strict avoidance. The rest were eating dairy products inconsistently, with intermittent or even frequent allergic reactions. “It's hard to know which comes first, whether they got complacent” about ingesting it “or backed off because [they were] having too many symptoms,” Wood says.

MORE AND MORE families are willing to live with those uncertainties because the alternative is greater anxiety. “We were scared senseless,” says Divya Balachandar, whose daughter Leena Wong, now 7 years old, had her first episode of anaphylaxis at age 4 after being touched by a cashew. Testing revealed Leena also was allergic to sesame, eggs, milk, other tree nuts, and peanuts. Balachandar, a pediatric pulmonologist in New York City, and her husband enrolled Leena in a federally funded oral immunotherapy trial for peanut allergy in 2015. “It made me nervous, really nervous, to put something in my daughter's mouth that she was allergic to,” Balachandar says. She gravitated toward a trial over treatment with a local allergist because, she says, “there were no rules” about how to treat in private practice. By this spring, Leena could eat two spoonfuls of peanut butter—about 25 peanuts—without a problem. She started second grade sitting with her classmates at lunchtime, liberated from a separate nut-free table.

Both companies developing peanut-based treatments say they had more volunteers for their trials than they could accommodate. Private practitioners usually have a waiting list; Greiwe's runs more than 4 months. At Stanford University in Palo Alto, California, which has a large food allergy research program, more than 2000 patients are waitlisted to enroll in the university's clinical trials, says Sharon Chinthrajah, an allergist-immunologist there.

More treatments are on the horizon. In Australia, Tang is working with a company that's testing an approach she pioneered, a combination of a probiotic and oral peanut immunotherapy. The probiotic should tilt the body toward producing the subset of T cells that tolerate the allergen and away from making cells that attack it, she says. Chinthrajah and others are enthusiastic about combining oral immunotherapy with a monoclonal antibody called omalizumab, which is FDA approved to treat allergic asthma. Clinical trials are also gearing up to test other monoclonal antibodies that target molecules involved in allergic inflammation.

Jacob's and Leena's families are eager to see what comes next. Jacob is also allergic to pistachios and cashews, but because he finds those foods easier to avoid than peanuts, the family has rejected immunotherapy that targets them. Leena's family is the opposite. With her older sister and her parents, Leena attends Indian functions regularly, where tree nuts are a common ingredient in sauces. In August, another episode of anaphylaxis landed her in the emergency room: She began to vomit and suffered chest tightness and eye swelling after eating Indian food her parents suspect contained cashews—despite having triple-checked with the restaurant that it did not. “I would love to do tree nuts,” Balachandar says, once immunotherapy “becomes more available and better understood.”

Physicians with deep roots in food allergy immunotherapy hope those new to it tread carefully. Doctors who offer such treatments “have to know the data cold,” including published results and side effects that may crop up, Greenhawt says. Still, he's thrilled that peanut immunotherapy treatments may soon be approved. The other day, talking with a peanut-allergic 4-year-old and his mother, Greenhawt shared what the next year might bring. “I said, ‘I'm going to see you a year from now; hopefully, we will have two products that are approved, and we can talk about which one might be best for you.’” The mother looked startled and delighted, Greenhawt says. “I've never seen somebody smile as brightly as that.”

[1]: pending:yes

Via Krishan Maggon
No comment yet.
Scooped by Gilbert C FAURE
October 4, 2018 8:14 AM
Scoop.it!

Scientists develop new test to safely diagnose peanut allergies - News - Medical Research Council

Back to listingNews Scientists develop a new test to safely and accurately diagnose peanut allergies 3 May 2018 MRC scientists have developed a new laboratory test to diagnose peanut allergy. The test has 98% specificity and, unlike current options, it doesn’t run the risk of false-positives or causing allergic reactions such as anaphylactic shock. The simple blood test is five times more cost-efficient compared to the oral food challenge (OFC) – the standard food allergy test – and could be adapted to test for other food allergies. Peanut allergies are among the most common food allergies in children*. Currently, doctors diagnose peanut allergy using a skin-prick test or specific IgE test but this may result in over-diagnosis or false-positives and it cannot differentiate between sensitivity and true food allergy. When skin-prick and IgE test results are unclear, allergists rely on an OFC, which consists of feeding peanut in incrementally larger doses to a patient in a highly-controlled setting in hospital to confirm allergy to the food. While the test is the gold-standard for diagnosing food allergies, there is risk of causing severe allergic reactions. Now, the researchers have developed a safer, accurate blood test in the lab. The new test, called the mast activation test (MAT), could act as a second line tool when skin-prick test results are inconclusive and before referring children and their families to specialists for an OFC, according to researchers from the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma. Their new study was published in the Journal of Allergy and Clinical Immunology. Dr Alexandra Santos, an MRC Clinician Scientist at King’s College London, paediatric allergist and study lead author, said: “The current tests are not ideal. If we relied on them alone, we’d be over diagnosing food allergies – only 22% of school-aged children in the UK with a positive test to peanuts are actually allergic when they’re fed the food in a monitored setting.” Dr Santos continued: “The new test is specific in confirming the diagnosis so when it’s positive, we can be very sure it means allergy. We would reduce by two-thirds the number of expensive, stressful oral food challenges conducted, as well as saving children from experiencing allergic reactions.” Food allergy symptoms are triggered when allergens interact with an antibody called immunoglobulin E (or IgE). The food allergens activate IgE antibodies, triggering symptoms such as skin reactions, itching or constricting of the mouth, throat and airways, and digestive problems (such as stomach cramps, nausea or vomiting). The current skin-prick test and IgE test, which have been in use for decades, measure the presence of IgE antibodies. The new test focuses on mast cells, which play a crucial role in triggering allergic reactions. Mast cells activate by recognising the IgE in plasma and, in allergic patients, produce biomarkers associated with allergic reactions, which can be detected in the lab. Using blood samples from 174 children participating in allergy testing – 73 peanut allergic and 101 peanut-tolerant – the scientists added peanut protein to mast cells to screen for IgE-mediated activation. The MAT accurately identified peanut allergy with 98 specificity. (Specificity is a statistical measure in determining efficacy for diagnosis. The MAT test rarely gives positive results in non-allergic patients.) The researchers also found the test reflected the severity of peanut allergy – patients with more severe reactions have a higher number of activated mast cells. The MAT test is five times cheaper to conduct than the OFC, which requires an allergist and specialist nurses on hand to monitor for adverse reactions and provide medical support if symptoms arise. Dr Santos said: “We are adapting this test to other foods, such as milk, eggs, sesame and tree nuts. This test will be useful as we are seeing more and more children who have never been exposed to these foods because they have severe eczema or have siblings with allergies. Parents are often afraid to feed them a food that is known to cause allergic reactions.” The researchers believe the MAT test may have other uses, for example, in the food industry to detect the presence of allergens in products. Pharmaceutical companies could use it to monitor patients’ allergic response to drugs being evaluated during clinical trials. The scientists plan to transition the biomarker test out of the laboratory and into a clinical setting. They will be testing blood samples from patients with suspected allergies to further validate its utility. 5 to 8% of UK children have a food allergy with up to one in 55 children having a peanut allergy, according to Food Standards Agency estimates. Current UK guidelines recommend avoiding giving your child peanuts and foods containing peanuts before the age of six months. Other countries, such as Canada and the United States, have updated their recommendations – a move that is in the works in the UK. The researchers say updated guidelines may result in a rise in requests for peanut allergy diagnosing. This paper is available on Europe PMC. Categories Categories: Research Health categories: Blood, Inflammatory, Respiratory, Generic Strategic objectives: Lifestyles affecting health, Environment and health, Securing impact from medical research, Aim: Picking research that delivers, Aim: Research to people, Aim: Supporting scientists Locations: London Type: News article
No comment yet.
Rescooped by Gilbert C FAURE from Immunology and Biotherapies
June 15, 2018 4:04 AM
Scoop.it!

Safety and efficacy of epicutaneous immunotherapy for food allergy - Wang - 2018 - Pediatric Allergy and Immunology - Wiley Online Library

Safety and efficacy of epicutaneous immunotherapy for food allergy - Wang - 2018 - Pediatric Allergy and Immunology - Wiley Online Library | Allergy (and clinical immunology) | Scoop.it
Food allergy is increasingly common in children, affecting about 4%‐8%. The mainstays of management remain allergen avoidance and emergency preparedness to treat allergic reactions with emergency medications. Unfortunately, these approaches are unsatisfactory for many patients and their families as the restrictions, constant vigilance, and unpredictable severity of allergic reactions negatively impact quality of life. In recent decades, there has been significant interest in developing treatments for food allergy that lead to desensitization to increase thresholds for triggering allergic reactions and decrease the risk of reacting to allergen‐contaminated food products. Epicutaneous immunotherapy (EPIT) is a novel therapy that is currently under investigation, delivering allergen via repeated applications to the skin and targeting antigen‐presenting cells in the superficial skin layers. Murine models have demonstrated that allergen uptake is an active process by skin dendritic cells with subsequent migration to draining lymph nodes. Allergen exposure to the non‐vascularized epidermis limits systemic absorption, contributing to the high‐safety profile. Results from murine experiments showed that EPIT has comparable efficacy as subcutaneous immunotherapy in terms of challenge outcomes, airway hyper‐responsiveness, and immunologic parameters. Several clinical trials of EPIT have recently been completed or are ongoing. Results support the high safety and tolerability of this approach. Efficacy data suggest that the change in threshold eliciting dose following 1 year of therapy is less than that seen compared to high‐dose (2‐4 g peanut protein) oral immunotherapy, but more prolonged treatment with EPIT appears to lead to increasing desensitization. Additional data from larger‐scale studies should provide a more robust assessment of safety and efficacy of EPIT.

Via Krishan Maggon
No comment yet.
Scooped by Gilbert C FAURE
April 15, 2018 12:39 PM
Scoop.it!

Anti-hIgE gene therapy of peanut-induced anaphylaxis in a humanized murine model of peanut allergy

Anti-hIgE gene therapy of peanut-induced anaphylaxis in a humanized murine model of peanut allergy | Allergy (and clinical immunology) | Scoop.it
Peanuts are the most common food to provoke fatal or near-fatal anaphylactic reactions.
Treatment with an anti-hIgE mAb is efficacious but requires frequent parenteral administration.
No comment yet.
Rescooped by Gilbert C FAURE from Immunology and Biotherapies
February 21, 2018 6:47 AM
Scoop.it!

Aimmune Therapeutics biologic immunotherapy AR101: Pivotal Phase 3 PALISADE Trial Meets Primary Endpoint in Patients With Peanut Allergy

Aimmune Therapeutics biologic immunotherapy AR101: Pivotal Phase 3 PALISADE Trial Meets Primary Endpoint in Patients With Peanut Allergy | Allergy (and clinical immunology) | Scoop.it
BRISBANE, Calif.--(BUSINESS WIRE)--Feb. 20, 2018-- Aimmune Therapeutics, Inc. (Nasdaq:AIMT), a biopharmaceutical company developing treatments for potentially life-threatening food allergies, today announced that its pivotal Phase 3 PALISADE efficacy trial of AR101 met the primary endpoint. In the United States, AR101 has U.S. Food and Drug Administration (FDA) Breakthrough Therapy Designation for peanut-allergic patients ages 4–17.AR101 has U.S. Food and Drug Administration (FDA) Breakthrough Therapy Designation for peanut-allergic patients ages 4–17.

PALISADE (Peanut ALlergy Oral Immunotherapy Study of AR101 for DEsensitization in Children and Adults) is our core, pivotal Phase 3 clinical trial for AR101. PALISADE was the largest randomized clinical trial for peanut allergy to date, enrolling more than 550 participants ages 4-55 in the U.S., Canada, and Europe.

Landmark 554-patient Phase 3 study met the primary efficacy endpoint, as 67.2% of AR101 patients ages 4–17 tolerated at least a 600-mg dose of peanut protein in the exit food challenge, compared to 4.0% of placebo patients (p<0.00001) The lower-bound of the 95% confidence interval (CI) of the difference between treatment arms at the primary endpoint was 53.0%, greatly exceeding the pre-specified threshold of 15% (p<0.00001) 50.3% of AR101 patients ages 4–17 tolerated a 1000-mg dose of peanut protein in the exit food challenge, compared to 2.4% of placebo patients (p<0.00001) Among patients ages 4–17 who completed treatment with AR101, 96.3% tolerated a 300-mg dose of peanut protein in the exit food challenge, 84.5% tolerated a 600-mg dose, and 63.2% tolerated a 1000-mg dose 79.6% of AR101 patients ages 4–17 completed the trial; of the 20.4% who discontinued treatment, 12.4% withdrew due to treatment-related adverse events 2.4% of AR101 patients ages 4–17 and 0.8% of placebo patients experienced serious adverse events

Via Krishan Maggon
Krishan Maggon 's curator insight, February 21, 2018 3:40 AM
AR101 has U.S. Food and Drug Administration (FDA) Breakthrough Therapy Designation for peanut-allergic patients ages 4–17.

Gradually increasing doses of AR101 would desensitize patients to peanut over a period of about six months. Afterward, patients would continue to take maintenance doses of AR101 in order to maintain desensitization.
Scooped by Gilbert C FAURE
October 28, 2017 5:20 AM
Scoop.it!

Detection of the Peanut Allergens Ara h 2 and Ara h 6 in Human Breast Milk: Development of 2 Sensitive and Specific Sandwich ELISA Assays

Detection of the Peanut Allergens Ara h 2 and Ara h 6 in Human Breast Milk: Development of 2 Sensitive and Specific Sandwich ELISA Assays | Allergy (and clinical immunology) | Scoop.it
<b><i>Background:</i></b> Little is known about breast milk as a vehicle for tolerance development or sensitization to peanuts very early in life. Thus, well-characterized and
No comment yet.
Suggested by Société Francaise d'Immunologie
September 18, 2017 9:37 AM
Scoop.it!

Peanut allergy could be cured with probiotics

Peanut allergy could be cured with probiotics | Allergy (and clinical immunology) | Scoop.it
The vast majority of children in a large clinical trial continued to tolerate peanuts for 4 years after it had ended, new research shows.
No comment yet.
Suggested by LIGHTING
February 24, 2019 3:43 PM
Scoop.it!

Effect of Epicutaneous Immunotherapy vs Placebo on Reaction to Peanut Protein Ingestion Among Children With Peanut Allergy: The PEPITES Randomized Clinical Trial. | Allergy and Clinical Immunology ...

This randomized clinical trial compares the efficacy and safety of epicutaneous immunotherapy with a peanut patch vs placebo among peanut-allergic children.
No comment yet.
Scooped by Gilbert C FAURE
February 14, 2019 5:20 AM
Scoop.it!

DBV Rejoins Peanut Allergy Treatment Race, But Aimmune Takes the Lead

DBV Rejoins Peanut Allergy Treatment Race, But Aimmune Takes the Lead | Allergy (and clinical immunology) | Scoop.it
DBV Technologies is jumping back into the race to bring an FDA-approved peanut allergy treatment to the market, but it might not be enough to beat its...
No comment yet.
Scooped by Gilbert C FAURE
January 27, 2019 5:39 AM
Scoop.it!

Effect of Avoidance on Peanut Allergy after Early Peanut Consumption | NEJM

Effect of Avoidance on Peanut Allergy after Early Peanut Consumption | NEJM | Allergy (and clinical immunology) | Scoop.it
Original Article from The New England Journal of Medicine — Effect of Avoidance on Peanut Allergy after Early Peanut Consumption...
No comment yet.
Scooped by Gilbert C FAURE
December 31, 2018 8:08 AM
Scoop.it!

Peanut allergy

Peanut allergy | Allergy (and clinical immunology) | Scoop.it

CME / ABIM MOC

Peanut Allergy: Pathophysiologic Mechanisms and Therapeutic Options
  • Authors: Carla M. Davis, MD; J. Andrew Bird, MD; Jacqueline Pongracic, MD
  • CME / ABIM MOC Released: 12/27/2018
  • Valid for credit through: 12/27/2019
No comment yet.
Scooped by Gilbert C FAURE
December 4, 2018 4:09 AM
Scoop.it!

Recent developments and highlights in mechanisms of allergic diseases: Microbiome - Lunjani - - Allergy - Wiley Online Library

Abstract All body surfaces are exposed to a wide variety of microbes, which significantly influence immune reactivity within the host. This review provides an update on some of the critical novel findings that have been published on the influence of the microbiome on atopic dermatitis, food allergy and asthma. Microbial dysbiosis has consistently been observed in the skin, gut and lungs of patients with atopic dermatitis, food allergy and asthma, respectively, and the role of specific microbes in allergic disorders is being intensively investigated. However, many of these discoveries have yet to be translated into routine clinical practice. Abbreviations AAI allergic airway inflammation AD atopic dermatitis AHR airway hyper‐responsiveness AMP antimicrobial peptides COPD chronic obstructive pulmonary disease CRS chronic rhinosinusitis GCS glucocorticoids HDM house dust mite HMOs human milk oligosaccharides ICSs inhaled corticosteroids LABAs long‐acting β2 adrenergic receptor agonists OIT oral immunotherapy PARs protease‐activated receptors PSMs phenol‐soluble modulins RSV respiratory syncytial virus SCFAs short‐chain fatty acids TLR Toll‐like receptor 1 INTRODUCTION An enormous variety of microbes colonize the skin and mucosal body surfaces. These microbes are organized within complex community structures, utilizing nutrients from other microbes, host secretions and the diet. The microbiome is defined as the sum of these microbes, their genomic elements and interactions in a given ecological niche. In addition to bacteria, viruses are also considered to be an important component of the microbiome (virome). The composition of the microbiome is dependent on the specific body site examined, resulting in a series of unique habitats within and between individuals that can change substantially over time.1 This presents significant challenges to the local immune system, which should tolerate the presence of these microbes to avoid damaging host tissue while retaining the ability to respond appropriately to pathogens. The mechanisms that mediate host‐microbe communication are highly sophisticated and need to be constantly coordinated.2 Indeed, disrupted communication between the microbiome and the host due to altered microbiome composition and/or metabolism is thought to negatively influence immune homeostatic networks and may play a role in immune hypersensitivity to environmental exposures, such as allergens.3-5 For several years, epidemiological studies have suggested associations between the migration from traditional farming to urban environments, increase in processed food intake, lack of contact with animals and excessive hygiene practices with the increased incidence of asthma, atopic dermatitis and food allergy. However, it is only relatively recently that the importance of the gut, lung and skin microbiomes in regulation of immune tolerance and its aberrations in a variety of human diseases including allergy and asthma has been recognized.6, 7 In particular, early‐life events such as mode of delivery, breastfeeding, mother's diet and health status, antibiotics and other drug usage in pregnancy and early childhood, early‐life environment (ie, siblings, pets at home, proximity to farm animals and green areas) significantly influence the timing of bacterial colonization and establishment, which modify the risk of developing allergies and asthma, as summarized in Figure 1.8-17 In this review, we will highlight some of the recent advances in our knowledge regarding the influence of the microbiome on immune reactivity in the skin, gut and lungs of patients with atopic dermatitis, food allergy and asthma. In addition, we will discuss the potential translation and challenges associated with microbial‐based therapies in patients with these allergic disorders. 2 MICROBIOME IN ATOPIC DERMATITIS The skin microbiome is comprised of bacteria, fungi, viruses and archaeal communities, with bacteria being the most widely studied.18 The skin microbiome is influenced by age, gender, ethnicity, climate, UV exposure and lifestyle factors.19 16S ribosomal RNA (rRNA) sequencing has demonstrated that significantly diverse bacterial phyla exist on healthy skin with site‐specific differences in composition. This is primarily driven by the physiology of a skin niche. Propionibacterium species are predominantly found in sebaceous sites, with Corynebacterium and Staphylococcus species occurring in moist microenvironments. Malassezia represents the predominant fungal flora on human skin.20 Figure 2 illustrates the interactions between the skin microbiome and host cells. Atopic dermatitis (AD) is characterized by epidermal barrier dysfunction resulting from a synergistic decrease in epidermal barrier structural proteins, alteration in lipid composition and skin pH, activation of local and systemic inflammatory responses and decrease in skin microbiome diversity.19 Staphylococcus aureus overgrowth is consistently linked with AD pathogenesis and correlates with disease severity and eczematous flares.1, 21 High IL‐4 and IL‐13 levels within AD skin can deplete keratinocyte‐produced antimicrobial peptides (AMPs), cathelicidin LL‐37, human beta defensin hBD‐2 and hBD‐3, necessary for controlling pathogenic organisms.22 Defective TLR‐2 expression in Langerhans cells of AD skin has also been observed, which may contribute to the impairment in effective immune recognition and clearance of pathogenic bacteria such as S. aureus.23 Epidermal lipid composition strongly correlates with bacterial diversity and composition at typical sites for AD lesions. For example, S. aureus dominance was associated with elevated levels of ceramide AS.21 Staphylococcus aureus overgrowth with concomitant decline in Staphylococcus epidermidis is a general feature of AD and is not restricted to eczematous lesions.19, 21 Staphylococcus aureus colonization is evident in 90% of AD cases,24 associates with AD severity and increased allergen sensitization.25 Intervention studies with antimicrobials targeting S. aureus can reduce AD severity. Restoration of the epithelial barrier with anti‐inflammatory and emollient use is able to increase microbial diversity of lesional skin.1, 24 Patients with severe AD can be colonized with a single S. aureus strain, which persists even post‐eczematous flare albeit at a lower relative abundance. In contrast, S. epidermidis strains were more heterogeneous. Interestingly, patients with more severe AD were colonized with methicillin‐sensitive staphylococci, whereas less severe AD was more frequently associated with methicillin‐resistant strains. This observation may have significant treatment implications, particularly when methicillin‐sensitive S. aureus and methicillin‐resistant S. epidermidis strains are present.26 In a recent study, the skin microbiome of infants with AD showed a consistent absence of S. aureus sequences at multiple time points on lesional skin contrary to reported finding in patients with established AD. The most prevalent species were S. epidermidis and S. cohnii. However, those who developed AD at 12 months had significantly lower levels of these commensal staphylococci detectable at 2 months of age.27 This study suggests that S. aureus colonization may not always predate clinical AD and highlights the need for longitudinal studies to investigate the transition to microbial dysbiosis in AD. Commensal S. epidermidis strains can also increase during disease flares.24 Coagulase‐negative staphylococci (CoNS), which include S. epidermidis, S. hominis and S. lugdunensis, can secrete antimicrobials that limit S. aureus overgrowth and biofilm formation.1, 28 In addition, S. epidermidis activates TLR2, thereby promoting tight junction protein expression and inducing keratinocyte‐derived antimicrobial peptide secretion. Early occupation of the neonatal human skin by S. epidermidis is associated with induction of S. epidermidis‐specific FOXP3+ Treg cells that regulate local activation of host immune responses.29 Other members of the healthy skin microbiota, such as Propionibacterium, Streptococcus, Acinetobacter, Corynebacterium, Prevotella and Proteobacteria, are frequently reduced in AD patients.28, 30 Staphylococcus aureus can contribute to epidermal barrier disruption in a number of ways. Staphylococcus aureus downregulates terminal differentiation proteins such as filaggrin and loricrin, while secretion of proteases contributes to the disruption of the epidermal integrity via direct proteolytic activity or activation of protease‐activated receptors (PARs). Superantigens such as staphylococcal enterotoxins A and B or toxic shock syndrome toxin‐1 trigger a cytokine response that further disrupts the epidermal barrier. These enterotoxins also act as allergens, and toxin‐specific IgE contributes to cutaneous inflammation.28, 31 Staphylococcus aureus expresses exotoxins such as cytolytic α‐toxin, which damage keratinocytes, while β‐, γ‐ and δ‐toxins stimulate mast cell degranulation.28, 32 Phenol‐soluble modulins (PSMs) induce keratinocyte damage and secretion of the alarmins IL‐1α and IL‐36α, which further exaggerate skin inflammation.33 An impaired skin barrier results in increased exposure of the immune system to microbial components, resulting in a progressive cycle of inflammatory responses and tissue damage. It was recently suggested that reactivity to S. aureus can be facilitated via allergen co‐exposure and vice versa since patients with sensitization to house dust mite also show significantly more IgE reactivity to S. aureus and Escherichia coli, two abundant species in the house dust mite microbiome.34 A subset of AD patients is susceptible to eczema herpeticum (EH), and S. aureus may contribute to EH susceptibility as it has been shown to secrete products that enhance viral replication.1 Despite Malassezia species having a commensal role in healthy skin, in AD Malassezia may contribute to disease pathogenesis. Malassezia DNA has been detected in 90% of AD skin lesions, and colonization increases with disease severity.35 In addition, different Malassezia strains were found in AD and healthy individuals suggesting the existence of key pathogenic strains in AD.36 Higher levels of IgE sensitization to Malassezia have been detected in adult AD compared to healthy individuals and childhood AD.22, 36 Malassezia could contribute to AD pathogenesis by secreting immunogenic proteins that induce proinflammatory cytokines, expression of TLR2 and TLR4 on keratinocytes and induction of auto‐reactive T cells.22 Atopic dermatitis is considered a first step in the atopic diathesis, facilitated in part by the defective epidermal barrier of AD. The IL‐4/IL‐13 axis in AD is also thought to upregulate the pore‐forming claudin‐2 expression in the gut leading to barrier defects.19 In addition to the skin microbiota, AD has been associated with changes in the gut microbiota. Patients with AD have lower levels of Bifidobacterium in the gut compared to healthy controls, and Bifidobacterium levels were inversely correlated with AD disease severity.37 Several studies have shown that alterations in gut microbiota composition can precede the development of AD. Early gut colonization with C. difficile was associated with AD development,38 and low gut microbiota diversity and specifically low Bacteroidetes diversity at 1 month were associated with AD development at 2 years of age.35, 39 A recent whole‐metagenome analysis demonstrated a lower abundance of key metabolic pathways in AD children associated with depletion of mucin‐degrading bacteria such as Akkermansia muciniphila, Ruminococcus gnavus and Lachnospiraceae.40 These bacteria not only are able to influence immune development through directly influencing signalling pathways and antigen processing but also can lead to a reduced microbial diversity as these bacteria are able to degrade complex polysaccharides into short‐chain fatty acids (SCFAs)—nutrient sources that allow for gut colonization by other microbes.40 Dog exposure at birth was associated with a dose‐related reduced risk of AD in early life, suggesting that exposure to an environment rich in microbial components may be protective.41 In contrast, antibiotic exposure during the first 2 years of life is associated with an increased risk of AD.42 Infants with high faecal calprotectin levels (an antimicrobial protein used as a biomarker of intestinal inflammation) measured at 2 months of age had an increased risk of AD and asthma by 6 years of age. High faecal calprotectin was also shown to be inversely correlated with levels of E. coli. Reduced early colonization with E. coli was shown to impair IL‐10 regulation.43 3 MICROBIOME IN FOOD ALLERGY The human gut microbiome is increasingly being considered as a crucial factor in the development of food allergy, with a strong interrelation between the human gut microbiota, environmental factors, human genetics and gastrointestinal atopy.4, 44 In particular, the composition and metabolic activity of the gut microbiota are intimately linked with the development of oral tolerance.45, 46 Therefore, disturbed microbial homeostasis, especially early in life, appears to significantly influence allergic disease susceptibility. Figure 3 illustrates some of the known interactions between the gut microbiome and host mucosal cells. Recently, the oral bacterial composition in saliva samples from healthy and allergic children up to 7 years of age was described. The result confirmed that early changes in oral microbial composition seem to associate with immune maturation and allergy development.47 Milk‐allergic infants have higher total bacteria and anaerobic bacterial counts compared with healthy control children after 6 months of differential formula intake. In addition, higher proportions of Lactobacilli and lower proportions of Enterobacteria and Bifidobacteria were observed in 46 milk‐allergic infants.48 The spontaneous resolution of milk allergy in infants was associated with a specific gut microbiota composition.49 Bunyavanich et al showed that Clostridia and Firmicutes were enriched in the infant gut microbiome of subjects whose milk allergy spontaneously resolved. This result suggested that early infant gut microbiota may shape food allergy outcomes in childhood and bacterial taxa within Clostridia and Firmicutes species could be further investigated as probiotic candidates for milk allergy therapy.49 An additional study examining the gut microbiome of 141 children with egg allergy and healthy controls found that genera from Lachnospiraceae and Ruminococcaceae were associated with egg sensitization; however, there was no association between early‐life gut microbiota and egg allergy resolution by age 8 years.50 A prospective microbiome association study in 14 children with food allergy and 87 children with food sensitization showed that the genera Haemophilus, Dialister, Dorea and Clostridium were underrepresented among subjects with food sensitization, whereas the genera Citrobacter, Oscillospira, Lactococcus and Dorea were underrepresented among subjects with food allergy.51 An additional prospective study identified both temporal variation and long‐term variation in the differential abundance of specific bacterial genera in children developing IgE‐associated allergic disease, with Faecalibacterium correlating with IL‐10 and Foxp3 mRNA levels.52 Human milk oligosaccharides (HMOs) have been shown to be important in supporting the establishment of the infant gut microbiome as they are selective substrates for protective microbes such as Bifidobacteria.53 Two recent studies have described differences in HMO composition that are associated with cow's milk allergy or food sensitization.54, 55 One potential mechanism for this association is that different HMO profiles may support the establishment of different microbes early in life, thereby indirectly influencing immune maturation and education. In conclusion, a number of human studies now suggest that food allergy could be associated with changes in microbial exposures in early life, which modifies the development of host immunity and results in pathologic immune responses to food allergens. 4 MICROBIOME IN ASTHMA Composition of the microbiome at all mucosal sites changes dynamically in the first days, months and years of life. If the process of “healthy” and timely colonization is disrupted, the early‐life dysbiosis of the gut and lung becomes an important risk factor for atopy, allergy and asthma. In the Canadian Healthy Infant Longitudinal Development (CHILD) study, the lower relative abundance of the bacterial genera Lachnospira, Veillonella, Faecalibacterium and Rothia in the gut was associated with the development of asthma later in life and mechanistically linked with the reduced levels of faecal SCFAs.56 Another recent study also showed that high levels of SCFAs early in life were protective against later life sensitization and asthma.57 In a US birth cohort, lower relative abundance of Bifidobacterium, Akkermansia and Faecalibacterium, with higher relative abundance of Candida and Rhodotorula, in the gut of neonates significantly increased the risk of developing multisensitized atopy and asthma later in life.58 Interestingly, the faecal metabolome of those children at increased risk contained increased levels of pro‐inflammatory metabolites, among which 12, 13‐DiHOME was able to induce IL‐4 production in CD4+ T cells and decreased the abundance of Tregs.58 Increased abundance of nasopharyngeal Lactobacillus species during acute respiratory infection with respiratory syncytial virus (RSV) in infancy was associated with reduced risk of wheezing at 2 years of age.59 Colonization of the airways with Streptococcus, Moraxella or Haemophilus within the first 2 months of life was associated with virus‐induced acute respiratory infections in the first 60 weeks of life as well as increased risk of asthma later in life.60 Colonization of the hypopharynx within the first month of life with Moraxella catarrhalis, Haemophilus influenzae or Streptococcus pneumoniae was associated with low‐grade systemic inflammation as assessed by serum CRP, TNF‐alpha and IL‐6 levels.61 In addition, a positive association was observed between RSV infection and hospitalization in children with nasopharyngeal colonization with H. influenzae and Streptococcus.62, 63 Importantly, the relative nasopharyngeal abundance of Streptococcus and Staphylococcus negatively correlated with FEV1 and PC20 in children.64 Children who were breastfed and those who had low rates of respiratory infections in the first 2 years of life were colonized early within the upper respiratory tract with Staphylococcus species, followed by Corynebacterium, Dolosigranulum and Moraxella.65-67 However, the most impressive data regarding asthma protection have been observed in relation to traditional farming environments, associated with a high endotoxin and bacterial‐containing dust within the home.5, 15, 17, 68-71 Adult asthma patients treated with inhaled corticosteroids (ICSs) have greater upper and lower airway microbiota diversity compared to control subjects, especially enriched in the phylum Proteobacteria, which include Haemophilus, Comamonadaceae, Sphingomonadaceae, Nitrosomonadaceae, Oxalobacteraceae and Pseudomonadaceae families.72-76 The phylum Proteobacteria is also associated with worse asthma control, whereas Actinobacteria correlates with improvement or no change in asthma control.77 Interestingly, neutrophilic exacerbations of asthma and chronic obstructive pulmonary disease (COPD) correlated with the presence of Proteobacteria in the sputum, whereas eosinophilic exacerbations correlated with the presence of Bacteroidetes.78 Mycoplasma pneumoniae and Chlamydophila pneumoniae are also often found in the airways of the severe asthmatic.79 Macrolide antibiotic treatment may be useful in this subgroup of patients, but patients should be carefully selected.80 Both clarithromycin and azithromycin have been shown to reduce airway hyper‐responsiveness and decrease the abundance of Pseudomonas, Haemophilus and Staphylococcus,73, 81 while increasing the relative abundance of Streptococci.82 However, it is currently not clear how significant a role asthma medications play in directly influencing the composition of the airway microbiota. It has been reported that combination of ICS and oral glucocorticoids (GCS) correlates positively with the increased abundance of Proteobacteria, specifically Pseudomonas, and with a decreased abundance of Bacteroidetes, Fusobacteria and Prevotella.83 In corticosteroid‐resistant patients, Neisseria‐Haemophilus, Campylobacter and Leptotrichia species are present in the lower airways.75 Interestingly, treatment of COPD patients with ICS and long‐acting β2 adrenergic receptor agonists (LABAs), compared to LABA alone, significantly increased the bacterial load, increased bacterial diversity and changed composition of the microbiome in the airways.84 However, prospective longitudinal studies involving corticosteroid‐naïve asthma patients are still needed to address the issue of medication effects on the airway microbiome. The mechanisms responsible for changes in the airway microbiome are also not well understood, and in addition to medications, it is possible that the type of inflammatory response (ie, eosinophil vs neutrophil), changes in host secretions (eg, lipids85, 86) and cellular metabolism might influence microbial colonization and growth within the airways. Figure 4 illustrates the immune responses in the airways that can be influenced by the respiratory microbiome. In addition to asthma, the potential for microbes to play a role in the initial aetiology of rhinitis, or in exacerbations and progression to more severe inflammatory sequelae (such as asthma) is currently being examined. The phylum Proteobacteria is enriched in children with rhinitis, which may be clinically important given the Proteobacteria‐related asthma associations described above.69 Dysbiosis of the inferior turbinate mucosa microbiota, particularly an increase in S. aureus and a decrease in P. acnes, was associated with high total IgE levels in adults with allergic rhinitis.87 In adults with chronic rhinosinusitis (CRS), the genus Corynebacterium was depleted, accompanied by increased relative abundance of genera from the phyla Firmicutes (including Staphylococcus and Streptococcus), Proteobacteria (including Haemophilus, Pseudomonas and Moraxella) or Fusobacteria. This trend was particularly evident in subjects with comorbidities such as asthma and cystic fibrosis.88 Similarly, another study reported that middle meatus samples from CRS patients without nasal polyps were enriched in Streptococcus, Haemophilus and Fusobacterium but exhibited loss of diversity compared to healthy, CRS with nasal polyps and allergic rhinitis subject samples.89 5 LEARNING FROM ANIMAL MODELS Despite the compelling observations and associations in humans that link changes in the microbiota with allergic diseases, very often the causal relationship is not clear. Microbial dysbiosis can be the reason for the disease but can also be the consequence of inappropriate immune reactivity. Animal models have been used to better understand the role of microbes in directly influencing allergic diseases and to elucidate the molecular mechanisms underpinning host‐microbe crosstalk. 5.1 Atopic dermatitis Similar to humans, dogs naturally develop AD and associated allergen sensitization. Canine AD is associated with reduced bacterial diversity, with increased abundance of Staphylococcus pseudintermedius and Corynebacterium species.90 Canine AD lesions improve with antimicrobial treatment and a reduction in Staphylococcus species coincided with restoration of bacterial diversity.30 Filaggrin‐deficient flaky tail mice carry a loss‐of‐function filaggrin mutation, which is associated with a defective epidermal barrier, epidermal hydration and flexibility. Staphylococcus aureus abundance on the skin of these mice correlates with Th2 cytokine levels.91 Inbred DS‐Ng mice develop spontaneous dermatitis, and the skin lesions have been shown to be heavily colonized by S. aureus.29 Staphylococcus aureus triggered cutaneous inflammation involve the accessory gene regulatory (Agr) virulence systems of S. aureus and induced δ‐toxin molecules, which initiate Th2 type skin inflammation. Targeted S. aureus and Corynebacterium bovis antimicrobial therapy improved eczematous lesions and increased bacterial diversity in Adam 17 (a transmembrane metalloproteinase)‐deficient mice. Withdrawal of targeted antimicrobials resulted in a recurrence of eczema and microbial dysbiosis.30 In a mouse itch model, IL‐17A and IL‐22 drive neutrophils to limit the overgrowth of S. aureus on injured skin.25 C5aR‐deficient mice develop reduced microbial diversity, suggesting that the complement system may also regulate the skin microbiota.29 A mouse model of AD showed that application of a Vitreoscilla filiformis bacterial lysate reduced the inflammatory manifestations following allergen application.24 Studies in mice during the neonatal period suggest that tolerance to skin commensals such as S. epidermidis is preferentially established early in life. This supports the hypothesis that exposure to certain microbes at a critical window early in life is required for normal development of the immune system.30 5.2 Food allergy The potential role of the gut microbiome in food allergy has been studied in multiple murine models. Rodriguez et al92 demonstrated that intestinal colonization with Staphylococcus protects against oral sensitization and allergic responses. The microbiota of allergen‐sensitized IL‐4raF709 mice differentially promoted OVA‐specific IgE responses and anaphylaxis when reconstituted in wild‐type germ‐free mice, which could play a role in food allergy.93 The disease‐susceptible IL‐4raF709 mice display enhanced signalling through the interleukin‐4 receptor (IL‐4R) and exhibit STAT6‐dependent impaired generation and function of mucosal allergen‐specific Treg cells, which failed to suppress mast cell activation and expansion.94 Interestingly, STAT6 gene variants are also implicated in the pathophysiology of food allergy in humans.95 The gut microbiota can also regulate Th2 responses through the induction of RORγt Treg cells and Th17 cells.96 Certain bacterial strains such as Bifidobacterium longum 35624, Lactobacillus rhamnosus JB‐1, Clostridia species and Bacteroides fragilis can induce intestinal Treg cells that are able to suppress food allergy and colitis.97, 98 Pattern‐recognition receptor activation on DCs is a potential mechanism by which intestinal microbes may promote Treg cell differentiation.99 5.3 Asthma Important insights regarding the role of the microbiota in the pathogenesis of airway inflammation have come from mouse models. Neonatal mice are more susceptible to develop house dust mite (HDM)‐induced allergic airway inflammation (AAI) and airway hyper‐responsiveness (AHR) than mature mice.100 This phenomenon was associated with a shift from Gammaproteobacteria and Firmicutes towards a Bacteroidetes‐dominated microbiota and the development of PDL‐1–dependent Helios‐ Treg cells.100 Mice housed under germ‐free conditions display significantly more pronounced type 2 inflammation and AHR as compared to conventionally colonized mice. Recolonization, especially early in life, can reverse many of these immunological defects.101 Similarly, antibiotic‐driven dysbiosis in neonatal mice leads to impaired maturation of Tregs and enhanced Th2 responses and promotes proinflammatory colonic iNKT cells.80, 102-105 Conversely, specific bacterial strains, their components or metabolites can successfully induce a variety of anti‐inflammatory responses in the gut and in the lung. L. rhamnosus decreased AAI and AHR induced by Bet v 1 in mice.106 Bacterial strains isolated from neonatal mouse lungs and then administered intranasally very early in life (starting at day 2 after birth) can protect or worsen HDM‐induced airway inflammation, depending which cytokine profile they induced in vitro on precision‐cut lung slices.107 Intramuscular treatment with a DNA plasmid encoding a M. leprae 65 kDa heat‐shock protein (DNA‐HSP65) or subcutaneous injections with proteins from M. tuberculosis delivered in the presence of the TLR9 agonist CpG were able to significantly inhibit development of Der p 1‐induced AAI and AHR in MyD88‐ or Fas‐dependent manner.108 In addition, an exopolysaccharide from B. longum subsp. longum 35624 was shown to protect against colitis and AAI in murine models, which was dependent on TLR2‐induced IL‐10 secretion.109, 110 SCFAs or dietary fibres that are metabolized to SCFAs potently reduced experimental asthma, as well as increased the levels of colonic Bacteroidetes and Actinobacteria species, while decreasing the levels of Firmicutes and Proteobacteria.111, 112 Importantly, the beneficial effects of SCFAs or a high‐fibre diet were transferred to the offspring after treatment of pregnant mice via epigenetic mechanisms.112, 113 Mechanistically, SCFAs have been repeatedly shown to increase Treg numbers and effectiveness.114, 115 In addition, SCFAs influence bone marrow haematopoiesis,111 reduce effector T‐cell activity,116 improve epithelial barrier117, 118 and inhibit mast cell and ILC2 activation.119, 120 Other bacterial metabolites, such as histamine, can induce a wide and complex spectrum of regulatory mechanisms.121, 122 Increased numbers of histamine‐secreting bacteria were observed in adult patients with asthma and correlated with asthma severity.123 Histamine signalling through the H2R is involved in AAI,124 while the use of H2R antagonists in children during their first 6 months of life is associated with significantly increased risk of allergic diseases and asthma.10 6 THERAPEUTIC TARGETING OF THE MICROBIOME Despite the growing number of studies that associate changes in the microbiota with allergic and immune‐related outcomes, only a relatively small number of studies have shown clinical benefits and there are no microbe‐based therapies that are currently universally accepted for the prevention or treatment of allergies or asthma. A number of reasons can be suggested for this, which may include the poor choice of therapeutic microbes to begin with. It is likely that many confounding factors do influence the success of a microbiome therapeutic, such as diet, age, obesity, ethnicity and other environmental exposures. These need to be taken into account and controlled for. In addition, given the explosion in knowledge regarding disease endotypes, it is possible that specific microbes will need to be carefully selected to mechanistically fit with specific disease endotypes and it is likely that one intervention will not work for everyone. Certain interventions such as faecal transplantation may be too crude an approach, and until critical safety concerns are resolved, this type of intervention should not be considered outside the setting of carefully monitored clinical trials. 6.1 Atopic dermatitis Early intervention aimed at protecting the skin barrier may ameliorate progression of the atopic march in a subset of patients.19 Skin microbiome manipulation may offer novel therapeutic opportunities, as has been seen with the emollients supplemented with a Vitreoscilla filiformis lysate.125 Similarly, topically administration of Roseomonas mucosa improved clinical severity scores in adults and children with AD.125 Autologous microbiome transplant (AMT) of S. hominis and S. epidermidis showed efficacy in controlling S. aureus overgrowth.126 In addition to topical bacterial treatments, oral administration of probiotics has also been examined. Prenatal and post‐natal treatment with Lactobacillus and Bifidobacterium strains can reduce risk of AD development in infants,35, 127, 128 which may associate with changes in T cell–mediated responses.129 A mixture of probiotic strains was recently shown to reduce SCORAD index and topical steroid use in children with AD.130 Little has been reported on probiotic treatment of adults with AD, but administration of B. longum 35624 to adults with psoriasis resulted in reduced circulating CRP, TNF and IL‐17 levels, possibly due to increased numbers of Tregs, which suggests that bacteria in the gut can influence skin inflammatory activity in adults.131, 132 Taken together, supplementation with specific probiotic strains may modulate the gut bacteria in a way that influences inflammation within the skin and may protect some children against AD development.35 6.2 Food allergy The use of probiotics in food allergy treatment and prevention has been examined. Supplementation of cow's milk‐allergic children with Lactobacillus casei and Bifidobacterium lactis did not accelerate cow's milk allergy resolution.133 However, the combination of L. rhamnosus GG and extensively hydrolysed casein formula did accelerate milk allergy resolution after 6 and 12 months when compared to the formula‐only control group.134 The combination of L. rhamnosus supplementation and peanut oral immunotherapy (OIT) was evaluated in peanut‐allergic children for 18 months. The combination was effective in inducing possible sustained unresponsiveness and immune changes that suggested modulation of the peanut‐specific immune response.135 In addition, a sustained beneficial effect on psychosocial impact of food allergy at 3 and 12 months after end of treatment was recently reported.136 However, the major limitation of this study is that further work is required to determine the relative contributions of the probiotic vs OIT due to the lack of an OIT and L. rhamnosus supplementation control groups in this trial. 6.3 Asthma A significant number of studies have examined the effect of probiotic supplementation on asthma‐related outcomes. A recent systematic review of probiotic studies in children with asthma identified eleven studies eligible with a total of 910 children. The proportion of children with fewer episodes of asthma was significantly higher in the probiotic group than in the control group, but no statistical significance was observed in childhood asthma control test, asthmatic symptom in the day and night, the number of symptom‐free days, forced expiratory volume in the first second predicted and peak expiratory flow.137 In the future, it will be interesting to evaluate microbial administration directly to the airways, in addition to the gut.138 7 CONCLUSIONS Significant advances have been made in recent years in describing the composition of the microbiome in the gut, airways and skin. The changes in bacterial communities that associate with, or sometimes precede, atopic dermatitis, food allergy and asthma are being identified (summarized in Table 1). Accumulating evidence suggests that microbial exposures might be most effective at preventing atopic disorders during the first 1‐2 years of life. However, substantial gaps in our knowledge on the microbiome still exist. In particular, the field has been slow to translate potentially effective microbiome‐associated therapies into the clinic via appropriate clinical trials performed to high standards and showing meaningful clinical responses that are superior to current avoidance approaches. While the critical role of the microbiota in cancer immunotherapy has been established, there are currently no published data on the potential role of the microbiota in influencing the success of immunotherapy or biologics in allergy or asthma.139 In addition, novel probiotics and not just the traditional probiotic strains need to be clinically tested. Furthermore, microbial components or their metabolites should also be examined; in particular, the application of these novel microbial drugs to the diseased site (eg, the airways) must be explored. Lastly, there are no microbial therapeutics currently approved for routine clinical practice, and significant effort and investment are still required to identify the optimal microbial interventions for allergy and asthma. Location Phyll (Genus) Effect Reference Oral cavity ↑ Gemella haemolysans ↓ Lactobacillus gasseri, Lactobacillus crispatus Increased risk of allergic diseases 47 Intestine ↑ Staphylococcus species Protection against oral sensitization and allergic responses 92 Intestine ↑ Clostridia, Firmicutes Milk allergy resolution 49 Intestine ↑ Lachnospiraceae, Ruminococcaceae Associated with egg allergy 50 Intestine ↓ Haemophilus, Dialister, Dorea, Clostridium Associated with food sensitization 51 Intestine ↓ Citrobacter, Oscillospira, Lactococcus, Dorea Associated with food allergy 51 Intestine ↓ Escherichia coli High faecal calprotectin, impaired IL‐10 activation, increased risk of AD and asthma 43 Intestine ↓ Bifidobacterium Correlates with AD severity 37 Intestine Early colonization with C. difficile Associated with AD development 38 Intestine ↓ Bacteroidetes diversity Associated with AD development 39 Intestine ↓ Akkermansia muciniphila, Ruminococcus gnavus and Lachnospiraceae Associated with AD development 40 Intestine ↓ Lachnospira, Veillonella, Faecalibacterium, Rothia Reduced levels of faecal SCFAs, increased risk of asthma 56 Intestine ↓ Bifidobacterium, Akkermansia, Faecalibacterium ↑ Candida, Rhodotorula Increased risk of developing multisensitized atopy, increased circulating proinflammatory metabolites 58 Upper airways Early colonization with Staphylococcus species, Corynebacterium, Dolosigranulum, Moraxella Associated with lower rate of respiratory infections in the first 2 years of life 65-67 Upper airways Early colonization with Streptococcus, Moraxella, Haemophilus Increased risk of virus‐induced acute respiratory infections and increased risk of asthma 60 Upper airways ↑ Proteobacteria Associated with rhinitis in children 69 Nasopharynx ↑ Haemophilus influenzae, Streptococcus species Increased risk of hospitalization during RSV infection 62 Nasopharynx Colonization with Staphylococcus aureus Decreased risk of hospitalization during RSV infection 63 Nasopharynx ↑ Streptococcus, Staphylococcus Abnormalities in functional tests of the respiratory system 64 Nasopharynx ↑ Staphylococcus aureus ↓ P. acnes Associated with high IgE levels 87 Nasopharynx ↑ Firmicutes (Staphylococcus & Streptococcus), Proteobacteria (Haemophilus, Pseudomonas & Moraxella), Fusobacteria ↓ Corynebacterium Associated with CRS in adults 88 Nasopharynx ↑ Streptococcus, Haemophilus, Fusobacteria ↓ Diversity Associated with CRS without nasal polyps in adults 89 Nasopharynx ↑ Lactobacillus during acute respiratory infection with RSV Reduced risk of wheezing 59 Hypopharynx Colonization with Moraxella catarrhalis, Haemophilus influenzae, Streptococcus pneumoniae Low‐grade systemic inflammation 61 Lower airways ↑ Proteobacterium (Klebsiella species) (Mycoplasma pneumoniae, Chlamydophila pneumoniae) Associated with severe asthma 77, 79 Lower airways ↑ Actinobacteria Improvement in asthma control 77 Lower airways ↑ Neisseria, Haemophilus, Campylobacter, Leptotrichia Associated with resistance to corticosteroids in asthma 75 Sputum ↑ Proteobacteria Associated with neutrophilic asthma exacerbations 78 Sputum ↑ Bacteroidetes Associated with eosinophilic asthma exacerbations 78 Skin ↑ Staphylococcus aureus Epidermal barrier dysfunction, cutaneous inflammation, formation of AD skin lesions, associated with AD severity and allergen sensitization, associated with susceptibility to eczema herpeticum among AD patients 19, 21, 23 Skin Colonization with single clonal Staphylococcus aureus strains Associated with AD severity 26 Skin ↑ Malassezia species. Associated with AD severity 35 Skin ↑ Corynebacterium, Proteobacterium Associated with AD severity 21 Skin ↑ coagulase‐negative staphylococci: (Staphylococcus epidermidis, S. hominis, S. lugdunensis) Limits Staphylococcus aureus overgrowth 28 Skin Colonization with S. epidermidis TLR2 activation, epidermal barrier maintenance 1 Skin ↓ Proteobacteria (Propionibacterium, Streptococcus, Acinetobacter, Corynebacterium, Prevotella) Associated with AD 28, 30 Skin Early colonization with S. epidermidis Local activation of the host immune response through induction of S. epidermidis‐specific FOXP3 Treg cells 29 Skin ↑ in resident skin bacteria Associated with AD flares 24 This table summarizes the bacterial changes that have been associated with atopic dermatitis, food allergy or asthma. CONFLICTS OF INTEREST LOM is a consultant to Alimentary Health Ltd and has received research funding from GSK. NL, PS, ZL, MS and TE have no conflict of interest in relation to this work. AUTHOR CONTRIBUTIONS NL, PS, ZL, MS, TE and LOM contributed to drafting the manuscript. All authors read, reviewed and agreed the final version of this manuscript. REFERENCES Notes : This table summarizes the bacterial changes that have been associated with atopic dermatitis, food allergy or asthma.
No comment yet.
Rescooped by Gilbert C FAURE from Immunology and Biotherapies
November 19, 2018 3:46 AM
Scoop.it!

Oral Desensitization to Peanuts | NEJM

Oral Desensitization to Peanuts | NEJM | Allergy (and clinical immunology) | Scoop.it
Editorial from The New England Journal of Medicine — Oral Desensitization to Peanuts..

 

A series of case reports and small studies have shown that the systematic introduction of tiny amounts of peanut allergen, followed by gradual increases in dose, could prevent or attenuate systemic reactions.1-4 The concept gained traction when a group in Cambridge, United Kingdom, found that 12% defatted peanut flour could induce desensitization in children.5

Vickery and colleagues now present in the Journal6 the results of a randomized, controlled trial involving approximately 550 participants with peanut allergy. The trial used a Good Manufacturing Process–produced 12% defatted peanut flour preparation, known as AR101, as the allergen. 

 

AR101 and other, similar products such as CA002, which is being developed by the Cambridge group, would therefore appear to have a role in initial dose escalation. The potential market for these products is believed to be billions of dollars.10 It is perhaps salutary to consider that in the study conducted by the Cambridge group, children underwent desensitization with a bag of peanut flour costing peanuts.

.


Via Krishan Maggon
No comment yet.
Rescooped by Gilbert C FAURE from Immunology and Biotherapies
October 11, 2018 3:28 PM
Scoop.it!

Feasibility of desensitizing children highly allergic to peanut by high‐dose oral immunotherapy - Reier‐Nilsen - - Allergy - Wiley Online Library

1 Background There are limited data on the feasibility, efficacy and safety of high‐dose oral immunotherapy (OIT) in children highly allergic to peanuts. 2 Objective In children highly allergic to peanut, we primarily aimed to determine the feasibility of reaching the maximum maintenance dose (MMD) of 5000 mg peanut protein or, alternatively, a lower individual maintenance dose (IMD), by OIT up‐dosing. Secondarily, we aimed to identify adverse events (AEs) and determine factors associated with reaching a maintenance dose. 3 Methods The TAKE‐AWAY peanut OIT trial enrolled 77 children 5‐15 years old, with a positive oral peanut challenge. Fifty‐seven were randomized to OIT with biweekly dose step‐up until reaching MMD or IMD and 20 to observation only. Demographic and biological characteristics, AEs, medication and protocol deviations were explored for associations with reaching maintenance dose. 4 Results All children had anaphylaxis defined by objective symptoms in minimum two organ systems during baseline challenge. The MMD was reached by 21.1%, while 54.4% reached an IMD of median (minimum, maximum) 2700 (250, 4000) mg peanut protein, whereas 24.5% discontinued OIT. During up‐dosing, 19.4% experienced anaphylaxis. Not reaching the MMD was caused by distaste for peanuts (66.7%), unacceptable AEs (26.7%) and social reasons (6.7%). Increased peanut s‐IgG4/s‐IgE ratio (OR [95% CI]: 1.02 [1.00, 1.04]) was associated with reaching MMD. 5 Conclusion Although 75.5% of children with peanut anaphylaxis reached a maintenance dose of 0.25‐5 g, only 21.1% reached the MMD. Distaste for peanuts and AEs, including high risk of anaphylaxis, limited the feasibility of reaching MMD.

Via Krishan Maggon
No comment yet.
Scooped by Gilbert C FAURE
June 19, 2018 8:38 AM
Scoop.it!

Peanut allergy: an overview

Peanut allergy: an overview | Allergy (and clinical immunology) | Scoop.it
PEANUT ALLERGY ACCOUNTS FOR THE MAJORITY of severe food-related allergic reactions. It tends to present early in life, and affected individuals generally do not outgrow it. In highly sensitized people, trace quantities can induce an allergic reaction.
No comment yet.
Scooped by Gilbert C FAURE
May 3, 2018 11:52 AM
Scoop.it!

Identifying peanut allergies cheaper and easier with new test | Society | The Guardian

Identifying peanut allergies cheaper and easier with new test | Society | The Guardian | Allergy (and clinical immunology) | Scoop.it
Scientists says blood test could avoid costly, stressful, food tests for confirming allergy
No comment yet.
Rescooped by Gilbert C FAURE from Immunology and Biotherapies
April 5, 2018 1:52 PM
Scoop.it!

AR101 for Peanut Allergy | Aimmune | CODIT Oral Immunotherapy

AR101 for Peanut Allergy | Aimmune | CODIT Oral Immunotherapy | Allergy (and clinical immunology) | Scoop.it
Aimmune Therapeutics develops treatments to protect children with food allergies (peanut, milk, and egg) from the consequences of accidental exposure.

Via Krishan Maggon
No comment yet.
Scooped by Gilbert C FAURE
December 26, 2017 3:14 AM
Scoop.it!

What Is Happening with Peanut Allergy Guidelines? Tough nut to crack for parents and pediatricians

What Is Happening with Peanut Allergy Guidelines? Tough nut to crack for parents and pediatricians | Allergy (and clinical immunology) | Scoop.it
Adopting the new peanut guidelines may be difficult for doctors and parents and will require further education.

(Results from the December 20 poll: more than 80% of responders answered "Yes" to the question, "Should patients with drug allergies be periodically re-tested?")

Source



 
No comment yet.
Scooped by Gilbert C FAURE
October 3, 2017 6:35 AM
Scoop.it!

The efficacy of oral and subcutaneous antigen-specific immunotherapy in murine cow’s milk- and peanut allergy models

The efficacy of oral and subcutaneous antigen-specific immunotherapy in murine cow’s milk- and peanut allergy models | Allergy (and clinical immunology) | Scoop.it
Antigen-specific immunotherapy (AIT) is a promising therapeutic approach for both cow’s milk allergy (CMA) and peanut allergy (PNA), but needs optimization in terms of efficacy and safety. Compare oral immunotherapy (OIT) and subcutaneous immunotherapy (SCIT) in murine models for CMA and PNA and determine the dose of allergen needed to effectively modify parameters of allergy. Female C3H/HeOuJ mice were sensitized intragastrically (i.g.) to whey or peanut extract with cholera toxin. Mice were treated orally (5 times/week) or subcutaneously (3 times/week) for three consecutive weeks. Hereafter, the acute allergic skin response, anaphylactic shock symptoms and body temperature were measured upon intradermal (i.d.) and intraperitoneal (i.p.) challenge, and mast cell degranulation was measured upon i.g. challenge. Allergen-specific IgE, IgG1 and IgG2a were measured in serum at different time points. Single cell suspensions derived from lymph organs were stimulated with allergen to induce cytokine production and T cell phenotypes were assessed using flow cytometry. Both OIT and SCIT decreased clinically related signs upon challenge in the CMA and PNA model. Interestingly, a rise in allergen-specific IgE was observed during immunotherapy, hereafter, treated mice were protected against the increase in IgE caused by allergen challenge. Allergen-specific IgG1 and IgG2a increased due to both types of AIT. In the CMA model, SCIT and OIT reduced the percentage of activated Th2 cells and increased the percentage of activated Th1 cells in the spleen. OIT increased the percentage of regulatory T cells (Tregs) and activated Th2 cells in the MLN. Th2 cytokines IL-5, IL-13 and IL-10 were reduced after OIT, but not after SCIT. In the PNA model, no differences were observed in percentages of T cell subsets. SCIT induced Th2 cytokines IL-5 and IL-10, whereas OIT had no effect. We have shown clinical protection against allergic manifestations after OIT and SCIT in a CMA and PNA model. Although similar allergen-specific antibody patterns were observed, differences in T cell and cytokine responses were shown. Whether these findings are related to a different mechanism of AIT in CMA and PNA needs to be elucidated.
No comment yet.