Abstract
Anaphylaxis is an acute life threatening systemic allergic reaction that can have a wide range of clinical manifestations. The most common triggers for anaphylaxis include food, medication, and venom. What is curious regarding anaphylaxis is how so many different agents can induce a severe systemic clinical response but only in a select subgroup of patients. Over the past decade, several important advances have been made in understanding the underlying cellular and molecular mechanisms contributing to anaphylaxis, with mast cells being an essential component. Classically, crosslinked IgE bound to its high affinity receptor induces mast cell mediator release. However, toll-like, complement, or Mas-related G protein-coupled receptors also activate mouse and human mast cells. While anaphylaxis secondary to foods historically has been more extensively characterized clinically and mechanistically, more recent studies have shifted focus towards understanding drug-induced anaphylaxis. The focus of this review is to highlight recent basic science developments and compare what is currently known regarding anaphylaxis to food, medications, and venom.
Keywords: Anaphylaxis, Food Allergy, Venom Allergy, Drug Allergy, Mast cell, IgE, MRGPRX2, Complement
Introduction
Anaphylaxis is an acute life threatening systemic allergic reaction that can have a wide range of clinical manifestations including urticaria, respiratory distress, nausea, vomiting, diarrhea, and/or hypotension1. In children, the most common causes of anaphylaxis are foods including egg, cow’s milk, wheat, peanut, tree nut, fish, shellfish, and soy, yet it is possible that the induction and effector mechanism induced by each of these food allergens differs. While some food allergies can persist into adulthood, the most common triggers of anaphylaxis in adults are instead medications and venom2. Medications accounted for more than half of severe fatal anaphylaxis in the US over a 12-year period3. While the culprit drug was not specified in the vast majority of these cases, antibiotics, radiocontrast media, neuromuscular blocker agents (NMBAs), and antineoplastic agents were implicated at the highest frequencies3. In regard to venom-induced anaphylaxis, Hymenoptera (bee and wasp) stings are the most prominent causes in North America and Europe, however, reactions to the venoms of snakes, other invertebrates, and cold-blooded vertebrates have also been described4. Mast cells (MCs) are essential mediators of anaphylaxis, likely for all of these classes of triggers, although other cell types can contribute. Classically, crosslinked IgE bound to its high affinity receptor induces MC mediator release. However, toll-like, complement or Mas-related G protein-coupled (MRG) receptors and even non-IgE antibodies also activate mouse and human MCs5. This review will highlight basic science developments over the past five years regarding our evolving understanding of anaphylaxis to food, medications, and venom.
Food-Induced Anaphylaxis
Recent work has uncovered mechanistic aspects of how MCs are activated by food-specific antibodies and the lymphocytes that regulate this antibody response. Kita and colleagues recently confirmed numerous older studies that anaphylaxis in murine food allergy models can also be induced by activating MCs via both IgE and IgG Fc receptors6. An important link between gut mast cell numbers and skin inflammation has also been elucidated7 to explain a well-established clinical association between eczema and food allergies. Skin tape stripping results in release of systemic IL-33 that acts on gut ILC2s to make IL-4 and IL-13 and MC expansion7. Subsequently, mucosal MCs are triggered by allergens but how luminal antigens reach MCs remained unclear. Elegant work using mouse models demonstrated that particular types of gut epithelium transport food allergens to submucosal MCs thereby promoting anaphylaxis8.
Regulatory T cells (Tregs) have a clear role in tolerance to food antigens9; recent work using tetramers in mice tracked the early induction of food-specific anergic T cells showing that with oral tolerance these cells ultimately transform into Tregs and suppress other T effector populations10. The microbiome regulates the functionality of Tregs to food. Clostridial species activate Tregs through innate immune pathways that promote tolerance to food allergens11. Feehley et al. identified changes also in Clostridial species in children sensitized to milk and, using a mouse model, showed that gut microbiota from these children predispose to oral anaphylaxis and alter the transcriptional profiles of intestinal epithelial cells12. However, both IL-4 and particular microbiota can overcome the protective effects of TGFb produced by Tregs to food, thus predisposing to anaphylaxis13, highlighting the interplay between immune cells and the gut environment for food allergy.
Work over the past five years has advanced our understanding of the T cells that guide IgE induction; these studies demonstrate that Tfh cells, not Th2, are required for allergen specific IgE induction14 15. We demonstrated that a subset of Tfh cells, marked by expression of GATA3 and IL-13 (termed “Tfh13”), is responsible for induction of high affinity IgE that potently induces anaphylaxis14; where these cells act in the follicle to promote IgE, and the specific role of IL-13 remains unknown. Oral tolerance to allergens induced during early life prevents the induction of Il4-expressing Tfh cells in mice through a CTLA-4 dependent mechanism16. However, oral immunotherapy of patients with peanut allergy suppressed multiple T effector cell populations without altering allergen specific Tfh cells17 raising questions about how difficult Tfh cell repression might be in patients. Blocking the interaction of Tfh and B cells through PD1 blockade inhibits IgE production while promoting low affinity IgG, possibly by deregulating the germinal center reaction in mouse models and was associated with reduced anaphylaxis6, 18. Finally, the timing of adjuvant and allergen exposure in a mouse food allergy model was essential for induction of food specific IgE by promoting IgE+ B cell survival and expansion19. These insights into the nature and timing of T-B cell interactions driving food-reactive antibodies provide new mechanistic approaches to redirecting the allergic response in patients with food allergy while highlighting the challenges of doing so.
Alpha-gal syndrome is a novel form of food-induced anaphylaxis where, in contrast to other foods, clinical symptoms typically develop 2–6 hours following ingestion of a product containing the polysaccharide galactose alpha 1,3 galactose (alpha-gal)20. Alpha-gal is present on cells and tissues of non-primate mammals and patients with alpha-gal syndrome typically experience delayed reactions after ingestion of mammalian meats including beef, pork, and lamb. In the US, sensitization to alpha-gal is thought to occur through lone star tick bites which in part explains the geographical variation observed in disease prevalence21. However, there are many unanswered questions regarding alpha-gal syndrome including the underlying mechanisms by which tick bites induce alpha-gal specific IgE and why clinical symptoms are delayed following ingestion22. In regard to the latter, Román-Carrasco and colleagues found that alpha-gal can only pass through an intestinal epithelial monolayer in vitro when bound to lipids and not proteins. They suggest that alpha-gal may be entering the circulation mainly in the form of chylomicrons, and the slow release of the allergen from these structures may in turn be responsible for the delayed presentation of clinical symptoms following ingestion23.
Medication-Induced Anaphylaxis
Similar to food, IgE-mediated MC degranulation has also been associated with medication-induced anaphylaxis. However, unlike food, most drugs are too small by themselves to be recognized by IgE. Instead, some drugs are haptens, small molecules that are recognized by adaptive immune cells only when attached to a larger protein. Penicillin is one of the most well-characterized haptens and penicilloyl, the major hapten-carrier complex, is responsible for more than half of IgE-mediated reactions to the drug24. However, it is unclear how penicillin (or hapten)-specific IgE is generated and whether Tfh cells or Tregs, for example, play a similar role as in food-induced anaphylaxis. Identifying and quantifying drug specific IgE is also more limited than in food allergy as clinically validated skin testing and immunoassays are not available for most medications.
Cetuximab is a chimeric monoclonal antibody which targets the epidermal growth factor receptor and is associated with a unique form of IgE-mediated anaphylaxis. In this instance, specific IgE is not directed against an epitope of the drug itself but instead to the sugar glycosylating a portion of the cetuximab heavy chain, galactose alpha 1,3 galactose (alpha-gal)25. This is the first known instance of a specific IgE directed to a polysaccharide in a medication and is also related to the alpha-gal syndrome as discussed above.
In addition to IgE and mast cells, IgG and neutrophils may also play a role in certain forms of drug-induced anaphylaxis. Recently, Jonsson and colleagues found those patients with severe anaphylaxis to NMBA had elevated concentrations of anti-NMBA IgG as well as higher levels of neutrophil activation and platelet activating factor (PAF) compared to controls who did not develop anaphylaxis26. They propose that the anti-NMBA IgG complexes primarily engage FcγRIIA (CD32a) and FcγRIII (CD16) on the surface of neutrophils leading to their activation and downstream release of PAF, a potent mediator of anaphylaxis associated with smooth muscle contraction and vasodilation. Importantly, in this cohort, 31% of patients lacked biomarkers consistent with IgE-mediated anaphylaxis, which illustrates that IgG (and neutrophil) mediated anaphylaxis can occur with or without an IgE mediated hypersensitivity reaction. It is likely that other drugs can also induce IgG-mediated anaphylaxis, but an exhaustive list of these agents is not currently known.
Complement is another pathway that has been associated with drug-induced anaphylaxis. C3a and C5a, aptly named anaphylatoxins, are products of complement activation that can induce mast cell and basophil degranulation. Cremophor, a diluent commonly found in the chemotherapy agent, paclitaxel, can directly activate the complement system27. A recent study found that paclitaxel (independent of Cremophor) can also directly engage the C5aR1 to elicit anaphylaxis in murine models28.
Finally, the Mas-related G protein-coupled receptor X2 (MRGPRX2) is predominantly expressed on mast cells in the skin but can be found at lower levels in the lungs and gut. While the natural ligand for MRGPRX2 is not known, small molecules that carry a net positive charge have been identified that can bind to this receptor which in turns leads to direct mast cell activation, release of tryptase, histamine, and prostaglandins29. Numerous drugs including fluroquinolones, vancomycin, certain neuromuscular blockers, and certain anti-depressants have been shown to activate MRGPRX2 in human and murine mast cells in vitro29–32. In contrast to IgE-mediated reactions, activation of MRGPRX2 does not require prior exposure to the drug, can occur on the first administration, and requires high enough concentrations of the inciting drug to trigger receptor activation. This, in addition to the receptor mainly being expressed in the skin, raises questions as to the precise role MRGPRX2 has in the development of anaphylaxis (a systemic response) versus a cutaneous reaction (a local response) and warrants further investigation.
While outside the scope of this review, there are countless other medications that have been associated with anaphylaxis in select individuals and the underlying mechanisms by which this anaphylaxis occurs is likely to vary. Notably, during the COVID-19 pandemic, episodes of anaphylaxis following administration of COVID mRNA vaccines were reported albeit the exact component of the vaccine responsible for inducing anaphylaxis remains unclear33. Interestingly, in a recent study of patients with a history of a severe reaction to their first COVID vaccine (including 19 with anaphylaxis), all tolerated their second dose of the vaccine34. This would suggest against a traditional IgE-mediated driven mechanism for anaphylaxis but complement instead may be playing a role. Further studies exploring this phenomena are thus indicated.
Venom Induced Anaphylaxis
Systemic venom reactions occur more often in middle-aged and elderly patients and are more commonly associated with cardiovascular and mast cell diseases compared with food-induced anaphylaxis35. In particular, clonal mast cell disorders, mainly systemic mastocytosis, predisposes patients to severe venom-induced anaphylaxis. However, recently, hereditary α-tryptasemia, which is a non-clonal mast cell disease caused by increased germline copies of TPSAB1 that encodes α-tryptase (one of the major proteases secreted by mast cells)36, has been identified as a risk factor for severe venom-induced anaphylaxis37. The mechanism of how this increased α-tryptase expression contributes to or is associated with venom-induced anaphylaxis remains unknown.
The role of the adaptive immune system, including IgE, is less clear in venom-induced anaphylaxis than in food-induced anaphylaxis. Venom-specific IgE is detectable in almost all patients with a history of venom-induced anaphylaxis, and it enhances MC degranulation upon venom exposure38. Conversely, the majority of people with IgE to venom do not react with anaphylaxis39. Like drugs but unlike most food allergens, venom components can directly induce MC degranulation without specific IgE40. In addition, Elst and colleagues showed that bone marrow mast cells from patients with a clonal mast cell disorder and history of venom anaphylaxis overexpress FcεRI (but not MRGPRX2), emphasizing the complexity of the interactions between IgE and mast cells in venom-induced anaphylaxis41. Interestingly, in mice the ultimate result of MC degranulation has shown beneficial, detoxifying effects on venoms and thus the development of an IgE response to venom has been proposed to be a protective, rather than pathogenic, response to venoms40, 42.
The mechanism leading to venom sensitization also remains largely unknown. Foundational mechanistic studies in mice were performed by Palm et al. a decade ago, which revealed that enzymatically active phospholipase A2 (PLA2) - one of the main allergens in bee venom - induced release of interleukin-33 and the Th2 response but not sIgE42. Melittin, which is the major component of bee venom, induces inflammation via the inflammasome/caspase1/IL1β pathway43. Ogden et al. confirmed that enzymatically active PLA2 is a potent adjuvant that shifts the immune response toward Th2 cells44. Bruni et al. reported that the IgE-immune response to the venom of Thalassophryne nattereri (venomous fish from South America) was IL-4-dependent45 and that the main component of this venom, natterin, induced NLRP6 and NLRC4 inflammasome-dependent neutrophilic inflammation in the lung46. The relative roles of pro-type 2 signals such as IL-33 and IL-4 versus the IL-1beta/inflammasome pathways in protection from envenoming remain unclear and likely are complementary rather than directly linked43.
Unmet Needs and Future Directions
Anaphylaxis is a markedly heterogenous condition and therefore we and others47 have emphasized the need to interpret data in the context of specific triggers and patient populations rather than applying anaphylaxis mechanisms broadly to clinically related syndromes. Patients can present with a myriad of cutaneous, respiratory, cardiovascular, or gastrointestinal symptoms (i.e., phenotypes) that fulfill the clinical definition. Additionally, as discussed above, there are multiple different cellular and molecular mechanisms (i.e., endotypes) that can be involved. Two major endotypes of anaphylaxis have been recognized based on whether the response is IgE-dependent or IgE-independent. While the former endotype is more uniform, involving the classical activation of mast cells and basophils, the latter endotype is diverse and includes direct mast cell activation via MRGPRX2, IgG-mediated activation of mast cells, basophils, and neutrophils through the FcγRs, and complement activation. More mechanistic studies of anaphylaxis have been completed in murine models and translation to human models and clinical disease is needed.
Another unmet need in anaphylaxis is the identification of a factor that could reliably and quickly link a particular clinical phenotype with an underlying inflammatory endotype. In patients with chemotherapy-induced reactions, serum levels of IL-6 may be associated more specifically with cardiovascular and neuromuscular symptoms as opposed to cutaneous or respiratory symptoms48. Tryptase is a granule protein released by mast cells upon activation that clinically can be used as an indicator for anaphylaxis. However, not all cases of anaphylaxis are associated with elevated tryptase levels and vice versa. Additionally, elevated serum tryptase levels cannot discern if the mast cell was activated by an IgE-mediated, MRGPRX2-mediated or another pathway. Histamine metabolites N-methylhistamine and N-methylimidazole acetic acid, the prostaglandin D2 metabolite 2,3-dinor-11-beta-PGF2-alpha, and leukotriene (LT) E4 may also be elevated in the urine during and for a period of time after anaphylaxis. However, these mediators cannot distinguish which mechanism induced anaphylaxis and the clinical specificity and sensitivity of these markers has not been well established. Recently, hsa-miR-451a, a miRNA expressed mostly by neutrophils, was identified as a novel potential biomarker of acute anaphylaxis but further work is needed to evaluate its clinical utility49. Thus, while progress has been made, additional studies are still needed to identify biomarkers unique to specific mechanisms of anaphylaxis. By identifying the immunologic pathway contributing to a specific clinical reaction, the ideal goal would be to then initiate a more tailored and effective treatment.
While the acute treatment of anaphylaxis (epinephrine) is well established, the long-term management of these patients historically has been avoidance of the inciting agent. The exception being in patients with venom-induced anaphylaxis where venom immunotherapy has been shown to be highly effective and immune modulating50. Recently, oral immunotherapy to peanut has been FDA approved in children. However, to date, this therapy has not been shown to permanently modify the immune response to peanut but instead can potentially raise the threshold for which anaphylaxis to peanut is induced51. Likewise, drug desensitizations can be successfully performed to allow a patient to temporarily receive a drug that caused anaphylaxis in the past52. However, outside of the desensitization, the risk of anaphylaxis to that drug remains. A novel way of preventing anaphylaxis in mice was reported through the inhibition of FcεRI-mediated signaling with acalabrutinib, a BTK inhibitor53. Evaluation of the effects of acalabrutinib in humans is needed as is the development of other therapies for the chronic management of patients with anaphylaxis.
In addition to managing anaphylaxis after it has occurred, it is also important to focus on understanding why it develops in the first place. It is unclear why only certain patients develop anaphylaxis and not others. For MRGPRX2, elevated expression on mast cells has been observed in certain patient populations such as chronic spontaneous urticaria54 and maculopapular cutaneous mastocytosis55. Variants in MRGPRX2 associated with a loss of function have also been identified56. However, there is not yet an assay able to predict who is at high risk of developing a reaction.
Genetic factors likely play a role in anaphylaxis as, for example, increased copies of TPSAB1 have been associated with significantly higher frequency of anaphylaxis among patients with mastocytosis57 and are more common in patients with a history of idiopathic anaphylaxis compared to the general population 37. Regarding food-induced anaphylaxis, the HLADPB1*02:01:02 allele was associated with a significantly increased risk of wheat-dependent exercise-induced anaphylaxis58 and specific loci in the HLA-DR/DQ region have been associated with self-reported reactions to shrimp and peach in Japanese populations59. Additionally, the SERPINB gene cluster at 18q21.3 was identified as a susceptibility locus for food allergy60. Together these studies highlight the continued need to investigate genetic factors that could predict susceptibility of developing anaphylaxis.
Finally, while food, medication, and venom represent some of the most common triggers of anaphylaxis, as many as 14–59% of cases report no identifiable cause61. These cases of exclusion are diagnosed as having idiopathic anaphylaxis and even less known about the underlying mechanisms by which this form of anaphylaxis occurs. Some of the cases classified decades ago as idiopathic have now been demystified due to recognition of novel mechanisms involved in anaphylaxis with alpha-gal syndrome being a prime example. However, there are many cases of anaphylaxis that remain unresolved, and it is that hope that as more is understood about other underlying mechanisms important in causing anaphylaxis, less clinical cases will remain idiopathic.
In conclusion, these observations highlight the overall complexity inherent in studying the mechanisms of anaphylaxis but also the incredible impact research in this area could have on the lives of a wide spectrum of patients.
Acknowledgments
MK is supported by the German Foundation Walter Benjamin Fellowship from the German Research Foundation (464546188; KR 5534/1-1). WS is supported by K23AI141694, SCE is supported by R01AI136942, R56AI155497, FARE foundation, Food Allergy Fund and the Food Allergy Science Initiative.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest Disclosure:
W. Stevens has served as an advisor to GlaxoSmithKline, Bristol Myers Squibb, Genentech, and Regeneron. M. Kraft reports travel support from ALK-Abéllo and speaker honoraria from Bencard Allergie GmbH, outside the submitted work. Magdalena Kraft and Stephanie Eisenbarth have nothing to report.
References
- 1.Shaker MS, Wallace DV, Golden DBK, Oppenheimer J, Bernstein JA, Campbell RL, et al. Anaphylaxis-a 2020 practice parameter update, systematic review, and Grading of Recommendations, Assessment, Development and Evaluation (GRADE) analysis. J Allergy Clin Immunol 2020; 145:1082–123. [DOI] [PubMed] [Google Scholar]; *A recent evidence-based clinical guideline for the diagnosis and management of anaphylaxis.
- 2.Lee S, Hess EP, Lohse C, Gilani W, Chamberlain AM, Campbell RL. Trends, characteristics, and incidence of anaphylaxis in 2001–2010: A population-based study. J Allergy Clin Immunol 2017; 139:182–8 e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jerschow E, Lin RY, Scaperotti MM, McGinn AP. Fatal anaphylaxis in the United States, 1999–2010: temporal patterns and demographic associations. J Allergy Clin Immunol 2014; 134:1318–28 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Swiontek K, Planchon S, Ollert M, Eyer F, Fischer J, Hilger C. Phospholipase A2 Triggers Anaphylaxis to Snake Venom by Repeated Skin Sensitization: A Case Report. J Investig Allergol Clin Immunol 2021; 31:175–7. [DOI] [PubMed] [Google Scholar]
- 5.Cianferoni A Non-IgE-mediated anaphylaxis. J Allergy Clin Immunol 2021; 147:1123–31. [DOI] [PubMed] [Google Scholar]
- 6.Lama JK, Iijima K, Kobayashi T, Kita H. Blocking the inhibitory receptor programmed cell death 1 prevents allergic immune response and anaphylaxis in mice. J Allergy Clin Immunol 2022; 150:178–91 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Leyva-Castillo JM, Galand C, Kam C, Burton O, Gurish M, Musser MA, et al. Mechanical Skin Injury Promotes Food Anaphylaxis by Driving Intestinal Mast Cell Expansion. Immunity 2019; 50:1262–75 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]; *An important recent paper in the field establishing how a skin-gut connection might promote food allergy through cutenouse allergen exposure uing mouse models.
- 8.Noah TK, Lee JB, Brown CA, Yamani A, Tomar S, Ganesan V, et al. Thermoneutrality Alters Gastrointestinal Antigen Passage Patterning and Predisposes to Oral Antigen Sensitization in Mice. Front Immunol 2021; 12:636198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Stephen-Victor E, Crestani E, Chatila TA. Dietary and Microbial Determinants in Food Allergy. Immunity 2020; 53:277–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hong SW, Krueger PD, Osum KC, Dileepan T, Herman A, Mueller DL, et al. Immune tolerance of food is mediated by layers of CD4(+) T cell dysfunction. Nature 2022; 607:762–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Abdel-Gadir A, Stephen-Victor E, Gerber GK, Noval Rivas M, Wang S, Harb H, et al. Microbiota therapy acts via a regulatory T cell MyD88/RORgammat pathway to suppress food allergy. Nat Med 2019; 25:1164–74. [DOI] [PMC free article] [PubMed] [Google Scholar]; *Two papers (also see Feehley et al) helping clarify how microbiota might impact food allergy.
- 12.Feehley T, Plunkett CH, Bao R, Choi Hong SM, Culleen E, Belda-Ferre P, et al. Healthy infants harbor intestinal bacteria that protect against food allergy. Nat Med 2019; 25:448–53. [DOI] [PMC free article] [PubMed] [Google Scholar]; *Two papers (also see Abdel-Gadir et al) helping clarify how microbiota might impact food allergy.
- 13.Turner JA, Stephen-Victor E, Wang S, Rivas MN, Abdel-Gadir A, Harb H, et al. Regulatory T Cell-Derived TGF-beta1 Controls Multiple Checkpoints Governing Allergy and Autoimmunity. Immunity 2020; 53:1202–14 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Gowthaman U, Chen JS, Zhang B, Flynn WF, Lu Y, Song W, et al. Identification of a T follicular helper cell subset that drives anaphylactic IgE. Science 2019; 365. [DOI] [PMC free article] [PubMed] [Google Scholar]; * A study from our group identifying a new type of Tfh cell important for high affinity IgE to allergens, including food antigens.
- 15.Lama JK, Kita H. Tfh cells regulate antibody affinity and determine the outcomes of anaphylaxis. J Allergy Clin Immunol 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Krempski JW, Lama JK, Iijima K, Kobayashi T, Matsunaga M, Kita H. A mouse model of the LEAP study reveals a role for CTLA-4 in preventing peanut allergy induced by environmental peanut exposure. J Allergy Clin Immunol 2022; 150:425–39 e3. [DOI] [PMC free article] [PubMed] [Google Scholar]; * An important mechanistic dissection using mouse models of how early allergen introudction might prevent food allergy.
- 17.Monian B, Tu AA, Ruiter B, Morgan DM, Petrossian PM, Smith NP, et al. Peanut oral immunotherapy differentially suppresses clonally distinct subsets of T helper cells. J Clin Invest 2022; 132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bonamichi-Santos R, Aun MV, Kalil J, Castells MC, Giavina-Bianchi P. PD-L1 Blockade During Allergen Sensitization Inhibits the Synthesis of Specific Antibodies and Decreases Mast Cell Activation in a Murine Model of Active Cutaneous Anaphylaxis. Front Immunol 2021; 12:655958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chen Q, Xie M, Liu H, Dent AL. Development of allergen-specific IgE in a food-allergy model requires precisely timed B cell stimulation and is inhibited by Fgl2. Cell Rep 2022; 39:110990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tripathi A, Commins SP, Heymann PW, Platts-Mills TA. Delayed anaphylaxis to red meat masquerading as idiopathic anaphylaxis. J Allergy Clin Immunol Pract 2014; 2:259–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kersh GJ, Salzer J, Jones ES, Binder AM, Armstrong PA, Choudhary SK, et al. Tick bite as a risk factor for alpha-gal specific IgE antibodies and development of alpha-gal syndrome. Ann Allergy Asthma Immunol 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Platts-Mills TAE, Commins SP, Biedermann T, van Hage M, Levin M, Beck LA, et al. On the cause and consequences of IgE to galactose-alpha-1,3-galactose: A report from the National Institute of Allergy and Infectious Diseases Workshop on Understanding IgE-Mediated Mammalian Meat Allergy. J Allergy Clin Immunol 2020; 145:1061–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Roman-Carrasco P, Lieder B, Somoza V, Ponce M, Szepfalusi Z, Martin D, et al. Only alpha-Gal bound to lipids, but not to proteins, is transported across enterocytes as an IgE-reactive molecule that can induce effector cell activation. Allergy 2019; 74:1956–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Castells M, Khan DA, Phillips EJ. Penicillin Allergy. N Engl J Med 2019; 381:2338–51. [DOI] [PubMed] [Google Scholar]
- 25.Chung CH, Mirakhur B, Chan E, Le QT, Berlin J, Morse M, et al. Cetuximab-induced anaphylaxis and IgE specific for galactose-alpha-1,3-galactose. N Engl J Med 2008; 358:1109–17. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This paper describes a novel target for IgE-mediated medication induced anaphylaxis and is the first known instance of a specific IgE directed to a polysaccharide, galactose-alpha-1,3-galactose.
- 26.Jonsson F, de Chaisemartin L, Granger V, Gouel-Cheron A, Gillis CM, Zhu Q, et al. An IgG-induced neutrophil activation pathway contributes to human drug-induced anaphylaxis. Sci Transl Med 2019; 11. [DOI] [PubMed] [Google Scholar]; *This paper describes a new pathway for neuromuscular blocking agent induced anaphylaxis in humans involving IgG and neutrophil activation.
- 27.Szebeni J, Muggia FM, Alving CR. Complement activation by Cremophor EL as a possible contributor to hypersensitivity to paclitaxel: an in vitro study. J Natl Cancer Inst 1998; 90:300–6. [DOI] [PubMed] [Google Scholar]
- 28.Brandolini L, d’Angelo M, Novelli R, Castelli V, Giorgio C, Sirico A, et al. Paclitaxel binds and activates C5aR1: A new potential therapeutic target for the prevention of chemotherapy-induced peripheral neuropathy and hypersensitivity reactions. Cell Death Dis 2022; 13:500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.McNeil BD, Pundir P, Meeker S, Han L, Undem BJ, Kulka M, et al. Identification of a mast-cell-specific receptor crucial for pseudo-allergic drug reactions. Nature 2015; 519:237–41. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This paper highlights how several different classes of drugs can directly activate mast cells through the MRGPRX2 receptor.
- 30.Liu R, Hu S, Zhang Y, Che D, Cao J, Wang J, et al. Mast cell-mediated hypersensitivity to fluoroquinolone is MRGPRX2 dependent. Int Immunopharmacol 2019; 70:417–27. [DOI] [PubMed] [Google Scholar]
- 31.Wolf K, Kuhn H, Boehm F, Gebhardt L, Glaudo M, Agelopoulos K, et al. A group of cationic amphiphilic drugs activates MRGPRX2 and induces scratching behavior in mice. J Allergy Clin Immunol 2021; 148:506–22 e8. [DOI] [PubMed] [Google Scholar]
- 32.Chompunud Na Ayudhya C, Amponnawarat A, Roy S, Oskeritzian CA, Ali H. MRGPRX2 Activation by Rocuronium: Insights from Studies with Human Skin Mast Cells and Missense Variants. Cells 2021; 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Khalid MB, Frischmeyer-Guerrerio PA. “The Conundrum of COVID-19 mRNA Vaccine-Induced Anaphylaxis”. J Allergy Clin Immunol Glob 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Krantz MS, Kwah JH, Stone CA Jr., Phillips EJ, Ortega G, Banerji A, et al. Safety Evaluation of the Second Dose of Messenger RNA COVID-19 Vaccines in Patients With Immediate Reactions to the First Dose. JAMA Intern Med 2021; 181:1530–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Francuzik W, Rueff F, Bauer A, Bilo MB, Cardona V, Christoff G, et al. Phenotype and risk factors of venom-induced anaphylaxis: A case-control study of the European Anaphylaxis Registry. J Allergy Clin Immunol 2021; 147:653–62 e9. [DOI] [PubMed] [Google Scholar]
- 36.Lyons JJ, Yu X, Hughes JD, Le QT, Jamil A, Bai Y, et al. Elevated basal serum tryptase identifies a multisystem disorder associated with increased TPSAB1 copy number. Nat Genet 2016; 48:1564–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lyons JJ, Chovanec J, O’Connell MP, Liu Y, Selb J, Zanotti R, et al. Heritable risk for severe anaphylaxis associated with increased alpha-tryptase-encoding germline copy number at TPSAB1. J Allergy Clin Immunol 2021; 147:622–32. [DOI] [PubMed] [Google Scholar]; *Clinical study comparing a cohort of patients with systemic mastocytosis, or history of venom or idiopathic anaphylaxis with general population, demonstrating that hereditary a-tryptasemia is associated with increased risk of severe anaphylaxis to venom and more prevalent in patients with mastocytosis or history of idiopathic anaphylaxis than in general population.
- 38.Starkl P, Gaudenzio N, Marichal T, Reber LL, Sibilano R, Watzenboeck ML, et al. IgE antibodies increase honeybee venom responsiveness and detoxification efficiency of mast cells. Allergy 2022; 77:499–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lindstrom I, Holtta P, Suuronen K, Suomela S, Suojalehto H. High prevalence of sensitization to bumblebee venom among greenhouse workers. J Allergy Clin Immunol Pract 2022; 10:637–9. [DOI] [PubMed] [Google Scholar]
- 40.Marichal T, Starkl P, Reber LL, Kalesnikoff J, Oettgen HC, Tsai M, et al. A beneficial role for immunoglobulin E in host defense against honeybee venom. Immunity 2013; 39:963–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Elst J, De Puysseleyr LP, Ebo DG, Faber MA, Van Gasse AL, van der Poorten MM, et al. Overexpression of FcepsilonRI on Bone Marrow Mast Cells, but Not MRGPRX2, in Clonal Mast Cell Disorders With Wasp Venom Anaphylaxis. Front Immunol 2022; 13:835618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Palm NW, Rosenstein RK, Yu S, Schenten DD, Florsheim E, Medzhitov R. Bee venom phospholipase A2 induces a primary type 2 response that is dependent on the receptor ST2 and confers protective immunity. Immunity 2013; 39:976–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Palm NW, Medzhitov R. Role of the inflammasome in defense against venoms. Proc Natl Acad Sci U S A 2013; 110:1809–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ogden HL, Lai Y, Nolin JD, An D, Frevert CW, Gelb MH, et al. Secreted Phospholipase A2 Group X Acts as an Adjuvant for Type 2 Inflammation, Leading to an Allergen-Specific Immune Response in the Lung. J Immunol 2020; 204:3097–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bruni FM, Coutinho EMM, Andrade-Barros AI, Grund LZ, Lopes-Ferreira M, Lima C. Anaphylaxis induced by Thalassophryne nattereri venom in mice is an IgE/IgG1-mediated, IL-4-dependent phenomenon. Sci Rep 2020; 10:584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lima C, Falcao MAP, Andrade-Barros AI, Seni-Silva AC, Grund LZ, Balogh E, et al. Natterin an aerolysin-like fish toxin drives IL-1beta-dependent neutrophilic inflammation mediated by caspase-1 and caspase-11 activated by the inflammasome sensor NLRP6. Int Immunopharmacol 2021; 91:107287. [DOI] [PubMed] [Google Scholar]
- 47.Dribin TE, Schnadower D, Wang J, Camargo CA Jr., Michelson KA, Shaker M, et al. Anaphylaxis knowledge gaps and future research priorities: A consensus report. J Allergy Clin Immunol 2022; 149:999–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Jakubovic BD, Sanchez-Sanchez S, Hamadi S, Lynch DM, Castells M. Interleukin-6: A novel biomarker for monoclonal antibody and chemotherapy-associated hypersensitivity confirms a cytokine release syndrome phenotype-endotype association. Allergy 2021; 76:1571–3. [DOI] [PubMed] [Google Scholar]
- 49.Francuzik W, Pazur K, Dalke M, Dolle-Bierke S, Babina M, Worm M. Serological profiling reveals hsa-miR-451a as a possible biomarker of anaphylaxis. JCI Insight 2022; 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Boyle RJ, Elremeli M, Hockenhull J, Cherry MG, Bulsara MK, Daniels M, et al. Venom immunotherapy for preventing allergic reactions to insect stings. Cochrane Database Syst Rev 2012; 10:CD008838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Investigators PGoC, Vickery BP, Vereda A, Casale TB, Beyer K, du Toit G, et al. AR101 Oral Immunotherapy for Peanut Allergy. N Engl J Med 2018; 379:1991–2001. [DOI] [PubMed] [Google Scholar]
- 52.Khan DA, Banerji A, Blumenthal KG, Phillips EJ, Solensky R, White AA, et al. Drug Allergy: A 2022 Practice Parameter Update. J Allergy Clin Immunol 2022. [DOI] [PubMed] [Google Scholar]
- 53.Dispenza MC, Krier-Burris RA, Chhiba KD, Undem BJ, Robida PA, Bochner BS. Bruton’s tyrosine kinase inhibition effectively protects against human IgE-mediated anaphylaxis. J Clin Invest 2020; 130:4759–70. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This paper describes how BTK inhibitors may prevent the development of IgE-mediated anaphylaxis and could be a potential therapeutic target.
- 54.Fujisawa D, Kashiwakura J, Kita H, Kikukawa Y, Fujitani Y, Sasaki-Sakamoto T, et al. Expression of Mas-related gene X2 on mast cells is upregulated in the skin of patients with severe chronic urticaria. J Allergy Clin Immunol 2014; 134:622–33 e9. [DOI] [PubMed] [Google Scholar]
- 55.Deepak V, Komarow HD, Alblaihess AA, Carter MC, Metcalfe DD, Ali H. Expression of MRGPRX2 in skin mast cells of patients with maculopapular cutaneous mastocytosis. J Allergy Clin Immunol Pract 2021; 9:3841–3 e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Alkanfari I, Gupta K, Jahan T, Ali H. Naturally Occurring Missense MRGPRX2 Variants Display Loss of Function Phenotype for Mast Cell Degranulation in Response to Substance P, Hemokinin-1, Human beta-Defensin-3, and Icatibant. J Immunol 2018; 201:343–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sordi B, Vanderwert F, Crupi F, Gesullo F, Zanotti R, Bonadonna P, et al. Disease correlates and clinical relevance of hereditary alpha-tryptasemia in patients with systemic mastocytosis. J Allergy Clin Immunol 2022. [DOI] [PubMed] [Google Scholar]
- 58.Fukunaga K, Chinuki Y, Hamada Y, Fukutomi Y, Sugiyama A, Kishikawa R, et al. Genome-wide association study reveals an association between the HLA-DPB1( *)02:01:02 allele and wheat-dependent exercise-induced anaphylaxis. Am J Hum Genet 2021; 108:1540–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Khor SS, Morino R, Nakazono K, Kamitsuji S, Akita M, Kawajiri M, et al. Genome-wide association study of self-reported food reactions in Japanese identifies shrimp and peach specific loci in the HLA-DR/DQ gene region. Sci Rep 2018; 8:1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Marenholz I, Grosche S, Kalb B, Ruschendorf F, Blumchen K, Schlags R, et al. Genome-wide association study identifies the SERPINB gene cluster as a susceptibility locus for food allergy. Nat Commun 2017; 8:1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Carter MC, Akin C, Castells MC, Scott EP, Lieberman P. Idiopathic anaphylaxis yardstick: Practical recommendations for clinical practice. Ann Allergy Asthma Immunol 2020; 124:16–27. [DOI] [PubMed] [Google Scholar]