Skip to main content
VA Author Manuscripts logoLink to VA Author Manuscripts
. Author manuscript; available in PMC: 2020 Nov 3.
Published in final edited form as: J Allergy Clin Immunol. 2016 Apr 26;137(6):1674–1680. doi: 10.1016/j.jaci.2016.02.015

Rostrum: Human IgE-independent systemic anaphylaxis

Fred D Finkelman 1,2,3, Marat V Khodoun 1,2, Richard Strait 4,5
PMCID: PMC7607869  NIHMSID: NIHMS771114  PMID: 27130857

Abstract

Anaphylaxis is a rapidly developing, life-threatening, generalized or systemic allergic reaction that is classically elicited by antigen (Ag) crosslinking of Ag-specific IgE bound to the high affinity IgE receptor, FcεRI, on mast cells and basophils. This initiates signals that induce cellular degranulation, with release and secretion of vasoactive mediators, enzymes, and cytokines. IgE-independent mechanisms of anaphylaxis, however, have been clearly demonstrated in experimental animals. These include IgG-dependent anaphylaxis, which involves the triggering of mediator release by IgG/antigen complex crosslinking of FcγRs on macrophages, basophils and neutrophils; anaphylaxis mediated by the binding of complement-derived peptides, C3a and C5a, to their receptors on mast cells, basophils and other myeloid cells; and direct activation of mast cells by drugs that interact with receptors on these cells. Here, we review the mechanisms involved in these IgE-independent forms of anaphylaxis and the clinical evidence for their human relevance. We conclude that this evidence supports the existence of all three IgE-independent mechanisms as important causes of human disease, although practical and ethical considerations preclude their demonstration to the degree of certainty possible with animal models. Further, we cite evidence that different clinical situations can suggest different mechanisms as having a primal role in anaphylaxis and that IgE-dependent and distinct IgE-independent mechanisms can act together to increase anaphylaxis severity. As specific agents become available that can interfere with mechanisms involved in the different types of anaphylaxis, recognition of specific types of anaphylaxis is likely to become important for optimal prophylaxis and therapy.

Keywords: anaphylatoxin, complement, FcεR, FcγR, IgE, IgG, mast cell, basophil, mouse

Introduction

Anaphylaxis is a rapidly developing, life-threatening, generalized or systemic allergic reaction1. Foods, drugs, and insect stings are the most common causes of this disorder2. Classically, anaphylaxis is induced by antigen (Ag) crosslinking of Ag-specific IgE that has bound to the high affinity IgE receptor (FcεRI) on mast cells and basophils3. Crosslinking of IgE and its receptor induces a signaling cascade that results in mast cell degranulation with release of mediators, including histamine, as well as preformed cytokines and proteases, and synthesis and secretion of additional cytokines as well as lipid mediators, such as platelet activator factor (PAF), leukotrienes and prostaglandins4. Passive immunization studies in which mice were sensitized by injecting an Ag-specific IgE antibody (Ab), followed by enteral or parenteral exposure to that Ag, support the importance of IgE and mast cells in Ag-induced shock5. Indeed, both passive and active immunization studies in which mice were challenged orally with the appropriate Ag have generally demonstrated that genetic or Ab elimination of IgE, mast cells, or the IgE-binding chain of FcεRI, FcεRIα, completely suppresses anaphylaxis development6-8. In contrast, studies in which mice were actively immunized with an Ag, followed by parenteral challenge with the same Ag, have often revealed that anaphylaxis can occur in the absence of the classical IgE/FcεRI/mast cell pathway and demonstrated that a disorder that closely resembles IgE-mediated systemic anaphylaxis can be mediated by mechanisms that involve IgG rather than IgE9-11. Consistent with this, mice that are passively immunized with an IgG1, IgG2a, or IgG2b (but not IgG3) monoclonal Ab (mAb) specific for the hapten trinitrophenyl (TNP) develop anaphylaxis, that is nearly indistinguishable clinically from IgE-mediated anaphylaxis, when challenged parenterally, but not enterally, with a TNP-protein conjugate5, 6, 8. These observations, coupled with several human clinical observations, suggest that IgE-independent anaphylaxis may be clinically important. Here, we will first review observations that prove the existence of IgG-mediated anaphylaxis in mice and describe differences in the mechanisms behind the classical, IgE-mediated pathway and the alternative IgG-mediated pathway in this species, as well as the clinical implications of these differences. Next, we will review observations that support the existence of IgG-mediated anaphylaxis in humans, as well as the implications and limitations of these observations. Finally, we will discuss the evidence and its limitations for other, Ab-independent mechanisms of anaphylaxis in both mice and humans.

Murine evidence for IgG-mediated anaphylaxis

Evidence for IgE-independent, IgG-dependent anaphylaxis was provided by studies in which mice were immunized, then parenterally challenged with a potent antigen11. In some of these active immunization models, disease developed even if mice were first treated with an anti-IgE monoclonal antibody, but was suppressed if mice were instead treated with a rat IgG2b monoclonal Ab (mAb), 2.4G2. This mAb binds to and triggers, but then blocks the inhibitory low affinity IgG receptor, FcγRIIB, and the stimulatory low affinity IgG receptor, FcγRIII, and indirectly blocks the other murine FcγRs: FcγRI and FcγRIV11, 12. The existence of IgE-independent anaphylaxis in actively immunized mice was demonstrated most conclusively by studies that: (1) induced severe anaphylaxis in actively immunized IgE- or FcεRIα-deficient mice, but not in actively immunized mice that lacked all stimulatory FcRs (i.e.; FcRγ-deficient mice); and (2) demonstrated reduced severity or absence of anaphylaxis in actively immunized mice that lacked function of one or more of the stimulatory murine FcγRs11-14.

Subsequent passive immunization studies demonstrated that an anti-IgE mAb would block anaphylaxis when mice were sensitized with an Ag-specific IgE mAb, but not when mice were sensitized with an Ag-specific IgG1, IgG2a, or IgG2b mAb, while reciprocal results were found when passively immunized mice were treated with 2.4G210-12, 15, 16. The severity of systemic anaphylaxis in these IgG passive immunization models was normal or increased in mice that that were deficient in FcεRIα13. In contrast, anaphylaxis in mice passively sensitized with an Ag-specific IgG1 mAb was totally absent in mice deficient in FcγRIII (the only stimulatory murine FcγR that binds mouse IgG1), while total suppression of anaphylaxis in mice sensitized with an IgG2a mAb (which binds to all three stimulatory murine FcγRs) required the deletion or blocking of all of these receptors10, 12. The importance of FcγRs in murine IgG-dependent anaphylaxis was also shown by the unique inability of IgG3, among the murine IgG isotypes, to mediate anaphylaxis, which correlates with the observation that IgG3 is the only murine IgG isotype that fails to bind to any stimulatory murine FcγR17, 18.

Clinical implications of studies of murine IgG-mediated anaphylaxis

Studies of murine IgG-mediated anaphylaxis by several groups have evaluated the mediators involved, the responsible cell types, and the quantities of Ag required to induce shock. Nearly all studies have identified PAF, rather than histamine, as the mediator most important in IgG-mediated anaphylaxis in actively immunized mice11, 19, 20, although this has not been investigated thoroughly in passively immunized mice. In contrast to agreement about the importance of PAF in IgG-mediated anaphylaxis in actively immunized mice, different studies have identified monocyte/macrophages, basophils, or neutrophils as the critical cell type in IgG-mediated anaphylaxis11, 19, 20. All of these cell types express FcγRIII and FcγRIV in mice and all are capable of producing PAF in response to appropriate stimuli18, 20-24. Differences in cell types that appear to be responsible for IgG-mediated anaphylaxis may result from differences in mouse strains used, stimuli that elicit anaphylaxis, endogenous bacterial flora and/or animal husbandry practices.

Results of studies that compared the doses of antigen required to induce IgE- vs. IgG1-mediated anaphylaxis suggest that the dose of challenge Ag determines when IgG-mediated anaphylaxis can occur. In mice that were passively sensitized with high affinity IgE or IgG Abs to TNP, 100- to 1,000-fold less TNP-conjugated protein was required to induce shock in IgE- than in IgG-sensitized mice15. This was true regardless of the extent of TNP-labeling of the TNP-conjugated protein, although less TNP-conjugate was required to induce either IgE- or IgG-mediated anaphylaxis when the protein was heavily labeled15. These observations are consistent, respectively, with the much higher affinity of FcεRI than FcγRIII, the much higher ratio of cell-bound to serum IgE than IgG, and the better crosslinking of an Ag-specific mAb by an Ag that has multiple copies of the epitope bound by that mAb.

Because IgG-mediated anaphylaxis requires a much larger dose of Ag than IgE-mediated anaphylaxis, anaphylaxis induced by parenteral administration of a small quantity of Ag (e.g.; insect sting) is much more likely to be IgE-mediated. Anaphylaxis induced by Ag ingestion (e.g.; food allergy), similarly, always appears to be IgE-mediated6, 7, because induction of anaphylaxis in food allergy models requires systemic absorption of ingested Ag and only a very small percentage of ingested Ag is absorbed with all epitopes intact7, 25. In contrast, both IgE- and IgG-mediated anaphylaxis can be induced by parenteral administration of a relatively large quantity of Ag15 (e.g.; infusion of a therapeutic Ab or drug), particularly an Ag that has multiple iterations of an antibody-reactive epitope (e.g.; a carbohydrate Ag, such as dextran).

The difference in Ag dose requirement for IgE vs. IgG-mediated anaphylaxis allows IgG to act both as a mediator of anaphylaxis and a blocker of IgE-mediated anaphylaxis, depending on Ab and Ag concentrations (Figure 1). In the presence of Ag-specific IgE, Ag-specific IgG Ab will block anaphylaxis that would otherwise be induced by a low dose of Ag by intercepting Ag before it can bind to mast cell-associated IgE and by interacting with the inhibitory receptor, FcγRIIB15, 26 (Figure 1A and B), but mediate anaphylaxis induced by a higher Ag dose (Figure 1C and D). The ability of IgG to both block Ag access to mast cell-associated IgE and to mediate anaphylaxis through IgG/Ag complex binding to stimulatory FcγRs can create the counter-intuitive situation where an intermediate dose of Ag will induce IgG-, but not IgE-mediated anaphylaxis in the presence of both Ag-specific IgE and IgG15 (Figure 1C).

Figure 1. Relative concentrations of Ag and Ab determine the roles of IgE and IgG Abs in Ab-mediated anaphylaxis.

Figure 1

IgE-mediated anaphylaxis requires considerably less Ab and Ag than IgG-mediated anaphylaxis. Consequently, when Ab levels are low (A), only IgE-mediated anaphylaxis can occur. When Ag levels are low but Ab levels are high (B), IgG “blocking” Abs prevent IgE-mediated anaphylaxis by intercepting Ag before it can bind to FcεRI-associated IgE and by binding to the inhibitory receptor, FcγRIIB, but the quantity of IgG/Ag complexes is too low to trigger IgG-mediated anaphylaxis. Consequently, anaphylaxis does not occur. When Ag and Ab levels are both high, but Ab is in excess to Ag (C), IgG Abs block the binding of Ag to FcεRI-bound IgE but IgG/Ag complexes can bind to FcγRs; consequently, only IgG-mediated anaphylaxis occurs. When Ag and Ab levels are both high, but Ag is in excess (D), IgG/Ab complexes are sufficient to trigger IgG-mediated anaphylaxis and enough Ag escapes IgG blockade to bind to FcεRI-associated IgE and trigger IgE-mediated anaphylaxis.

Limitations of studies of mouse IgG-mediated anaphylaxis

Although experimental evidence that IgG-mediated anaphylaxis can occur in mice is unequivocal, there are concerns about the interpretation of studies that identify the importance of different cells and receptors in this process. Nearly all studies that analyze the importance of specific receptors use gene deletion to eliminate specific FcγRs or Abs to block these receptors, while studies that analyze the importance of specific cell types either use Abs or drugs that eliminate these cell types or transfer a specific cell type to a recipient mouse. Although gene deletion can cause complete deficiency of a specific receptor, the elimination of one receptor under at least some circumstances can increase the expression and/or signaling capacity of the remaining receptors15, 24. This may lead investigators to exaggerate the importance of the remaining receptors. IgG mAbs to a specific receptor may influence non-targeted receptors by signaling through the targeted receptor and/or binding of the Fc part of the IgG mAb to non-targeted receptors (e.g.; mAb 2.4G2, which binds to FcγRIIB and FcγRIII, at least partially suppresses the expression and function of FcγRI and FcγRIV12). Techniques used to delete macrophages, such as silica, clodronate liposomes, and gadolinium, may activate these cells before killing them27; cytokines or mediators produced by the activated cells may influence the ability of remaining cell types to contribute to anaphylaxis. Antibodies used to delete specific cell types, including neutrophils, basophils and platelets, may activate and deplete complement upon binding the targeted cell type or form immune complexes with cell membrane antigens that bind to FcγRs on other cells; both may influence the ability of these other cells to contribute to anaphylaxis. In this regard, for example, we have found that IgG-mediated anaphylaxis is suppressed when platelets are eliminated with IgG anti-platelet antibodies, but not when platelets are eliminated with neuraminidase11. Abs that appear to eliminate a cell type, when that cell type is studied in one organ (e.g.; blood), may actually cause redistribution of that cell type to another organ (e.g.; spleen). Finally, because cells, such as neutrophils, may be partially activated by in vitro purification procedures, it is possible that transfer studies with purified neutrophils19 may exaggerate the importance of this cell type in IgG-mediated anaphylaxis. Although none of these techniques are used to evaluate whether IgG-mediated anaphylaxis exists in humans, concerns about their use in mice affects hypotheses about which receptors and cell types might contribute to putative human IgG-mediated anaphylaxis.

Theoretical considerations about the possibility of human IgG-mediated anaphylaxis

Because the kinds of experiments that have proven the existence of IgG-mediated anaphylaxis in mice would not be appropriate in humans, even when possible, evidence for human IgE-mediated anaphylaxis is typically anecdotal and correlative, rather than definitive. In addition, important differences between mice and humans, including differences in the properties of their IgG isotypes, differences in their FcγRs, differences in cellular FcγR distribution, and differences in the properties of FcγR-expressing cells themselves raise questions about the applicability of observations about IgG-mediated anaphylaxis in mice to humans. However, consideration of each of these differences fails to provide a reason for thinking that human IgG-mediated anaphylaxis is unlikely.

Human IgG1 and IgG3, and possibly IgG4, bind to human FcγRs with an affinity range similar to what is observed in mice24. Although humans lack FcγRIV, they express FcγRI and FcγRIII and have stimulatory FcγRs, FcγRIIA and FcγRIIC, which are not present in mice24. Activation of human basophils, monocyte/macrophages and neutrophils can cause these cells to produce PAF, which has been associated with human anaphylaxis28, 29. In addition, as noted earlier, human neutrophils can mediate IgG-dependent anaphylaxis when infused into mice19.

Differences in cellular FcγR expression may actually make it more likely for IgG to mediate anaphylaxis in humans than in mice, because while both human and mouse mast cells express a stimulatory FcγR (FcγRIII in mice, FcγRIIA and possibly, FcγRIIC in humans), human mast cells express relatively little or no inhibitory FcγRIIB, while mouse mast cells express relatively large amounts of this receptor24, 30, 31. Similarly, the much larger number of granules in human than in murine basophils32 makes degranulation of these cells more likely to induce anaphylaxis in the former species. Taken together, there is no reason to believe that differences in IgG isotypes, FcγRs, cellular distribution of these receptors, or the physiology of FcγR-expressing cells make IgG less likely to mediate anaphylaxis in people than in mice.

Clinical evidence for human IgG-mediated anaphylaxis

Several clinical observations support the importance of IgG-mediated anaphylaxis in humans, although each of these observations is open to alternative conclusions. Multiple researchers have described anaphylaxis in patients who were treated with a biologic therapeutic and had IgG, but not detectable IgE Ab that was specific for that therapeutic33. This has been reported in transfused and intravenous immunoglobulin-treated IgA-deficient individuals (who had developed IgG anti-IgA Abs)34; individuals treated with a variety of chimeric, humanized and even fully human mAbs35; individuals treated with dextran36 or aprotinin37; and von Willebrand factor-deficient individuals who have been infused with von Willebrand factor38. It is noteworthy that all of these examples of putative IgG-mediated anaphylaxis involve the parenteral administration of a large quantity of an Ag – precisely the condition that favors IgG-mediated anaphylaxis in mice. It remains possible, however, that the individuals who developed anaphylaxis may have had cell-bound, FcεRI-associated therapeutic-specific IgE without detectable Ag-specific IgE in serum. This is possible because the high affinity of FcεRI for IgE and the relatively low level of FcεRI crosslinking required to induce mast cell degranulation allow sufficient IgE to bind to mast cells to mediate their activation, even when serum IgE levels are very low. Similarly, other evidence that supports the existence of IgE-independent anaphylaxis, such as anaphylaxis without evidence of basophil activation, anaphylaxis in the absence of increased serum tryptase39, and anaphylaxis in skin test-negative individuals might be explained by a lack of sensitivity of the tests used, the small time window in which a test reflective of IgE-mediated anaphylaxis remains positive, or by restrictions in the location and properties of mast cells responsible for IgE-mediated anaphylaxis (e.g.; Ag-specific IgE might be bound to vascular mast cells, but not to skin mast cells).

Additional evidence in favor of IgG-mediated anaphylaxis comes from a study of individuals treated with the chimeric mAb infliximab, which demonstrated that the presence of IgG anti-infliximab Ab levels ≥ 8 μg/ml was associated with a relative risk of anaphylaxis of 2.440, 41. Although this association suggests that IgG may have been involved in anaphylaxis pathogenesis, it is also possible that higher IgG antibody levels were a marker for higher IgE Ab levels.

Because PAF is more strongly associated with IgG- than with IgE-mediated anaphylaxis in mice, reports that serum PAF levels are higher in patients undergoing anaphylaxis than in a control group of patients and that serum concentrations of PAF acetylhydrolase, the enzyme that breaks down PAF, correlate inversely with anaphylaxis severity28, 29, are also consistent with the existence of human IgG-mediated anaphylaxis. However, because PAF can also be produced by mast cells and basophils in response to IgE crosslinking42, 43, and because human myeloid cells other than mast cells and basophils can express FcεRIα24, these observations may instead reflect a role for PAF in human IgE-mediated anaphylaxis. This alternative explanation is somewhat refuted by evidence that human neutrophils, monocyte/macrophages and basophils can produce PAF in response to FcγR crosslinking and that human neutrophils can mediate anaphylaxis in mice19; however, it could be argued that there is no direct evidence that the amount of PAF or other mediators produced by these cells is sufficient to induce anaphylaxis in humans.

One last, intriguing piece of evidence in favor of human IgG-mediated anaphylaxis comes from a study demonstrating increased frequency of a gain of function allele of the stimulatory FcγR, FcγRIIA, in common variable immunodeficiency patients who have IgG anti-IgA antibodies and develop anaphylaxis after IVIG infusion44. The impact of this elegant work, however, is limited by the small number of patients studied, the possibility that the mechanism that associates increased FcγRIIA activity with anaphylaxis may be indirect (e.g.; increased FcγRIIA function might promote an IgE response), and the lack of other reported associations of FcγR polymorphisms with human anaphylaxis.

Taken together, these observations appear to us to make the existence of human IgG-mediated anaphylaxis highly likely, particularly when anaphylaxis occurs in the presence of relatively high titers of specific IgG Ab and undetectable specific IgE in individuals who have been injected or infused with relatively large quantities of the recognized Ag. We concede, however, that these observations do not provide absolute proof of the existence or clinical importance of human IgG-mediated anaphylaxis.

Murine complement-mediated anaphylaxis

Studies in mice demonstrate that C3a and C5a, small peptides derived from C3 and C5, respectively and known as anaphylatoxins, can activate mast cells and other myeloid cells45; however, there is a lack of convincing evidence that they are either required for antibody-mediated anaphylaxis or can produce shock in the absence of other factors in this species. Passive anaphylaxis studies have demonstrated that ligation of mast cell C3aR or C5aR are required for the induction of skin swelling by injected C3a and C5a, respectively, and that both anaphylatoxins stimulate mast cell degranulation46. Studies in which mice were injected with soluble peanut extract demonstrated that components of that extract activated complement through both the classical and the lectin pathways47, 48. C3a produced in this manner could stimulate hypothermia through a macrophage and PAF-dependent process, but only when mice were treated with a β-adrenergic antagonist and/or a long-acting formulation of IL-4 to increase their responsiveness to PAF47. Perhaps more importantly, complement activation by peanut extract acted synergistically with IgE-mediated mast cell activation to cause shock in the absence of exogenous β-adrenergic antagonist or IL-447. This suggested that combined IgE-dependent mast cell activation and complement activation by peanuts might be one explanation for the severity of peanut-induced anaphylaxis. The suggestion that complement activation exacerbates anaphylaxis induced by other mechanisms, but is insufficient to independently induce murine systemic anaphylaxis, is also consistent with the inability of Ag-specific mouse IgG3, which efficiently activates complement, but does not bind to FcγRs, to sensitize mice to develop anaphylaxis following relevant Ag challenge. Murine studies may, however, underestimate the importance of complement-derived anaphylatoxins in human anaphylaxis, because complement components are less capable of inducing anaphylaxis in another rodent (the rat) than in some larger mammals, such as dogs and pigs49.

These observations suggest that Ab-mediated anaphylaxis should be less severe in the absence of C3 or anaphylatoxin receptors when anaphylaxis is mediated by a complement-activating isotype, such as mouse IgG2a. Even IgE-mediated anaphylaxis might be expected to be less severe in the absence of complement or anaphylatoxin receptors, if IgE-mediated mast cell activation results indirectly in anaphylatoxin production. However, it is also possible that decreased stimulation of the G-protein-dependent anaphylatoxin receptors increases the responsiveness of other G-protein-dependent receptors, such as the histamine receptors, that mediate anaphylaxis, (this would be analogous to the increased signaling through FcγRIII that is observed in the absence of FcεRIα50). Studies are required to evaluate these possibilities.

Complement-mediated anaphylaxis: Observations in humans

The potential for complement-mediated human anaphylaxis is suggested by studies showing expression of one or both anaphylatoxin receptors on human mast cells, basophils, other myeloid cells, and vascular endothelial cells51-54. A role for complement in human Ab-mediated anaphylaxis is suggested by a correlation between the severity of anaphylaxis and serum anaphylatoxin levels, although the risk associated with elevated anaphylatoxin levels is not as high as the risk associated with elevated tryptase or histamine levels55. Complement may have a particularly important anaphylaxis-enhancing role in vespid toxin-induced anaphylaxis, in which complement activation by proteases in vespid toxins is likely to exacerbate disease caused by IgE antibodies to vespid toxin antigens56. There is also considerable clinical evidence for the induction of anaphylaxis by agents that directly activate complement in the absence of agent-specific IgE or IgG Abs. This can be observed in association with hemodialysis (particularly during the first use of a new dialysis membrane); protamine neutralization of heparin, liposomal drug infusion; infusions of drugs that are dissolved or suspended in certain lipid vehicles, such as Cremophor EL, and polyethylene glycol infusion49. Two limitations of these correlative studies are that: 1) complement might not be the only factor activated that could contribute to shock; activation of the contact/kinin system is just one alternative possibility); and 2) no human studies have been performed to try to prevent anaphylaxis in any of these situations with inhibitors of complement activation or anaphylatoxin receptors. Taken together with the mouse data, the most likely interpretation of the clinical studies is that acute complement activation can induce anaphylaxis, particularly when other factors are present (e.g.; FcεRI or FcγR crosslinking, pre-existing vasculopathies) that can add to or synergize with anaphylatoxin effects.

Ab- and complement-independent anaphylaxis

Several drugs have been associated with anaphylaxis in susceptible individuals in the absence of a direct Ig-mediated mechanism or complement activation. These drugs include over-sulfated heparin, aspirin and non-steroidal anti-inflammatory drugs (NSAIDs), antibiotics, including vancomycin and the fluoroquinilones, opiates, and drugs used in general anesthesia, particularly neuromuscular blocking agents39, 57-59. Different mechanisms for anaphylaxis induction have been implicated for these different drugs in in vitro studies with human cells and plasma and in vivo animal studies. Over-sulfated heparin directly activates the kinin system, with increased production of bradykinin59. NSAIDs, including aspirin, block cyclooxygenases, which are essential for prostaglandin production. This results in decreased levels of prostaglandin E2, which can suppress anaphylaxis, and increased levels of cysteinyl leukotrienes, which, among other effects, increase pulmonary smooth muscle contraction and vascular permeability39. Aspirin, unlike other NSAIDs, has also been reported to increase FcεRI-mediated basophil activation by enhancing phosphorylation of the signaling molecule, Syk.58 Vancomycin activates mast cells to release histamine and other mediators through a mechanism that is calcium-, phospholipase C- and phospholipase A2-dependent, but is otherwise unknown60. Opiates also induce histamine release, through a mechanism that involves binding to central opioid receptors39. Fluroquinolone antibiotics and nicotinic receptor antagonist non-steroidal neuromuscular blocking agents, such as tubocurare, which have a tetrahydroisoquinalone motif, directly activate mast cells by binding to MRGPRX2, a G-protein-coupled receptor57. Taken together, these observations provide considerable reason to believe that the direct effects of these drugs on mast cells and basophils contribute to anaphylaxis. However, Ab/FcR interactions can also contribute to the ability of at least some of these drugs to induce anaphylaxis. With the exception that opioid-induced anaphylaxis has been reversed in humans by administration of opioid receptor antagonists39, 61, the importance of direct mast cell activation for anaphylaxis induction by these drugs has not been proven in humans in vivo. For anaphylaxis associated with other drugs, such as iodinated radiological contrast media, the relative roles of IgE Abs, IgG Abs, complement, and direct effects on myeloid cells are still debated39. It also remains to be determined whether any anaphylaxis-associated drugs cause disease solely through direct effects on mast cells or as part of a two- or multi-hit mechanism, and why some people are much more susceptible than others.

Conclusion

Although it is currently impossible to prove beyond a doubt that non-IgE-mediated anaphylaxis is clinically relevant, considerable evidence supports the occurrence and clinical importance of human IgE-independent anaphylaxis that is mediated by IgG, complement, or direct basophil and mast cell activation. Clinical situations that have been associated with the four different putative types of anaphylaxis are summarized in Table I, which also summarizes the cells, receptors and mediators that are thought to contribute to the pathogenesis of each form of this disorder. IgG-mediated anaphylaxis should be suspected when there are large infusions of Ag and high titers of IgG Ab specific for the infused Ag, while complement-mediated anaphylaxis and direct mast cell/basophil activation should be suspected in patients who have received drugs, biologicals, or excipients that are known to have the appropriate complement or mast cell/basophil-activating properties. Identifying the probable cause of a specific episode of anaphylaxis is likely to increase in importance as therapeutics that block a specific pathway become available; pretreatment with such therapeutics may also be useful as prophylaxis for patients who require specific anaphylaxis-associated drugs. Finally, it is important that a single episode of anaphylaxis may involve more than one mechanism. Simultaneous occurrence of IgG- and IgE-mediated anaphylaxis has been demonstrated in mice62, as has synergy between anaphylatoxin- and IgE-mediated anaphylaxis47; IgG/Ag complexes that bind to stimulatory FcγRs may also initiate anaphylaxis by activating complement, and direct mast cell activation by drugs is likely to act additively or synergistically with Ab- or complement-dependent activation of these and other myeloid cells to increase anaphylaxis severity.

Table I.

Etiologic mechanisms of anaphylaxis and their distinguishing characteristics

Type Inciting Agents Cells Receptors Mediators
IgE-Mediated Food allergy
Insect sting allergy
Drug allergy
Mast cells
Basophils
FcεRI Histamine
PAF
IgG-Mediated Biologicals
Drugs
Dextrans
Aprotinin
Transfusions
Macrophages
Neutrophils
Basophils
FcγRIII
FcγRI
FcγRIV (mouse)
FcγRIIA (human)
PAF
Histamine
Complement-Mediated Lipid incipients
Micellar drugs
Liposome
Other nanoparticles
Polyethylene glycol
Cellulose membranes
Macrophages
Mast cells
C3aR
C5aR
PAF
Histamine
Direct Mast Cell Activation
(mechanisms differ for
 different agents)
NSAIDs, including aspirin
Vancomycin
Opiates
Local Anesthetics
Fluoroquinolone antibiotics
Neuromuscular blockers
Octreotide
Leuprolide
Mast cells
Other myeloid cells
MRGPRX2
Other receptors
Cysteinyl leukotrienes
Histamine

Acknowledgements

Some of the work mentioned in this article has been supported by the National Institutes of Health (R01AI113162 and R21AI103816), a Merit Award from the U. S. Department of Veterans Affairs, the U. S. Department of Defense (PR120718), and Food Allergy Research and Education, Inc.

Abbreviations

Ab

antibody

Ag

antigen

FcεRI

high affinity IgE receptor

FcεRIα

α polypeptide chain of FcεRI

FcγR

IgG receptor

mAb

monoclonal antibody

PAF

platelet activating factor

TNP

trinitrophenyl

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 citable 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.

References

  • 1.Bochner BS, Lichtenstein LM. Anaphylaxis. N Engl J Med. 1991;324:1785–90. doi: 10.1056/NEJM199106203242506. [DOI] [PubMed] [Google Scholar]
  • 2.Decker WW, Campbell RL, Manivannan V, Luke A, St Sauver JL, Weaver A, et al. The etiology and incidence of anaphylaxis in Rochester, Minnesota: a report from the Rochester Epidemiology Project. J Allergy Clin Immunol. 2008;122:1161–5. doi: 10.1016/j.jaci.2008.09.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ishizaka T, Ishizaka K, Tomioka H. Release of histamine and slow reacting substance of anaphylaxis (SRS-A) by IgE-anti-IgE reactions on monkey mast cells. J Immunol. 1972;108:513–20. [PubMed] [Google Scholar]
  • 4.Schwartz LB. Effector cells of anaphylaxis: mast cells and basophils. Novartis Found Symp. 2004;257:65–74. discussion −9, 98-100, 276-85. [PubMed] [Google Scholar]
  • 5.Hirayama N, Hirano T, Kohler G, Kurata A, Okumura K, Ovary Z. Biological activities of antitrinitrophenyl and antidinitrophenyl mouse monoclonal antibodies. Proc Natl Acad Sci U S A. 1982;79:613–5. doi: 10.1073/pnas.79.2.613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Brandt EB, Strait RT, Hershko D, Wang Q, Muntel EE, Scribner TA, et al. Mast cells are required for experimental oral allergen-induced diarrhea. J Clin Invest. 2003;112:1666–77. doi: 10.1172/JCI19785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kucuk ZY, Strait R, Khodoun M, Mahler A, Hogan S, Finkelman FD. Induction and suppression of allergic diarrhea and systemic anaphylaxis in a mouse model of food allergy. J Allergy Clin Immunol. 2012 doi: 10.1016/j.jaci.2012.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Strait R, A M, S H, Khodoun M, A S, FD F. Ingested allergens must be absorbed systemically to induce systemic anaphylaxis. J Allergy Clin Immunol. 2011;127:982–9. doi: 10.1016/j.jaci.2011.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Takeishi T, Martin TR, Katona IM, Finkelman FD, Galli SJ. Differences in the expression of the cardiopulmonary alterations associated with anti-immunoglobulin E-induced or active anaphylaxis in mast cell-deficient and normal mice. Mast cells are not required for the cardiopulmonary changes associated with certain fatal anaphylactic responses. J Clin Invest. 1991;88:598–608. doi: 10.1172/JCI115344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Miyajima I, Dombrowicz D, Martin TR, Ravetch JV, Kinet JP, Galli SJ. Systemic anaphylaxis in the mouse can be mediated largely through IgG1 and Fc gammaRIII. Assessment of the cardiopulmonary changes, mast cell degranulation, and death associated with active or IgE- or IgG1- dependent passive anaphylaxis. J Clin Invest. 1997;99:901–14. doi: 10.1172/JCI119255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Strait RT, Morris SC, Yang M, Qu XW, Finkelman FD. Pathways of anaphylaxis in the mouse. J Allergy Clin Immunol. 2002;109:658–68. doi: 10.1067/mai.2002.123302. [DOI] [PubMed] [Google Scholar]
  • 12.Khodoun MV, Kucuk ZY, Strait RT, Krishnamurthy D, Janek K, Clay CD, et al. Rapid desensitization of mice with anti-FcγRIIb/FcγRIII mAb safely prevents IgG-mediated anaphylaxis. J Allergy Clin Immunol. 2013;132:1375–87. doi: 10.1016/j.jaci.2013.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Dombrowicz D, Flamand V, Miyajima I, Ravetch JV, Galli SJ, Kinet JP. Absence of FcεRIα chain results in upregulation of FcγRIII-dependent mast cell degranulation and anaphylaxis. Evidence of competition between FcεRI and FcγRIII for limiting amounts of FcR β and γ chains. J Clin Invest. 1997;99:915–25. doi: 10.1172/JCI119256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Oettgen HC, Martin TR, Wynshaw-Boris A, Deng C, Drazen JM, Leder P. Active anaphylaxis in IgE-deficient mice. Nature. 1994;370:367–70. doi: 10.1038/370367a0. [DOI] [PubMed] [Google Scholar]
  • 15.Strait RT, Morris SC, Finkelman FD. IgG-blocking antibodies inhibit IgE-mediated anaphylaxis in vivo through both antigen interception and FcγRIIb cross-linking. J Clin Invest. 2006;116:833–41. doi: 10.1172/JCI25575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Khodoun M, Kucuk ZY, Strait RT, Krishnamurthy D, Janek K, Lewkowich IP, et al. Rapid polyclonal desensitization with antibodies to IgE and FcεRIα. J Allergy Clin Immunol. 2013;133:1555–64. doi: 10.1016/j.jaci.2013.02.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Grey HM, Hirst JW, Cohn M. A new mouse immunoglobulin: IgG3. J Exp Med. 1971;133:289–304. doi: 10.1084/jem.133.2.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Nimmerjahn F, Bruhns P, Horiuchi K, Ravetch JV. FcγRIV: a novel FcR with distinct IgG subclass specificity. Immunity. 2005;23:41–51. doi: 10.1016/j.immuni.2005.05.010. [DOI] [PubMed] [Google Scholar]
  • 19.Jonsson F, Mancardi DA, Kita Y, Karasuyama H, Iannascoli B, Van Rooijen N, et al. Mouse and human neutrophils induce anaphylaxis. J Clin Invest. 2011;121:1484–96. doi: 10.1172/JCI45232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tsujimura Y, Obata K, Mukai K, Shindou H, Yoshida M, Nishikado H, et al. Basophils play a pivotal role in immunoglobulin-G-mediated but not immunoglobulin-E-mediated systemic anaphylaxis. Immunity. 2008;28:581–9. doi: 10.1016/j.immuni.2008.02.008. [DOI] [PubMed] [Google Scholar]
  • 21.Nimmerjahn F, Ravetch JV. Divergent Immunoglobulin G subclass activity through selective Fc receptor binding. Science. 2005;310:1510–2. doi: 10.1126/science.1118948. [DOI] [PubMed] [Google Scholar]
  • 22.Mencia-Huerta JM, Benveniste J. Platelet-activating factor and macrophages. I. Evidence for the release from rat and mouse peritoneal macrophages and not from mastocytes. Eur J Immunol. 1979;9:409–15. doi: 10.1002/eji.1830090512. [DOI] [PubMed] [Google Scholar]
  • 23.Sanchez-Crespo M, Alonso F, Egido J. Platelet-activating factor in anaphylaxis and phagocytosis. I. Release from human peripheral polymorphonuclears and monocytes during the stimulation by ionophore A23187 and phagocytosis but not from degranulating basophils. Immunology. 1980;40:645–55. [PMC free article] [PubMed] [Google Scholar]
  • 24.Bruhns P, Jonsson F. Mouse and human FcR effector functions. Immunol Rev. 2015;268:25–51. doi: 10.1111/imr.12350. [DOI] [PubMed] [Google Scholar]
  • 25.Strait RT, Mahler A, Hogan S, Khodoun M, Shibuya A, Finkelman FD. Ingested allergens must be absorbed systemically to induce systemic anaphylaxis. J Allergy Clin Immunol. 2011;127:982–9.e1. doi: 10.1016/j.jaci.2011.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Takai T, Ono M, Hikida M, Ohmori H, Ravetch JV. Augmented humoral and anaphylactic responses in FcγRII-deficient mice. Nature. 1996;379:346–9. doi: 10.1038/379346a0. [DOI] [PubMed] [Google Scholar]
  • 27.Schmidt-Lauber C, Bossaller L, Abujudeh HH, Vladimer GI, Christ A, Fitzgerald KA, et al. Gadolinium-based compounds induce NLRP3-dependent IL-1β production and peritoneal inflammation. Ann Rheum Dis. 2015;74:2062–9. doi: 10.1136/annrheumdis-2013-204900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Vadas P, Gold M, Perelman B, Liss GM, Lack G, Blyth T, et al. Platelet-activating factor, PAF acetylhydrolase, and severe anaphylaxis. N Engl J Med. 2008;358:28–35. doi: 10.1056/NEJMoa070030. [DOI] [PubMed] [Google Scholar]
  • 29.Vadas P, Perelman B, Liss G. Platelet-activating factor, histamine, and tryptase levels in human anaphylaxis. J Allergy Clin Immunol. 2012;131:144–9. doi: 10.1016/j.jaci.2012.08.016. [DOI] [PubMed] [Google Scholar]
  • 30.Ujike A, Ishikawa Y, Ono M, Yuasa T, Yoshino T, Fukumoto M, et al. Modulation of immunoglobulin (Ig)E-mediated systemic anaphylaxis by low-affinity Fc receptors for IgG. J Exp Med. 1999;189:1573–9. doi: 10.1084/jem.189.10.1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhao W, Kepley CL, Morel PA, Okumoto LM, Fukuoka Y, Schwartz LB. FcγRIIa, not FcγRIIb, is constitutively and functionally expressed on skin-derived human mast cells. J Immunol. 2006;177:694–701. doi: 10.4049/jimmunol.177.1.694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lee JJ, McGarry MP. When is a mouse basophil not a basophil? Blood. 2007;109:859–61. doi: 10.1182/blood-2006-06-027490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Cheifetz A, Smedley M, Martin S, Reiter M, Leone G, Mayer L, et al. The incidence and management of infusion reactions to infliximab: a large center experience. Am J Gastroenterol. 2003;98:1315–24. doi: 10.1111/j.1572-0241.2003.07457.x. [DOI] [PubMed] [Google Scholar]
  • 34.Schmidt AP, Taswell HF, Gleich GJ. Anaphylactic transfusion reactions associated with anti-IgA antibody. N Engl J Med. 1969;280:188–93. doi: 10.1056/NEJM196901232800404. [DOI] [PubMed] [Google Scholar]
  • 35.Steenholdt C, Svenson M, Bendtzen K, Thomsen OO, Brynskov J, Ainsworth MA. Acute and delayed hypersensitivity reactions to infliximab and adalimumab in a patient with Crohn's disease. J Crohns Colitis. 2012;6:108–11. doi: 10.1016/j.crohns.2011.08.001. [DOI] [PubMed] [Google Scholar]
  • 36.Hedin H, Richter W, Messmer K, Renck H, Ljungstrom KG, Laubenthal H. Incidence, pathomechanism and prevention of dextran-induced anaphylactoid / anaphylactic reactions in man. Dev Biol Stand. 1980;48:179–89. [PubMed] [Google Scholar]
  • 37.Umeda Y, Fukumoto Y, Miyauchi T, Imaizumi M, Shimabukuro K, Mori Y, et al. Anaphylactic shock related to aprotinin induced by anti-aprotinin immunoglobulin G antibody alone; report of a case. Kyobu Geka. 2007;60:69–71. [PubMed] [Google Scholar]
  • 38.Bergamaschini L, Mannucci PM, Federici AB, Coppola R, Guzzoni S, Agostoni A. Posttransfusion anaphylactic reactions in a patient with severe von Willebrand disease: role of complement and alloantibodies to von Willebrand factor. J Lab Clin Med. 1995;125:348–55. [PubMed] [Google Scholar]
  • 39.Farnam K, Chang C, Teuber S, Gershwin ME. Nonallergic drug hypersensitivity reactions. Int Arch Allergy Immunol. 2012;159:327–45. doi: 10.1159/000339690. [DOI] [PubMed] [Google Scholar]
  • 40.Miele E, Markowitz JE, Mamula P, Baldassano RN. Human antichimeric antibody in children and young adults with inflammatory bowel disease receiving infliximab. J Pediatr Gastroenterol Nutr. 2004;38:502–8. doi: 10.1097/00005176-200405000-00008. [DOI] [PubMed] [Google Scholar]
  • 41.Baert F, Noman M, Vermeire S, Van Assche G, G DH, Carbonez A, et al. Influence of immunogenicity on the long-term efficacy of infliximab in Crohn's disease. N Engl J Med. 2003;348:601–8. doi: 10.1056/NEJMoa020888. [DOI] [PubMed] [Google Scholar]
  • 42.Mencia-Huerta JM, Lewis RA, Razin E, Austen KF. Antigen-initiated release of platelet-activating factor (PAF-acether) from mouse bone marrow-derived mast cells sensitized with monoclonal IgE. J Immunol. 1983;131:2958–64. [PubMed] [Google Scholar]
  • 43.Schwartz LB. Mediators of human mast cells and human mast cell subsets. Ann Allergy. 1987;58:226–35. [PubMed] [Google Scholar]
  • 44.van der Heijden J, Geissler J, van Mirre E, van Deuren M, van der Meer JW, Salama A, et al. A novel splice variant of FcγRIIa: a risk factor for anaphylaxis in patients with hypogammaglobulinemia. J Allergy Clin Immunol. 2013;131:1408–16. e5. doi: 10.1016/j.jaci.2013.02.009. [DOI] [PubMed] [Google Scholar]
  • 45.Szebeni J. Complement activation-related pseudoallergy: a new class of drug-induced acute immune toxicity. Toxicology. 2005;216:106–21. doi: 10.1016/j.tox.2005.07.023. [DOI] [PubMed] [Google Scholar]
  • 46.Schafer B, Piliponsky AM, Oka T, Song CH, Gerard NP, Gerard C, et al. Mast cell anaphylatoxin receptor expression can enhance IgE-dependent skin inflammation in mice. J Allergy Clin Immunol. 2013;131:541–8. e1–9. doi: 10.1016/j.jaci.2012.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Khodoun M, Strait R, Orekov T, Hogan S, Karasuyama H, Herbert DR, et al. Peanuts can contribute to anaphylactic shock by activating complement. J Allergy Clin Immunol. 2009;123:342–51. doi: 10.1016/j.jaci.2008.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kodama T, Sekine H, Takahashi M, Iwaki D, Machida T, Kanno K, et al. Role of complement in a murine model of peanut-induced anaphylaxis. Immunobiology. 2013;218:844–50. doi: 10.1016/j.imbio.2012.10.003. [DOI] [PubMed] [Google Scholar]
  • 49.Szebeni J. Complement activation-related pseudoallergy: a stress reaction in blood triggered by nanomedicines and biologicals. Mol Immunol. 2014;61:163–73. doi: 10.1016/j.molimm.2014.06.038. [DOI] [PubMed] [Google Scholar]
  • 50.Miyajima I, Dombrowicz D, Martin TR, Ravetch JV, Kinet J-P, Galli SJ. Systemic anaphylaxis in the mouse can be mediated largely through IgG1 and FcγRIII. Assessment of the cardiopulmonary changes, mast cell degranulation, and death associated with active or IgE- or IgG1- dependent passive anaphylaxis. J Clin Invest. 1997;99:901–914. doi: 10.1172/JCI119255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Van Epps DE, Simpson SJ, Johnson R. Relationship of C5a receptor modulation to the functional responsiveness of human polymorphonuclear leukocytes to C5a. J Immunol. 1993;150:246–52. [PubMed] [Google Scholar]
  • 52.Van Epps DE, Simpson SJ, Chenoweth DE. C5a and formyl peptide receptor regulation on human monocytes. J Leukoc Biol. 1992;51:393–9. doi: 10.1002/jlb.51.4.393. [DOI] [PubMed] [Google Scholar]
  • 53.Füreder W, Agis H, Willheim M, Bankl HC, Maier U, Kishi K, Müller MR, Czerwenka K, Radaszkiewicz T, Butterfield JH, Klappacher GW, Sperr WR, Oppermann M, Lechner K, Valent P. Differential expression of complement receptors on human basophils and mast cells. Evidence for mast cell heterogeneity and CD88/C5aR expression on skin mast cells. J Immunol. 1995;155:3152–3160. [PubMed] [Google Scholar]
  • 54.Skeie JM, Fingert JH, Russell SR, Stone EM, Mullins RF. Complement component C5a activates ICAM-1 expression on human choroidal endothelial cells. Invest Ophthalmol Vis Sci. 2010;51:5336–42. doi: 10.1167/iovs.10-5322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Brown SG, Stone SF, Fatovich DM, Burrows SA, Holdgate A, Celenza A, et al. Anaphylaxis: clinical patterns, mediator release, and severity. J Allergy Clin Immunol. 2013;132:1141–9. e5. doi: 10.1016/j.jaci.2013.06.015. [DOI] [PubMed] [Google Scholar]
  • 56.van der Linden PW, Hack CE, Kerckhaert JA, Struyvenberg A, van der Zwan JC. Preliminary report: complement activation in wasp-sting anaphylaxis. Lancet. 1990;336:904–6. doi: 10.1016/0140-6736(90)92272-j. [DOI] [PubMed] [Google Scholar]
  • 57.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: 10.1038/nature14022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Matsuo H, Yokooji T, Morita H, Ooi M, Urata K, Ishii K, et al. Aspirin augments IgE-mediated histamine release from human peripheral basophils via Syk kinase activation. Allergol Int. 2013;62:503–11. doi: 10.2332/allergolint.13-OA-0536. [DOI] [PubMed] [Google Scholar]
  • 59.Kishimoto TK, Viswanathan K, Ganguly T, Elankumaran S, Smith S, Pelzer K, et al. Contaminated heparin associated with adverse clinical events and activation of the contact system. N Engl J Med. 2008;358:2457–67. doi: 10.1056/NEJMoa0803200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Veien M, Szlam F, Holden JT, Yamaguchi K, Denson DD, Levy JH. Mechanisms of nonimmunological histamine and tryptase release from human cutaneous mast cells. Anesthesiology. 2000;92:1074–81. doi: 10.1097/00000542-200004000-00026. [DOI] [PubMed] [Google Scholar]
  • 61.Mushlin N, Vahid B, Roberts R. Use of naloxone in hydromorphone-induced anaphylactoid reaction. South Med J. 2007;100:611. doi: 10.1097/SMJ.0b013e318030ef88. [DOI] [PubMed] [Google Scholar]
  • 62.Liu E, Moriyama H, Abiru N, Miao D, Yu L, Taylor RM, et al. Anti-peptide autoantibodies and fatal anaphylaxis in NOD mice in response to insulin self-peptides B:9-23 and B:13-23. J Clin Invest. 2002;110:1021–7. doi: 10.1172/JCI15488. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES