Next Article in Journal
Targeting Casein Kinase 1 (CK1) in Hematological Cancers
Next Article in Special Issue
Clinical Impact of Inherited and Acquired Genetic Variants in Mastocytosis
Previous Article in Journal
An Automated Functional Annotation Pipeline That Rapidly Prioritizes Clinically Relevant Genes for Autism Spectrum Disorder
Previous Article in Special Issue
Results from a Genome-Wide Association Study (GWAS) in Mastocytosis Reveal New Gene Polymorphisms Associated with WHO Subgroups
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diagnosis, Classification and Management of Mast Cell Activation Syndromes (MCAS) in the Era of Personalized Medicine

1
Department of Internal Medicine I, Division of Hematology and Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
2
Division of Allergy and Clinical Immunology, University of Michigan, Ann Arbor, MI 48106, USA
3
Department of Dermatology, Medical University of Gdansk, 80-211 Gdansk, Poland
4
Allergy Unit, Verona University Hospital, 37126 Verona, Italy
5
Division of Allergy, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
6
Department of Allergology, Medical University of Gdansk, 80-211 Gdansk, Poland
7
Department of Dermatology and Allergy Biederstein, Technical University of Munich, D-80802 Munich, Germany
8
Dermatological Allergology, Department of Dermatology and Allergy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
9
Division of Allergy and Clinical Immunology, University of Salerno, 84131 Salerno, Italy
10
Department of Hematological Biology, Pitié-Salpêtrière Hospital, Pierre et Marie Curie University (UPMC), 75005 Paris, France
11
Department of Internal Medicine, Division of Rheumatology, Allergy & Immunology, Virginia Commonwealth University, Richmond, VA 23284, USA
12
Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, MD 20852, USA
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(23), 9030; https://doi.org/10.3390/ijms21239030
Submission received: 26 October 2020 / Revised: 16 November 2020 / Accepted: 24 November 2020 / Published: 27 November 2020

Abstract

:
Mast cell activation (MCA) is seen in a variety of clinical contexts and pathologies, including IgE-dependent allergic inflammation, other immunologic and inflammatory reactions, primary mast cell (MC) disorders, and hereditary alpha tryptasemia (HAT). MCA-related symptoms range from mild to severe to life-threatening. The severity of MCA-related symptoms depends on a number of factors, including genetic predisposition, the number and releasability of MCs, organs affected, and the type and consequences of comorbid conditions. In severe systemic reactions, MCA is demonstrable by a substantial increase of basal serum tryptase levels above the individual’s baseline. When, in addition, the symptoms are recurrent, involve more than one organ system, and are responsive to therapy with MC-stabilizing or mediator-targeting drugs, the consensus criteria for the diagnosis of MCA syndrome (MCAS) are met. Based on the etiology of MCA, patients can further be classified as having i) primary MCAS where KIT-mutated, clonal, MCs are detected; ii) secondary MCAS where an underlying IgE-dependent allergy or other reactive MCA-triggering pathology is found; or iii) idiopathic MCAS, where neither a triggering reactive state nor KIT-mutated MCs are identified. Most severe MCA events occur in combined forms of MCAS, where KIT-mutated MCs, IgE-dependent allergies and sometimes HAT are detected. These patients may suffer from life-threatening anaphylaxis and are candidates for combined treatment with various types of drugs, including IgE-blocking antibodies, anti-mediator-type drugs and MC-targeting therapy. In conclusion, detailed knowledge about the etiology, underlying pathologies and co-morbidities is important to establish the diagnosis and develop an optimal management plan for MCAS, following the principles of personalized medicine.

1. Introduction

Mast cells (MCs) are tissue-fixed effector cells involved in the initiation and perpetuation of allergic inflammation, as well as in a number of other inflammatory states [1,2,3,4,5]. In common with blood basophils, MCs express high-affinity IgE-binding sites and produce both pro-inflammatory and vasoactive mediators, some of which are stored in their metachromatic secretory granules (Table 1) [1,2,3,4,5]. During a severe anaphylactic reaction, allergen-induced cross-linking of IgE-binding sites (FceRI) on MCs is followed by an explosive release of granular-associated mediators. In addition, activated MCs release newly formed cell membrane-derived (lipid-type) mediators of hypersensitivity reactions and cytokines into the extracellular space [1,2,3,4,5]. Blood basophils may also participate in allergic and other inflammatory reactions in the same way as MCs [1,3,6]. However, not all hypersensitivity reactions may involve both cell types, even if the reaction is systemic. In addition, some of the mediators involved in anaphylactic reactions are produced and released primarily by MCs, but not by basophils.
The ability of MCs and basophils to liberate mediators of anaphylaxis in the context of cell activation, also known as ‘releasability’, depends on several different factors, including the underlying (primary) disease/pathology, the numbers and type of activated surface receptors and signaling molecules, and the genetic pattern [7,8,9,10,11]. The severity of a hypersensitivity reaction is dependent on additional variables, such as the number of MCs (and sometimes also basophils) involved in the reaction; presence and type of triggering allergen; the amount and type of allergen-specific IgE; triggering cofactors, like exercise, acetylsalicylic acid or alcohol; presence of comorbidities; the local (tissue) microenvironment; and the cytokine and chemokine networks involved [1,2,3,4,12,13,14,15,16,17].
MC activation (MCA) occurs in a number of pathologic conditions. Acute MCA is commonly seen in allergic reactions and may be associated with clinical signs and symptoms of anaphylaxis [1,2,3,4,5,12,13,14,15,16,17]. Severe or even life-threatening MCA may occur when the burden of MCs is high or/and these cells are in a ‘hyperactivated’ state [15,16,17]. In such patients, a MCA syndrome (MCAS) may be diagnosed when the symptoms are recurrent and MCAS criteria are fulfilled [15,16,17,18,19,20,21,22,23]. Historically, clinical symptoms arising from MCA have primarily been studied in the context of allergic/atopic disorders [1,2,3,4,5]. More recently, however, MCA has also been studied in the context of other pathologic conditions, including systemic mastocytosis (SM) [2,4,5,12,13,14,15,16,17,18,19,20,21,22,23].
During the past 10 years, criteria for MCA and MCAS have been developed by an international (EU/US-based) consensus group consisting of experts in the fields of allergy, dermatology, hematology, pathology, and molecular medicine [18,19,20,21,22]. These MCAS criteria have been validated in various studies [24,25,26]. In addition, diagnostic algorithms and recommendations for clinical management and therapy of patients with MCAS have been published recently [23].
In the current article, these criteria and the classification of MCAS as well as management strategies are discussed in light of new developments in the field; the growing number of available clinical, serological and biochemical markers; and the emerging medical need to diagnose and manage these patients based on scientific evidence and according to the principles of personalized medicine.

2. Symptoms recorded in Patients with MCA

MCA-related symptoms range from mild headache or urticaria to severe or even life-threatening anaphylaxis. These symptoms are caused by a number of vasoactive and pro-inflammatory mediators released from MCs (or alternatively basophils) when these cells are activated by an allergen via IgE and IgE receptors, or by other triggers [1,2,3,4,5,12,13,14,15,16,17]. An overview of MC-derived mediators and their clinical impact are shown in Table 1.
As mentioned, the severity of MCA-related symptoms depends on a number of independent factors and correlates with the amount and type of mediators released from MCs during an MCA event. In general, the symptoms that can be provoked by MC mediators are manifold and the type of MCA can be divided into (i) local versus systemic, (ii) acute versus chronic, and (iii) mild versus severe (Table 2) [23].
Typical manifestations of immediate-type hypersensitivity (allergic) reactions consistent with the diagnosis of systemic MCA (anaphylaxis), include acute urticaria, flushing, pruritus, headache, abdominal cramping, diarrhea, respiratory symptoms, and vascular instability (hypotension) [20,21,22,23]. Although none of these symptoms are absolutely specific for MCA, many are typically found in these patients. Especially, when occurring together in one patient at the same time, these symptoms are suggestive of MCA, although basophil activation is considered to manifest with a similar spectrum of symptoms. The likelihood of MCA is even higher when multiple (two or more) of these symptoms are recorded and are responsive to drugs blocking MC mediator effects, mediator production, or mediator release in MCs. Responsiveness to such intervention has been proposed as a diagnostic criterion of MCAS [20,21,22]. This is particularly relevant when the symptoms are unusual or when the patient has another unrelated disease that may cause symptoms mimicking (or resembling) MCA. Another important consideration is that several different mediators may be involved in MCA-related symptoms [1,2,3,4,5,17,18,19,20,21,22]. Likewise, severe hypotension may be triggered by both histamine and prostaglandin D2 (PGD2) derived from activated MCs in the same patient (Table 1).
Whether MCAS may also present as a chronic disease in the absence of recognized systemic anaphylactic events is controversial. Non-specific symptoms such as headache, fatigue, nausea and sleep disturbance do not fulfill well-established criteria of MCA and therefore of MCAS. Nevertheless, these symptoms are clinically relevant and thus should be treated appropriately to provide symptomatic relief. Also, it is important to recognize that the differential diagnoses to be considered in such patients are broad, including neurologic, infectious and cardiac disorders [20,21,22,23].
Finally, MCA may occur as a local, non-systemic event, which may also represent a clinical challenge for the treating physician. MCAS criteria are not fulfilled in these patients (Table 2). Examples include MCA in the skin presenting as urticaria or MCA in the lungs presenting as bronchial asthma. Although the MCAS criteria are not fulfilled, the impact of local MCA in these conditions needs to be acknowledged, and specific therapy using anti-mediator type drugs or MC-stabilizing agents may be required. On the other hand, it often remains unclear whether MC are indeed involved and if yes, to what extent. In addition, the etiology of local MCA often remains unknown. Whether local MCA is triggered by various infectious or toxic reactions is certainly worth considering, but precise criteria are needed to prove such an association and will be important to develop. Such criteria could also lead to better therapeutic interventions.

3. Laboratory Assessments in Patients with Systemic MCA

MCA is typically accompanied by the release of preformed and newly produced vasoactive and pro-inflammatory mediators and other MC-dependent compounds [1,2,3,4,5]. In most patients with severe systemic reactions, increased levels of MC-derived mediators are measurable in biological fluids (serum, plasma, urine) [20,21,22,23,24,25,26]. Some of these mediators, such as tryptase or prostaglandin D2, are more specific for MCs [1,2,3,4]. Other mediators are less specific and also produced by other cell types. Whereas histamine is released from MCs and basophils in similar quantities, tryptase is considered rather specific for MCs, although basophils can also express and release small quantities of the enzyme [27,28,29,30]. Therefore, most experts agree that for daily clinical practice, a rapid increase in the serum tryptase level from the individual’s baseline is MCA-specific and thus a reliable diagnostic parameter [20,21,22,23,24,25,26,31,32]. If no pre-therapeutic baseline is available, the baseline serum tryptase level has to be assessed after complete recovery (at least 24–48 h after complete resolution of symptoms) or in a symptom-free interval [20,21,22,23].
Apart from tryptase, other mediators, when increasing transiently over baseline during a clinically defined attack (MCA-related event), may also serve as supportive evidence of systemic MCA. These include, among others, histamine (plasma, urine), histamine metabolites (urine) and prostaglandin D2 metabolites (Table 3) [31,32,33,34,35,36,37].
However, these mediators as noted are less specific for MCA when compared to tryptase. Moreover, no threshold criteria have been proposed and validated to define what minimal increase of these mediators would count as a reliable indicator of systemic MCA (Table 3). Nevertheless, some of these mediators, such as histamine metabolites or prostaglandin D2 metabolites, may be helpful in the evaluation and ultimate diagnosis of MCA and should therefore be considered, especially when the serum tryptase assay is not available or when results are equivocal (Table 3). Other mediators are under preclinical or clinical evaluation. One of these mediators is diamine oxidase (DAO) (Table 3) [38]. Indeed, DAO levels and tryptase levels appear to increase in parallel during an MCA event [38]. It does remain unclear, however, whether DAO is indeed produced by MCs or by other cell types involved in these patients. Alternative mediators that have been discussed in the past (or more recently) may not qualify as robust indicators of MCA (Table 3).

4. Biochemical Indication of Substantial Systemic MCA

The normal (physiologic) serum tryptase level determined in healthy adults ranges between 0 and 11.4 ng/mL. Note that tryptase genotyping should be considered in any individual with a tryptase level ≥8 ng/mL to evaluate for HAT which, if present, will affect baseline tryptase levels and is further discussed below under “Impact of Genetic Predisposition in MCA and MCAS”. During a systemic MCA event that involves multiple organ systems and leads to the clinical symptoms of anaphylaxis, a substantial, event-related increase in tryptase over the individual’s baseline is typically found [20,21,22,23,31,32]. Such an increase in tryptase is measurable in patients with MCAS regardless of the underlying etiology or serum basal tryptase (sBT) level. Therefore, a diagnostic increase in tryptase is the most important criterion of MCAS. Because of the different levels in basal tryptase in these patients, a generally applicable equation has been developed by the consensus group [21,22,23]. By this consensus, a minimal increase of tryptase to plus 20% of baseline plus absolute 2 ng/mL (=sBT × 1.2 + 2), meets the definition of a substantial systemic MCA and is thus regarded as biochemical criterion of MCAS [21,22,23]. For example: the baseline tryptase level (sBT) is 10 ng/mL: an increase to 20 ng/mL is suggestive of MCA [1.2 × 10 + 2(absolute) = 14 ng/mL)], qualifying this as a MCA event (biochemical evidence of MCAS). The consensus equation works in all patient groups regardless of the baseline serum tryptase level (normal, slightly elevated or high), has been validated in several studies, and is regarded an acceptable standard in the diagnosis of MCAS (Table 3) [20,21,22,23,24,25,26].
As mentioned before, an increase in other MC-related biochemical parameters may also indicate systemic MCA. For example, a substantial increase in histamine metabolites or prostaglandin D2 metabolites should be regarded as indication of MCA [34,35,36,37]. Currently, a discussion is ongoing to define the minimal diagnostic threshold levels of these markers to qualify as MCAS criterion.

5. Diagnostic Consensus Criteria of MCAS

Minimal diagnostic criteria of MCAS include (i) typical clinical symptoms indicative of multi-organ involvement, usually meeting criteria for anaphylaxis; (ii) an increase in the serum tryptase level above the individual’s baseline serum tryptase (sBT), meeting the 20% + 2 threshold (= sBT × 1.2 + 2) and/or a clear increase in another MC mediator in biological fluids; and (iii) response of the symptoms to drugs targeting MC activation, mediator release from MCs and/or MC mediator effects (Supplemental Table S1). All three criteria must be fulfilled to arrive at the diagnosis MCAS by consensus criteria [20,21,22,23,39]. Prior to the diagnosis of MCAS, the physician may have established the initial diagnosis as ‘anaphylaxis’. However, a solid confirmation of the involvement and impact of the MC lineage is needed to establish the final diagnosis of MCAS (Figure 1).
In a first step, it is important to document mast cell (MC) lineage involvement and to confirm that MCAS criteria are fulfilled. The most important biomarker is tryptase, a portion of which is stored in secretory granules of MCs. During severe systemic MC activation (MCA), tryptase increases in biological fluids, including serum or plasma. When the diagnosis MCAS is established, it is important to ask whether the patient is suffering from a known primary disorder affecting MCs (clonality due to a somatic KIT gain-of-function mutation), secondary MCAS whereby MCs manifest an excessive response to an IgE-dependent allergen or to other IgE-independent agonists, and idiopathic MCAS where no underlying etiology can be documented. In some patients with mastocytosis, a combined form of MCAS (= primary and secondary MCAS) is diagnosed. These patients are at a very high risk to develop severe, life-threatening anaphylaxis. Abbreviations: MCAS, mast cell activation syndrome; MCs, mast cells.
When there is clear indication of MC involvement but the formal criteria of MCAS are not met, the condition may be considered to show MCA or even a (mild or chronic) MC activation disorder. However, such ‘sub-MCAS’ conditions are difficult to diagnose and difficult to separate from unrelated conditions in the differential diagnoses, as no criteria for these pathologies exist. The term ‘syndrome’ and thus MCAS should be reserved for patients who demonstrably suffer from significant MCA-related events meeting MCAS criteria.
There are also conditions that predispose individuals to MCA events and thus to MCAS, including hereditary alpha tryptasemia (HAT) and/or systemic mastocytosis (SM), which also can result in more severe IgE-dependent reactions, particularly to insect venoms (Supplemental Table S2) [21,22,23,40,41,42,43,44,45,46,47,48]. In SM, these patients may be labeled with the appendix SY (symptomatic) when they require continuous interventional treatment with MC stabilizers or mediator-targeting drugs, even if no MCAS is diagnosed [21]. For example, a patient with indolent SM (ISM) and repeated anaphylaxis after hymenoptera stings (but with only a slight increase in tryptase levels) should be labeled as ISMSY although MCAS criteria are not met.

6. Differential Diagnoses and Robustness of the Consensus Criteria

A number of medical conditions and disorders can mimic MCAS clinically [20,21,22,23]. These include, among others, infectious diseases, autoimmune disease, cardiac disorders, endocrinologic diseases, neurologic diseases, psychiatric disorders, and intoxications (Table 4).
The key problem is that the symptoms and symptom-patterns in MCAS and in these conditions are often overlapping and that it is often difficult to confirm MC involvement, especially when no MC-specific biochemical markers are available. Table 4 shows a compilation of specific medical conditions that may be confused with MCAS based on overlapping symptoms when only clinical criteria were applied. Therefore, it is of great importance to apply robust, MCA-indicative criteria and to base the diagnosis MCAS on solid diagnostic assays.
A particular problem is that the serum tryptase test is not always available during or after a systemic attack. In these cases, the diagnosis MCAS cannot be established, but the patients may be diagnosed as having had an episode of anaphylaxis, and only subsequent studies may or may not lead to the final diagnosis of MCAS (Figure 1).
Another specific problem is the co-incidence of MCAS with other underlying conditions that could mimic MCAS, for example a severe cardiac disease where hypotension and syncope have been recorded (e.g., aortic valve stenosis plus MCAS). Based on this (hypothetical) possibility, the criteria of MCAS should not include an ‘exclusive’ element. Rather, such patients have to be managed by an interdisciplinary approach, and only a close collaboration with specialists such as cardiologists and experts in MCAS/allergy will be able to establish an optimal treatment plan.
All in all, we opine that the optimal approach is i) to base MC involvement on a solid biochemical marker like tryptase and ii) to establish the diagnosis MCAS only in those patients in whom a substantial (event-related) increase in tryptase over the individual’s baseline (20% + 2 formula = 1.2 * sBT+2 = 1.2 × sBT + 2) has been documented [20,21,22,23]. This approach is most likely to confirm MCAS with high specificity, though the sensitivity will vary, depending on factors such as the clinical severity of the reaction, particularly with respect to hypotension, and the timing of acute sample collection, as sensitivity declines the longer is the time of collection after the peak, which usually occurs about 1 h after onset of clinical signs or symptoms, and post-peak levels decline with a half-life of about 2 h. Other diagnostic models in which only clinical parameters are employed with no robust biochemical makers integrated do not confirm MC involvement and thus are not suitable to differentiate between MCAS and MCAS-mimickers with certainty. This is a critical point, as patients may suffer from other relevant diseases that may need to be diagnosed and treated without delay.
During the preparation of this article, we reviewed other diagnostic criteria and models that have been proposed in the MCAS context in the recent past. In some of these proposals, the authors intertwine diagnostic MCAS criteria with diagnostic criteria of SM. It is worth noting in this regard that neoplastic MCs in SM are often silent clinically (without therapy) so that the patient does not suffer from MCA. In addition, although KIT is activated by certain mutations, MCs in SM have been reported to not exhibit increased releasability compared to normal MCs and the same holds true for ROSAKITD816V cells compared to ROSAKITWT cells [49]. Therefore, we are of the opinion that MC clonality and abnormal MC morphologies or phenotypes do not qualify as MCAS criteria.

7. Underlying Disorders and Classification of MCAS

MCAS can be divided into several different variants, depending on the underlying condition and pathology. In particular, patients with MCAS are grouped into primary (clonal = monoclonal) MCAS, secondary MCAS and idiopathic MCAS (Supplemental Table S3) [20,21,22,23]. Primary MCAS is defined by the presence of clonal MCs exhibiting a KIT-activating mutation in the KIT gene, usually KIT D816V. In most of these patients, an underlying cutaneous mastocytosis (CM) or SM is found. However, in a few of these patients, only the clonal MCs can be detected, whereas the full spectrum of criteria to diagnose SM or CM are not (yet) fulfilled [18,19,20,21,22,23]. In other patients, an underlying IgE-dependent allergy, another hypersensitivity disorder or another inflammatory condition associated with MCA may be detected [20,21,22,23,50]. These patients thus suffer from secondary MCAS. If no clonal MCs and no underlying reactive condition (explaining MCA) are found, the diagnosis is idiopathic MCAS and a search for etiology should continue [20,21,22,23]. Finally, as mentioned before, patients may suffer from more than one form of MCAS, for example when they have an IgE-dependent allergy as well as SM. In these patients, the likelihood of life-threatening anaphylaxis is high and the diagnosis MCAS can often be established in symptomatic cases [50]. These patients are often treated with combinations of drugs targeting MCs, MC mediators, and IgE [50,51].

8. Impact of Genetic Predisposition in MCA and MCAS

A number of genetic factors, such as germline mutations, polymorphisms, and gene replications are considered to predispose to more severe hypersensitivity reactions, atopic disorders and MCA. Examples are IL-4 and IL-13 gene polymorphisms leading to an excess production of IgE and thus predisposing to more extensive IgE-dependent allergies; or certain filaggrin gene (FLG) mutations predisposing to the development of atopic dermatitis [52,53,54,55,56].
A genetically determined increase in the number of MCs and/or production of MC mediators, such as tryptase, have also been discussed as predisposing factors. Recently, a familial form of hyper-tryptasemia has been described. This condition, termed hereditary alpha tryptasemia (HAT), is an autosomal dominant genetic trait caused by increased germline copies of the TPSAB1 gene encoding alpha-tryptase where baseline tryptase levels increase in a step-wise fashion with each additional TPSAB1 gene replication [46,47,57,58,59]. Individuals with HAT thus have elevated basal serum tryptase levels. Patients with HAT have been reported to variably present with complaints indicating multi-organ disease and MCA. More recent data suggest that HAT is a valid genetic biomarker that associates with more severe mediator-related symptoms and thus MCA in anaphylaxis, IgE-dependent allergies and mastocytosis [46,47,48,55,56,60,61].
Especially in patients who have HAT as well as a co-existing allergy and/or a co-existing clonal MC disorder (mastocytosis), the risk for development of MCAS may be greater [46,47,48,56,60,61]. Based on these studies, HAT can be regarded as a novel emerging genetic biomarker and modifying risk factor for MCA and MCAS in various patient cohorts, including SM and IgE-dependent allergies. It is also worth noting that HAT is more prevalent in SM than in healthy controls or other myeloid neoplasms [48].

9. Management of Patients with MCAS

Management of MCAS is based on both prophylactic and (active) therapeutic approaches. First, it is of considerable importance to advise patients and family members (e.g., parents of children) to avoid any agents or situations that might potentially provoke anaphylactic (allergic) reactions [15,16,17,18,19,20,21,22,23,44,50]. MCAS patients are also advised to take prophylactic anti-mediator therapy (e.g., histamine receptor blocker) for their entire life and to carry two (or more) epinephrine self-injectors after having been informed and instructed how to use these injectors in case of an emergency event. In patients with venom-induced (IgE-dependent) anaphylaxis, and thus secondary MCAS, immunotherapy (IT) is usually recommended, with recognition of potential side effects [17,44,50]. Cytoreductive agents are usually not prescribed for treatment of mediator-related events in MCAS. However, in those with a huge burden of MCs (e.g., patients with SSM or ASM/MCL), cytoreductive therapy might be considered. In fact, the frequency of life-threatening MCAS events may decrease substantially after reducing the mass of neoplastic MCs [50,51]. Aspirin has also been considered previously for therapy of anaphylaxis in patients with SM. However, the doses of aspirin necessary to block MC activation are high and cannot be tolerated by many patients. Other drugs include MC-stabilizing agents and glucocorticosteroids. A new emerging class of MC-targeting drugs are broadly acting tyrosine kinase inhibitors (TKI), such as midostaurin or avapritinib [62,63]. Some of these drugs, like midostaurin, block not only MC proliferation but also IgE-dependent (allergen-induced) activation of MCs [62,64]. Therefore, midostaurin may be a promising agent to suppress MCAS events in patients with primary MCAS and SM. Indeed, midostaurin rapidly improves mediator-induced symptoms and the quality of life in patients with advanced SM [65,66]. Another emerging strategy is to deplete specific IgE (e.g., omalizumab) in patients with secondary MCAS who have an underlying IgE-dependent allergy [67,68]. In patients with severe mixed MCAS (primary+secondary MCAS), multiple specific therapies may be required, following the principles of personalized medicine. For example, in patients with a high mass of neoplastic MC, advanced SM, and severe IgE-dependent allergies, combined therapy with a KIT-targeting drug or cladribine and omalizumab may be required to bring MCAS events under control [51].

10. Concluding Remarks and Future Perspectives

MCAS is an unusual MC-induced multi-organ condition characterized by clinical signs and symptoms of anaphylaxis or related pathologies. Because of the multiple mimickers of MCAS and a plethora of circulating proposals, it is important to diagnose MCAS using solid criteria through which MC involvement can be documented. In most cases, an event-related increase of the individual’s serum tryptase level over baseline is sufficient to confirm MC involvement. By contrast, the clinical symptoms of MCA are not specific and may be confused with symptoms of cardiac, infectious, neurologic, endocrinologic, or gastrointestinal disease. Once diagnosed, MCAS is classified into primary forms, secondary MCAS and idiopathic MCAS. Whereas, in some cases, initial therapy has to be applied before a definitive diagnosis of MCAS can be established it is important to be aware of this diagnosis and to establish it in the follow-up. The type of therapy varies in patients with MCAS, depending on the underlying disease, type of MCAS, and response to initial therapy. In severe forms and especially those with mixed MCAS, combination therapies are often recommended, including the use of anti-IgE antibodies, MC-targeted agents, and/or allergen-specific immunotherapies.

Supplementary Materials

The following are available online at https://www.mdpi.com/1422-0067/21/23/9030/s1.

Author Contributions

All co-authors contributed by drafting parts of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This project was supported by the Austrian Science Fund (FWF)—grants F4704-B20 and Polish Ministry of Science and Higher Education—grant no ST 02-0066/07/253. Metcalfe (DDM) and his laboratory is supported by the Division of Intramural Research, NIAID, NIH.

Conflicts of Interest

The authors declare that they have no conflict of interest in this study. LBS: VCU receives royalties from Thermo Fisher for the tryptase assay that are shared with its inventor, LBS.

References

  1. Schwartz, L.B. Mast cells and basophils. Clin. Allergy Immunol. 2002, 16, 3–42. [Google Scholar] [PubMed]
  2. Metcalfe, D.D. Mast cells and mastocytosis. Blood 2008, 112, 946–956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Galli, S.J.; Tsai, M. IgE and mast cells in allergic disease. Nat. Med. 2012, 18, 693–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Theoharides, T.C.; Valent, P.; Akin, C. Mast cells, mastocytosis, and related disorders. N. Engl. J. Med. 2015, 373, 1885–1886. [Google Scholar] [CrossRef]
  5. Valent, P.; Akin, C.; Hartmann, K.; Nilsson, G.; Reiter, A.; Hermine, O.; Sotlar, K.; Sperr, W.R.; Escribano, L.; George, T.I.; et al. Mast cells as a unique hematopoietic lineage and cell system: From Paul Ehrlich’s visions to precision medicine concepts. Theranostics 2020, 10, 10743–10768. [Google Scholar] [CrossRef] [PubMed]
  6. Gibbs, B.F. Human basophils as effectors and immunomodulators of allergic inflammation and innate immunity. Clin. Exp. Med. 2005, 5, 43–49. [Google Scholar] [CrossRef]
  7. Marone, G.; Spadaro, G.; Patella, V.; Genovese, A. The clinical relevance of basophil releasability. J. Allergy Clin. Immunol. 1994, 94, 1293–1303. [Google Scholar] [CrossRef]
  8. Kepley, C.L.; Youssef, L.; Andrews, R.P.; Wilson, B.S.; Oliver, J.M. Multiple defects in Fc epsilon RI signaling in Syk-deficient nonreleaser basophils and IL-3-induced recovery of Syk expression and secretion. J. Immunol. 2000, 165, 5913–5920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Macdonald, S.M.; Vonakis, B.M. Association of the Src homology 2 domain-containing inositol 5′ phosphatase (SHIP) to releasability in human basophils. Mol. Immunol. 2002, 38, 1323–1327. [Google Scholar] [CrossRef]
  10. Okayama, Y.; Kashiwakura, J.I.; Matsuda, A.; Sasaki-Sakamoto, T.; Nunomura, S.; Yokoi, N.; Ebihara, N.; Kuroda, K.; Ohmori, K.; Saito, H.; et al. The interaction between Lyn and FcεRIβ is indispensable for FcεRI-mediated human mast cell activation. Allergy 2012, 67, 1241–1249. [Google Scholar] [CrossRef] [PubMed]
  11. Havard, S.; Scola, A.M.; Kay, L.J.; Ishmael, S.S.; MacGlashan, D.W., Jr.; Peachell, P.T. Characterization of syk expression in human lung mast cells: Relationship with function. Clin. Exp. Allergy 2011, 41, 378–388. [Google Scholar] [CrossRef]
  12. Peavy, R.D.; Metcalfe, D.D. Understanding the mechanisms of anaphylaxis. Curr. Opin. Allergy Clin. Immunol. 2008, 8, 310–315. [Google Scholar] [CrossRef] [PubMed]
  13. Metcalfe, D.D.; Peavy, R.D.; Gilfillan, A.M. Mechanisms of mast cell signaling in anaphylaxis. J. Allergy Clin. Immunol. 2009, 124, 639–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kalesnikoff, J.; Galli, S.J. Anaphylaxis: Mechanisms of mast cell activation. Chem. Immunol. Allergy 2010, 95, 45–66. [Google Scholar] [CrossRef] [PubMed]
  15. Valent, P. Risk factors and management of severe life-threatening anaphylaxis in patients with clonal mast cell disorders. Clin. Exp. Allergy 2014, 44, 914–920. [Google Scholar] [CrossRef] [PubMed]
  16. Bonadonna, P.; Lombardo, C.; Zanotti, R. Mastocytosis and allergic diseases. J. Investig. Allergol. Clin. Immunol. 2014, 24, 288–297. [Google Scholar] [PubMed]
  17. Niedoszytko, M.; Bonadonna, P.; Elberink, J.N.G.O.; Golden, D.B.K. Epidemiology, diagnosis, and treatment of hymenoptera venom allergy in mastocytosis patients. Immunol. Allergy Clin. North Am. 2014, 34, 365–381. [Google Scholar] [CrossRef]
  18. Akin, C.; Scott, L.M.; Kocabas, C.N.; Kushnir-Sukhov, N.; Brittain, E.; Noel, P.; Metcalfe, D.D. Demonstration of an aberrant mast-cell population with clonal markers in a subset of patients with “idiopathic” anaphylaxis. Blood 2007, 110, 2331–2333. [Google Scholar] [CrossRef]
  19. Sonneck, K.; Florian, S.; Müllauer, L.; Wimazal, F.; Födinger, M.; Sperr, W.R.; Valent, P. Diagnostic and subdiagnostic accumulation of mast cells in the bone marrow of patients with anaphylaxis: Monoclonal mast cell activation syndrome. Int. Arch. Allergy Immunol. 2007, 142, 158–164. [Google Scholar] [CrossRef]
  20. Akin, C.; Valent, P.; Metcalfe, D.D. Mast cell activation syndrome: Proposed diagnostic criteria. J. Allergy Clin. Immunol. 2010, 126, 1099–1104. [Google Scholar] [CrossRef] [Green Version]
  21. Valent, P.; Akin, C.; Arock, M.; Brockow, K.; Butterfield, J.H.; Carter, M.C.; Castells, M.; Escribano, L.; Hartmann, K.; Lieberman, P.; et al. Definitions, criteria and global classification of mast cell disorders with special reference to mast cell activation syndromes: A consensus proposal. Int. Arch. Allergy Immunol. 2012, 157, 215–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Valent, P. Mast cell activation syndromes: Definition and classification. Allergy 2013, 68, 417–424. [Google Scholar] [CrossRef] [Green Version]
  23. Valent, P.; Akin, C.; Bonadonna, P.; Hartmann, K.; Brockow, K.; Niedoszytko, M.; Nedoszytko, B.; Siebenhaar, F.; Sperr, W.R.; Elberink, J.N.O.; et al. Proposed diagnostic algorithm for patients with suspected mast cell activation syndrome. J. Allergy Clin. Immunol. Pract. 2019, 7, 1125–1133.e1. [Google Scholar] [CrossRef] [PubMed]
  24. De Schryver, S.; Halbrich, M.; Clarke, A.; La Vieille, S.; Eisman, H.; Alizadehfar, R.; Joseph, L.; Morris, J.; Ben-Shoshan, M. Tryptase levels in children presenting with anaphylaxis: Temporal trends and associated factors. J. Allergy Clin. Immunol. 2016, 137, 1138–1142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Baretto, R.L.; Beck, S.; Heslegrave, J.; Melchior, C.; Mohamed, O.; Ekbote, A.; Huissoon, A.; Krishna, M.T. Validation of international consensus equation for acute serum total tryptase in mast cell activation: A perioperative perspective. Allergy 2017, 72, 2031–2034. [Google Scholar] [CrossRef] [PubMed]
  26. Passia, E.; Jandus, P. Using baseline and peak serum tryptase levels to diagnose anaphylaxis: A review. Clin. Rev. Allergy Immunol. 2020, 58, 366–376. [Google Scholar] [CrossRef] [PubMed]
  27. Xia, H.Z.; Kepley, C.L.; Sakai, K.; Chelliah, J.; Irani, A.M.; Schwartz, L.B. Quantitation of tryptase, chymase, Fc epsilon RI alpha, and Fc epsilon RI gamma mRNAs in human mast cells and basophils by competitive reverse transcription-polymerase chain reaction. J. Immunol. 1995, 154, 5472–5480. [Google Scholar] [PubMed]
  28. Castells, M.C.; Irani, A.M.; Schwartz, L.B. Evaluation of human peripheral blood leukocytes for mast cell tryptase. J. Immunol. 1987, 138, 2184–2189. [Google Scholar]
  29. Jogie-Brahim, S.; Min, H.K.; Fukuoka, Y.; Xia, H.Z.; Schwartz, L.B. Expression of alpha-tryptase and beta-tryptase by human basophils. J. Allergy Clin. Immunol. 2004, 113, 1086–1092. [Google Scholar] [CrossRef]
  30. Samorapoompichit, P.; Kiener, H.P.; Schernthaner, G.H.; Jordan, J.H.; Agis, H.; Wimazal, F.; Baghestanian, M.; Rezaie-Majd, A.; Sperr, W.R.; Lechner, K.; et al. Detection of tryptase in cytoplasmic granules of basophils in patients with chronic myeloid leukemia and other myeloid neoplasms. Blood 2001, 98, 2580–2583. [Google Scholar] [CrossRef] [Green Version]
  31. Schwartz, L.B.; Metcalfe, D.D.; Miller, J.S.; Earl, H.; Sullivan, T. Tryptase levels as an indicator of mast-cell activation in systemic anaphylaxis and mastocytosis. N. Engl. J. Med. 1987, 316, 1622–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Schwartz, L.B.; Sakai, K.; Bradford, T.R.; Ren, S.; Zweiman, B.; Worobec, A.S.; Metcalfe, D.D. The alpha form of human tryptase is the predominant type present in blood at baseline in normal subjects and is elevated in those with systemic mastocytosis. J. Clin. Investig. 1995, 96, 2702–2710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Serafin, W.E.; Austen, K.F. Mediators of immediate hypersensitivity reactions. N. Engl. J. Med. 1987, 317, 30–34. [Google Scholar] [CrossRef] [PubMed]
  34. Watkins, J.; Wild, G. Improved diagnosis of anaphylactoid reactions by measurement of serum tryptase and urinary methylhistamine. Ann. Fr. Anesth. Reanim. 1993, 12, 169–172. [Google Scholar] [CrossRef]
  35. Ravi, A.; Butterfield, J.; Weiler, C.R. Mast cell activation syndrome: Improved identification by combined determinations of serum tryptase and 24-hour urine 11β-prostaglandin2α. J. Allergy Clin. Immunol. Pract. 2014, 2, 775–778. [Google Scholar] [CrossRef] [PubMed]
  36. Tomasiak-Łozowska, M.M.; Klimek, M.; Lis, A.; Moniuszko, M.; Bodzenta-Łukaszyk, A. Markers of anaphylaxis—A systematic review. Adv. Med. Sci. 2018, 63, 265–277. [Google Scholar] [CrossRef] [PubMed]
  37. Butterfield, J.H.; Weiler, C.R. The utility of measuring urinary metabolites of mast cell mediators in systemic mastocytosis and mast cell activation syndrome. J. Allergy Clin. Immunol. Pract. 2020, 8, 2533–2541. [Google Scholar] [CrossRef]
  38. Boehm, T.; Reiter, B.; Ristl, R.; Petrosczi, K.; Sperr, W.; Stimpfl, T.; Valent, P.; Jilma, B. Massive release of the histamine-degrading enzyme diamine oxidase during severe anaphylaxis in mastocytosis patients. Allergy 2019, 74, 583–593. [Google Scholar] [CrossRef]
  39. Giannetti, M.P.; Akin, C.; Castells, M. Idiopathic anaphylaxis: A form of mast cell activation syndrome. J. Allergy Clin. Immunol. Pract. 2020, 8, 1196–1201. [Google Scholar] [CrossRef]
  40. Metcalfe, D.D.; Schwartz, L.B. Assessing anaphylactic risk? Consider mast cell clonality. J. Allergy Clin. Immunol. 2009, 123, 687–688. [Google Scholar] [CrossRef] [Green Version]
  41. Bonadonna, P.; Zanotti, R.; Pagani, M.; Caruso, B.; Perbellini, O.; Colarossi, S.; Olivieri, E.; Dama, A.; Schiappoli, M.; Senna, G.; et al. How much specific is the association between hymenoptera venom allergy and mastocytosis? Allergy 2009, 64, 1379–1382. [Google Scholar] [CrossRef] [PubMed]
  42. Gülen, T.; Hägglund, H.; Sander, B.; Dahlén, B.; Nilsson, G. The presence of mast cell clonality in patients with unexplained anaphylaxis. Clin. Exp. Allergy 2014, 44, 1179–1187. [Google Scholar] [CrossRef] [PubMed]
  43. Zanotti, R.; Lombardo, C.; Passalaqua, G.; Caimmi, C.; Bonifacio, M.; De Matteis, G.; Perbellini, O.; Rossini, M.; Schena, D.; Busa, M.; et al. Clonal mast cell disorders in patients with severe Hymenoptera venom allergy and normal serum tryptase levels. J. Allergy Clin. Immunol. 2015, 136, 135–139. [Google Scholar] [CrossRef] [Green Version]
  44. Bonadonna, P.; Bonifacio, M.; Lombardo, C.; Zanotti, R. Hymenoptera allergy and mast cell activation syndromes. Curr. Allergy Asthma Rep. 2016, 16. [Google Scholar] [CrossRef] [PubMed]
  45. González-De-Olano, D.; Esteban-López, M.I.; Alonso-Díaz-De-Durana, M.D.; González-Mancebo, E.; Prieto-García, A.; Gandolfo-Cano, M.; Mohedano-Vicente, E.; Balugo-López, V.; Fiandor, A.; Mielgo-Ballesteros, R.; et al. Frequency of clonal mast cell diseases among patients presenting with anaphylaxis: A prospective study in 178 patients from 5 tertiary centers in Spain. J. Allergy Clin. Immunol. Pract. 2019, 7, 2924–2926.e1. [Google Scholar] [CrossRef]
  46. Khoury, P.; Lyons, J.J. Mast cell activation in the context of elevated basal serum tryptase: Genetics and presentations. Curr. Allergy Asthma Rep. 2019, 19, 55. [Google Scholar] [CrossRef]
  47. Lyons, J.J.; Chovanec, J.; O’Connell, M.P.; Liu, Y.; Šelb, J.; Zanotti, R.; Bai, Y.; Kim, J.; DiMaggio, T.; Schwartz, L.B.; et al. Heritable risk for severe anaphylaxis associated with increased α-tryptase-encoding germline copy number at TPSAB1. J. Allergy Clin. Immunol. 2020, S0091–6749, 31029. [Google Scholar] [CrossRef]
  48. Greiner, G.; Sprinzl, B.; Gó>rska, A.; Ratzinger, F.; Gurbisz, M.; Witzeneder, N.; Schmetterer, K.G.; Gisslinger, B.; Uyanik, G.; Hadzijusufovic, E.; et al. Hereditary alpha tryptasemia is a valid genetic biomarker for severe mediator-related symptoms in mastocytosis. Blood 2020, in press. [Google Scholar] [CrossRef]
  49. Saleh, R.; Wedeh, G.; Herrmann, H.; Bibi, S.; Cerny-Reiterer, S.; Sadovnik, I.; Blatt, K.; Hadzijusufovic, E.; Jeanningros, S.; Blanc, C.; et al. A new human mast cell line expressing a functional IgE receptor converts to tumorigenic growth by KIT D816V transfection. Blood 2014, 124, 111–120. [Google Scholar] [CrossRef] [Green Version]
  50. Valent, P.; Akin, C.; Gleixner, K.V.; Sperr, W.R.; Reiter, A.; Arock, M.; Triggiani, M. Multidisciplinary challenges in mastocytosis and how to address with personalized medicine approaches. Int. J. Mol. Sci. 2019, 20, 2976. [Google Scholar] [CrossRef] [Green Version]
  51. Smiljkovic, D.; Kiss, R.; Lupinek, C.; Hoermann, G.; Greiner, G.; Witzeneder, N.; Krajnik, G.; Trautinger, F.; Vrtala, S.; Mittermann, I.; et al. Microarray-based detection of allergen-reactive IgE in patients with mastocytosis. J. Allergy Clin. Immunol. Pract. 2020, 8. [Google Scholar] [CrossRef] [PubMed]
  52. Reséndiz-Hernández, J.M.; Falfán-Valencia, R. Genetic polymorphisms and their involvement in the regulation of the inflammatory response in asthma and COPD. Adv. Clin. Exp. Med. 2018, 27, 125–133. [Google Scholar] [CrossRef] [PubMed]
  53. Mu, Z.; Zhang, J. The role of genetics, the environment, and epigenetics in atopic dermatitis. Adv. Exp. Med. Biol. 2020, 1253, 107–140. [Google Scholar] [CrossRef] [PubMed]
  54. Brown, S.; Elias, M.; Bradley, M. Genetics in atopic dermatitis: Historical perspective and future prospects. Acta Derm. Venereol. 2020, 100, adv00163. [Google Scholar] [CrossRef] [PubMed]
  55. Nedoszytko, B.; Reszka, E.; Gutowska-Owsiak, D.; Trzeciak, M.; Lange, M.; Jarczak, J.; Niedoszytko, M.; Jablonska, E.; Romantowski, J.; Strapagiel, D.; et al. Genetic and epigenetic aspects of atopic dermatitis. Int. J. Mol. Sci. 2020, 21, E6484. [Google Scholar] [CrossRef]
  56. Lee, E.; Kim, J.H.; Lee, S.Y.; Kang, M.J.; Park, Y.M.; Park, M.J.; Rhee, E.-S.; Ahn, K.; Kim, K.W.; Shin, Y.H.; et al. Association of IL13 genetic polymorphisms with atopic dermatitis. Ann. Allergy Asthma Immunol. 2020, 125. [Google Scholar] [CrossRef]
  57. Lyons, J.J.; Sun, G.; Stone, K.D.; Nelson, C.M.; Wisch, L.B.; O’Brien, M.; Jones, N.; Lindsley, A.; Komarow, H.D.; Bai, Y.; et al. Mendelian inheritance of elevated serum tryptase associated with atopy and connective tissue abnormalities. J. Allergy Clin. Immunol. 2014, 133, 1471–1474. [Google Scholar] [CrossRef] [Green Version]
  58. Lyons, J.J.; Yu, X.; Hughes, J.D.; Le, Q.T.; Jamil, A.; Bai, Y.; Ho, N.; Zhao, M.; Liu, Y.; O’Connell, M.P.; et al. Elevated basal serum tryptase identifies a multisystem disorder associated with increased TPSAB1 copy number. Nat. Genet. 2016, 48, 1564–1569. [Google Scholar] [CrossRef]
  59. Lyons, J.J.; Stotz, S.C.; Chovanec, J.; Liu, Y.; Lewis, K.L.; Nelson, C.; DiMaggio, T.; Jones, N.; Stone, K.D.; Sung, H.; et al. A common haplotype containing functional CACNA1H variants is frequently coinherited with increased TPSAB1 copy number. Genet. Med. 2018, 20, 503–512. [Google Scholar] [CrossRef]
  60. Carrigan, C.; Milner, J.D.; Lyons, J.J.; Vadas, P. Usefulness of testing for hereditary alpha tryptasemia in symptomatic patients with elevated tryptase. J. Allergy Clin. Immunol. Pract. 2020, 8, 2066–2067. [Google Scholar] [CrossRef]
  61. O’Connell, M.P.; Lyons, J.J. Hymenoptera venom-induced anaphylaxis and hereditary alpha-tryptasemia. Curr. Opin. Allergy Clin. Immunol. 2020, 20, 431–437. [Google Scholar] [CrossRef] [PubMed]
  62. Valent, P.; Akin, C.; Hartmann, K.; George, T.I.; Sotlar, K.; Peter, B.; Gleixner, K.V.; Blatt, K.; Sperr, W.R.; Manley, P.W.; et al. Midostaurin: A magic bullet that blocks mast cell expansion and activation. Ann. Oncol. 2017, 28, 2367–2376. [Google Scholar] [CrossRef] [PubMed]
  63. Reiter, A.; George, T.I.; Gotlib, J. New developments in diagnosis, prognostication, and treatment of advanced systemic mastocytosis. Blood 2020, 135. [Google Scholar] [CrossRef] [PubMed]
  64. Krauth, M.T.; Mirkina, I.; Herrmann, H.; Baumgartner, C.; Kneidinger, M.; Valent, P. Midostaurin (PKC412) inhibits immunoglobulin E-dependent activation and mediator release in human blood basophils and mast cells. Clin. Exp. Allergy 2009, 39, 1711–1720. [Google Scholar] [CrossRef] [PubMed]
  65. Gotlib, J.; Kluin-Nelemans, H.C.; George, T.I.; Akin, C.; Sotlar, K.; Hermine, O.; Awan, F.T.; Hexner, E.; Mauro, M.J.; Sternberg, D.W.; et al. Efficacy and safety of midostaurin in advanced systemic mastocytosis. N. Engl. J. Med. 2016, 374, 2530–2541. [Google Scholar] [CrossRef] [PubMed]
  66. Hartmann, K.; Gotlib, J.; Akin, C.; Hermine, O.; Awan, F.T.; Hexner, E.; Mauro, M.J.; Menssen, H.D.; Redhu, S.; Knoll, S.; et al. Midostaurin improves quality of life and mediator-related symptoms in advanced systemic mastocytosis. J. Allergy Clin. Immunol. 2020, 146, 356–366.e4. [Google Scholar] [CrossRef] [PubMed]
  67. Broesby-Olsen, S.; Vestergaard, H.; Mortz, C.G.; Jensen, B.; Havelund, T.; Hermann, A.P.; Siebenhaar, F.; Møller, M.B.; Kristensen, T.K.; Bindslev-Jensen, C.; et al. Omalizumab prevents anaphylaxis and improves symptoms in systemic mastocytosis: Efficacy and safety observations. Allergy 2018, 73, 230–238. [Google Scholar] [CrossRef] [PubMed]
  68. Lemal, R.; Fouquet, G.; Terriou, L.; Vaes, M.; Livideanu, C.B.; Frenzel, L.; Barete, S.; Canioni, D.; Lhermitte, L.; Rossignol, J.; et al. Omalizumab therapy for mast cell-mediator symptoms in patients with ISM, CM, MMAS, and MCAS. J. Allergy Clin. Immunol. Pract. 2019, 7, 2387–2395.e3. [Google Scholar] [CrossRef]
Figure 1. Algorithm in Patients with Suspected MCAS.
Figure 1. Algorithm in Patients with Suspected MCAS.
Ijms 21 09030 g001
Table 1. Mast Cell-derived Mediators and Related Clinical Symptoms
Table 1. Mast Cell-derived Mediators and Related Clinical Symptoms
Symptoms/PathologyMediator(s) Potentially Involved *
Systemic Symptoms
Vascular instabilityHistamine, LTC4, LTE4, PGD2, PAF
Edema formationHistamine, VEGF, LTC4, LTE4, PAF
Tissue remodelingCytokines, Proteases (Tryptase, Chymase)
Bleeding tendencyHeparin, Tissue Type Plasminogen Activator
Fever and cachexiaTumor Necrosis Factor
Eosinophilia, eosinophil infiltrationCytokines (GM-CSF, IL-5), Chemokines
Lymphocyte infiltrationCytokines, Chemokines
Neurologic symptoms, fatigueCytokines, Histamine
Headache and nauseaHistamine
Skin
PruritusHistamine, Cytokines
UrticariaHistamine, PAF, PGE2, PGD2
AngioedemaHistamine, Bradykinin
FlushingHistamine
GI-Tract
Gastric hypersecretionHistamine
Peptic ulcer diseaseHistamine
Cramping, abdominal painHistamine, LTC4, PAF
DiarrheaHistamine
Respiratory System
Nasal congestion, wheezingHistamine
BronchoconstrictionHistamine, PGD2, LTC4, LTD4, PAF, Endothelin
Secretion of mucusHistamine, Proteases, PGD2, LTC4
Pulmonary edemaHistamine, LTC4, PAF
Skeletal System
Bone remodelingProteases, Cytokines
OsteoporosisHeparin, Proteases
* Whereas some of the mast cell mediators (like histamine or heparin) are produced and stored in metachromatic granules, others (PGs/LTs) are produced and released but are not stored. LT, leukotriene; PG, prostaglandin; GM-CSF, granulocyte/macrophage colony-stimulating factor; VEGF, vascular permeability factor; IL, interleukin, GI-Tract, gastrointestinal tract; PAF, platelet-activating factor.
Table 2. Classification of Mast Cell Activation (MCA) and Related Conditions.
Table 2. Classification of Mast Cell Activation (MCA) and Related Conditions.
i. According to Organs involved and Severity of Symptoms
    Systemic MCA *
      Mild or moderate systemic MCA (MCAS criteria not fulfilled)
      Severe systemic MCA = MCAS (MCAS criteria fulfilled)
    Local MCA (mild/moderate or severe) (MCAS criteria not fulfilled)
ii. According to the Underlying Condition
    Primary (clonal) MCA
      Cutaneous mastocytosis
      Systemic mastocytosis (SM)
      1-2 minor SM criteria but no SM and no CM detected
    IgE-dependent allergy (or atopy)
      Organ-specific variants
      IgE-independent hypersensitivity reactions
    Other Conditions
      Reactive conditions (inflammation)
      Toxic tissue damage (intoxication)
      Physical, neurologic and others
iii. According to Frequency and Symptom-Free Intervals
    Acute single event
      With a known trigger (e.g., allergen)
      Without a known trigger
    Episodic recurrent
      With a known trigger (e.g., allergen)
      Without a known trigger
    Chronic persistent
      With a known trigger (e.g., allergen)
      Without a known trigger
* Systemic MCA involves two or more organ systems.
Table 3. Mast Cell Mediators recommended as Markers of Systemic MCA in Practice.
Table 3. Mast Cell Mediators recommended as Markers of Systemic MCA in Practice.
Recommendation LevelValidated Diagnostic Thresholds (Increase from Baseline) Available
Recommended as First Line Standard:
Tryptase (serum)yes: plus 20%+2 equation
Recommended as Alternative or Confirmatory:
Histamine metabolites (urinary)no
Prostaglandin D2 metabolites (urinary)no
Potentially Useful or Under Development:
Histamine (plasma)no
Diamine oxidase (DAO) *no
Soluble IgE receptor alpha chainno
Not Recommended:
Heparinno
Chymaseno
Chromogranin Bno
Bradykininno
Stem cell factorno
Interleukinsno
Chemokinesno
Basogranulinno
Platelet activating factor (PAF)no
* So far it remains unclear whether DAO is expressed by or released from mast cells.
Table 4. MCAS Mimickers: Differential Diagnoses to MCAS *.
Table 4. MCAS Mimickers: Differential Diagnoses to MCAS *.
Disorder/Mimicker Clinical Findings/Symptoms Presenting as Mimicry
Cardiovascular Mimickers
Myocardial InfarctionHypotension, Shock, Syncope
Endocarditis/Endomyocarditis Hypotension, Shock
Aortic Stenosis with Syncope Syncope
Pulmonary Infarction Dyspnea, Hypotension
Endocrinologic Mimickers
Acute HypothyroidismHypotension, Shock
Acute Hypoglycemia Hypotension, Shock
Adrenal Insufficiency Hypotension, Shock
HypopituitarismHypotension, Shock
Gastrointestinal Mimickers
Acute Inflammatory Bowel DiseaseDiarrhea, Pain, Dehydration, Hypotension
VIP-secreting Tumor (VIPoma)Diarrhea, Dehydration, Hypotension
Active Crohn’s Disease or Colitis UlcerosaDiarrhea, Pain, Dehydration, Hypotension
Food Intoxication Diarrhea, Dehydration, Hypotension
Infectious Disease Mimickers
Severe Bacterial or Viral Infections Septic Shock
Acute Gastrointestinal InfectionDiarrhea, Cramps, Dehydration, Hypotension
Acute Encephalitis / Meningitis Headache, Confusion, Fatigue, Hypotension
Acute Parasitic Diseases (e.g., Acute Chagas Disease) Dehydration, Rash, Hypotension
(e.g., Acute Chagas Disease) Headache, Dyspnea, Hypotension
Neurologic/Central Nervous System (CNS) Mimickers
EpilepsyHeadache, Confusion, Fatigue, Shock
CNS Tumors Headache, Confusion, Fatigue, Hypotension
Other CNS Diseases Headache, Fatigue, Hypotension
IntoxicationHeadache, Confusion, Fatigue, Hypotension
Somatoform disorders Headache, Fatigue, Hypotension
Psychiatric conditions Headache, Confusion, Fatigue, Hypotension
Cutaneous Mimickers
Hereditary or Acquired Angioedema Angioedema, Rash, Hypotension
Acute Lupus ErythematosusRash, Headache, Fatigue
Acute Toxic Dermatoses Exanthema, Hypotension
Hematologic Mimickers: Acute Anemia
Acute Gastrointestinal Bleeding Hypovolemic Shock
Massive Hypermenorrhea Hypovolemic Shock
Drug-related Mimickers (Adverse Events after Drug Intake)
Drug-induced Hypoglycemia Fatigue, Loss of Consciousness
Drug-induced HypotensionHypotension, Shock
Drug-induced DiarrheaDiarrhea, Cramps, Dehydration, Hypotension
Drug-Induced CNS Damage Headache, Fatigue, Hypotension, Confusion
* In most instances, symptoms of acute hypotension are recorded. If additional MCA-mimicking symptoms, such as skin lesions, headache and/or diarrhea are found, it is often difficult to separate the pathology from MCA and thus MCAS. Abbreviations: MCA, mast cell activation; MCAS, MCA syndrome; VIP, vasoactive intestinal peptide.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Valent, P.; Akin, C.; Nedoszytko, B.; Bonadonna, P.; Hartmann, K.; Niedoszytko, M.; Brockow, K.; Siebenhaar, F.; Triggiani, M.; Arock, M.; et al. Diagnosis, Classification and Management of Mast Cell Activation Syndromes (MCAS) in the Era of Personalized Medicine. Int. J. Mol. Sci. 2020, 21, 9030. https://doi.org/10.3390/ijms21239030

AMA Style

Valent P, Akin C, Nedoszytko B, Bonadonna P, Hartmann K, Niedoszytko M, Brockow K, Siebenhaar F, Triggiani M, Arock M, et al. Diagnosis, Classification and Management of Mast Cell Activation Syndromes (MCAS) in the Era of Personalized Medicine. International Journal of Molecular Sciences. 2020; 21(23):9030. https://doi.org/10.3390/ijms21239030

Chicago/Turabian Style

Valent, Peter, Cem Akin, Boguslaw Nedoszytko, Patrizia Bonadonna, Karin Hartmann, Marek Niedoszytko, Knut Brockow, Frank Siebenhaar, Massimo Triggiani, Michel Arock, and et al. 2020. "Diagnosis, Classification and Management of Mast Cell Activation Syndromes (MCAS) in the Era of Personalized Medicine" International Journal of Molecular Sciences 21, no. 23: 9030. https://doi.org/10.3390/ijms21239030

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop