CC BY 4.0 · Semin Thromb Hemost
DOI: 10.1055/s-0042-1758820
Review Article

Hereditary Angioedema: The Clinical Picture of Excessive Contact Activation

Remy S. Petersen
1   Department of Vascular Medicine, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
,
Lauré M. Fijen
1   Department of Vascular Medicine, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
,
Marcel Levi
1   Department of Vascular Medicine, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
,
Danny M. Cohn
1   Department of Vascular Medicine, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Amsterdam, the Netherlands
› Author Affiliations
 

Abstract

Hereditary angioedema is a rare, genetic disorder characterized by painful, debilitating and potentially life-threatening angioedema attacks in subcutaneous and submucosal tissue. While usually unpredictable, attacks can be provoked by a variety of triggers including physical injury and certain medication and are often preceded by prodromal symptoms. Hereditary angioedema has a profound influence on the patients' lives. The fundamental cause of hereditary angioedema in almost all patients is a mutation in the SERPING1 gene leading to a deficiency in C1-inhibitor. Subsequently, the contact activation cascade and kallikrein-kinin pathway are insufficiently inhibited, resulting in excessive bradykinin production triggering vascular leakage. While C1-inhibitor is an important regulator of the intrinsic coagulation pathway, fibrinolytic system and complement cascade, patients do not have an increased risk of coagulopathy, autoimmune conditions or immunodeficiency disorders. Hereditary angioedema is diagnosed based on C1-inhibitor level and function. Genetic analysis is only required in rare cases where hereditary angioedema with normal C1-inhibitor is found. In recent years, new, highly specific therapies have greatly improved disease control and angioedema-related quality of life. This article reviews the clinical picture of hereditary angioedema, the underlying pathophysiology, diagnostic process and currently available as well as investigational therapeutic options.


#

Clinical Case

A 21-year-old female was referred to the emergency department with an acute swelling of the throat. She had a history of recurrent episodes of non-pruritic swelling of extremities that usually resolved spontaneously within three days. These episodes started in puberty, occurred about once a month and were never accompanied by hives. The patient had presented to the emergency department twice before, both times with extreme abdominal pain accompanied by vomiting and diarrhea without an identified cause. Her mother had similar episodes of edema, although less frequently. Her grandfather passed away at the age of 30 as a result of asphyxiation. The patient used combined estrogen-progestin oral contraceptives and in the past had tried high-dose anti-histamines without effect on the episodes of swelling. At presentation, epinephrine, clemastine, and dexamethasone, were administered immediately, with no effect on what was diagnosed as laryngeal edema. Laryngeal intubation was necessary to secure the airway and the patient was admitted to the intensive care unit. An empirical dose of plasma-derived C1-inhibitor was administered, after which the edema swiftly resolved. The patient was discharged with a subcutaneous bradykinin B2 receptor inhibitor to be administered in case of another attack. Several weeks later, laboratory tests confirmed the diagnosis of hereditary angioedema (HAE) type 1, due to deficiency of C1-inhibitor. The patient started with prophylactic subcutaneous plasma-derived C1-inhibitor concentrates twice weekly and switched to a type of contraception without estrogen. In the following months, she continued to experience occasional break-through attacks that responded well to self-administered subcutaneous bradykinin B2 receptor inhibitor.


#

Clinical Features

HAE is a rare, genetic disease that is estimated to affect about 1 in 50,000 to 100,000 individuals.[1] Patients with HAE have recurring episodes of non-pitting, non-pruritic angioedema. Angioedema occurs in subcutaneous and submucosal areas and causes different symptoms depending on the location. If untreated, attacks typically worsen in the first 24 hours before resolving in two to five days.[2] Contrary to mast cell-mediated angioedema, attacks are not accompanied by urticaria or anaphylaxis.[2] Onset of symptoms is usually during childhood or adolescence, with a mean age at first symptoms of 11 years.[3] Frequency and locations of attacks vary significantly between patients and can even vary during a single patient's lifetime.[2] Up to 30% of patients experience edema at multiple anatomically unrelated regions simultaneously.[4] Hindering and debilitating dermal swelling predominantly occurs in the extremities, in the orofacial or genital region. When submucosa of the gastrointestinal tract is involved, patients present with extreme abdominal pain and vomiting, mimicking an acute abdomen, in some cases leading to unnecessary surgical interventions. HAE patients are estimated to be two-and-a-half times more likely to undergo abdominal surgery compared to the general population.[5] In the case of upper airway edema, impending airway obstruction can lead to asphyxiation if the airway is not secured or the edema treated in time. A review of literature published in 1962, before adequate treatment was developed, reported that 77% of HAE patients died due to acute laryngeal edema.[6] More recently, worldwide mortality amongst HAE patients due to laryngeal edema was still estimated to be as high as 5%.[7]

Various events and circumstances can trigger attacks of angioedema. Trauma caused by invasive procedures is well known to induce attacks and therefore pre-procedural prophylactic medication is recommended.[8] Patients should refrain from inhibitors of angiotensin-converting enzyme or dipeptidyl peptidase IV for inhibition of these enzymes is known to increase attack frequency as illustrated in [Fig. 1].[8] Increased exogenous or endogenous estrogen levels typically enhance attacks in female patients. This can occur in physiological circumstances like pregnancy and during the menstrual cycle, and also with the use of oral contraceptives or hormonal replacement therapy.[9] Other reported triggers include infection, fever, mental stress, and physical activity.[10] [11]

Zoom Image
Fig. 1 Degradation of bradykinin. Bradykinin binds to the bradykinin B2 receptor (B2R) and is transformed to des-arg9-bradykinin by carboxypeptidase N and M (CPN/CPM), which in turn binds to the bradykinin B1 receptor (B1R). Bradykinin is principally degraded by angiotensin converting enzyme (ACE), while angiotensin converting enzyme 2 (ACE2) has a higher affinity for des-arg9-bradykinin. Both bradykinin and des-arg9-bradykinin are broken down into inactive parts by aminopeptidase P (APP), dipeptidyl peptidase IV (DPP-IV), and neutral endopeptidase (NEP).

The occurrence of angioedema attacks is often unpredictable; however, 90% of patients experience prodromal symptoms hours to days before an attack.[12] [13] [14] These symptoms span a wide range including dermal manifestations, tiredness, emotional disturbances and gastro-intestinal symptoms. The most common dermal manifestation described to precede HAE attacks is erythema marginatum. This non-pruritic skin rash is sometimes misinterpreted as urticaria and is associated with a longer diagnostic delay.[12] The majority of patients report to be able to predict the onset of an attack and a recent study described the sensitivity and specificity of prodromes as a predictor of attacks as 89.5 and 63.1%, respectively.[14]

Frequent unpredictable and debilitating angioedema attacks can have a significant impact on the daily life of HAE patients. Depression and anxiety are reported in a substantial percentage of HAE patients, corresponding with a lower than average quality of life with regard to mental health and social relationships.[15] [16] [17] [18] Patients report a loss of productivity and absence from work due to attacks. Especially facial, abdominal, and laryngeal attacks correlate with higher absenteeism and activity impairment.[17] Even in attack-free periods, patients report anxiety due to the unpredictability of attacks, as well as worries about passing HAE on to their offspring.[18] HAE is shown to influence reproductive choices in approximately 20% of HAE patients.[19]


#

Pathophysiology

In HAE patients, angioedema is caused by insufficient inhibition of the contact activation pathway and kallikrein-kinin system. In the majority of patients, this is caused by an absolute C1-inhibitor deficiency (type 1, 85% of HAE with C1-inhibitor deficiency) or C1-inhibitor dysfunction (type 2, 15% of HAE with C1-inhibitor deficiency). C1-inhibitor is a heavily glycosylated single chain polypeptide of 478 amino acid residues with a molecular weight of 105 kDa. The physiological plasma concentration is about 240 mg/L with a long estimated half-life of about 70 hours.[20] C1-inhibitor regulates various pathways, including the kallikrein-kinin system, intrinsic coagulation system, fibrinolytic pathway, and the complement cascade ([Fig. 2]). The contact activation route and subsequent kallikrein-kinin system are initiated when factor XII (FXII) is autoactivated upon contact with negatively charged surfaces and subsequently induces cleavage of the zymogen prekallikrein into plasma kallikrein (PKa). PKa activates more FXII in a positive feedback loop and induces cleavage of high-molecular-weight kininogen, thereby producing bradykinin,[21] a nonapeptide of the kinin family. Bradykinin is considered to be principally responsible for the vasodilation and subsequent transfer of fluid to extravasal tissue in the case of angioedema formation in HAE patients. Bradykinin binds to the bradykinin B2 receptor (BKB2R) expressed on endothelial cells, initiating a signaling cascade leading to increased vascular permeability through several mechanisms including the release of nitric oxide, contraction of endothelial cells, and the uncoupling of tight junctions between endothelial cells. Carboxypeptidases N and M cleave the terminal arginine from bradykinin, forming des-Arg9-bradykinin, an agonist for the bradykinin B1 receptor (BKB1R).[21] This receptor is not constitutively present on endothelial cells, but is expressed following tissue injury or inflammatory stimuli. It is unclear what the function of the BKB1R is in the formation of angioedema in the context of HAE. The receptor is slowly and partially desensitized after binding, in contrast to the BKB2R which is rapidly desensitized.[22] Therefore, it has been hypothesized that stimulation of the BKB1R could explain the long duration of angioedema attacks.[23] Bradykinin and des-Arg9bradykinin are rapidly broken down into inactive metabolites by aminopeptidase P, neutral endopeptidase, dipeptidyl peptidase IV, and angiotensin-converting enzyme.[24]

Zoom Image
Fig. 2 Functions of C1-inhibitor. C1-inhibitor (C1-INH) is a crucial regulator of several intravascular cascades, including the contact-activation system and kallikrein-kinin pathway. The contact-activation system starts when factor XII (FXII) is converted in active factor XII (FXIIa), illustrated by a solid arrow. The activation of plasma prekallikrein (PK) in plasma kallikrein (PKa) is subsequently stimulated by FXIIa, as illustrated by a dotted arrow. Kallikrein, in turn, induces further activation of FXII and facilitates the cleavage of high-molecular weight kininogen (HK) into cleaved high-molecular weight kininogen (cHK) and bradykinin, which in turn binds to the bradykinin B2 receptor (B2R), resulting in the efflux of fluid to extravasal tissue. In the coagulation system, C1-INH inhibits both the activation of factor XI (FXI) and the function of active FXI (FXIa). In the fibrinolytic pathway, kallikrein induces the activation of plasminogen (Plg) to plasmin (Pl) directly and indirectly through the stimulation of urokinase plasminogen activator (uPA). C1-INH inhibits these functions of kallikrein as well as the direct effects of plasmin on fibrin degradation. In the complement system, both the classical pathway, involving complement 1 complex (C1 complex), complement 2 (C2), and complement 4 (C4, as well as the lectin pathway involving mannose-associated serine protease 1 and 2 (MASP-1/2) are inhibited by C1-INH.

C1-inhibitor is an important regulator of other pathways as well. C1-inhibitor regulates the intrinsic coagulation pathway by partially inhibiting the activation of factor XI and the functions of activated factor XI.[25] Likewise, C1-inhibitor regulates the fibrinolytic system directly by inhibiting plasmin activity, though less efficient than α2-antiplasmin,[26] and indirectly by inhibiting the conversion of plasminogen to plasmin by PKa.[20] Signs of activation of the thrombotic and fibrinolytic systems in HAE patients include elevated levels of activated factor XII (FXIIa), D-dimer, plasmin-α2-antiplasmin complexes and thrombin generation during attacks.[27] [28] [29] [30] Despite the impact of C1-inhibitor on both coagulation and fibrinolysis, C1-inhibitor deficiency does not translate into an increased risk of thrombosis or bleeding.[31] Furthermore, C1-inhibitor is a key player in the complement system, for it is the only known inhibitor of C1r and C1s proteases in the classical complement pathway. It also regulates the lectin pathway by inactivating mannose-associated serine proteases 1 and 2.[32] [33] There is no proven association between immunodeficiency disorders or autoimmune conditions and HAE.[34]

In HAE type 1 and 2, lack of (functional) C1-inhibitor is caused by a mutation in the SERPING1 gene. About 750 different pathogenic mutations are reported to be associated with HAE type I, including missense, nonsense, deletions, insertions, duplications and splice site mutations.[35] [36] Most cases of HAE type 2 are caused by a missense mutation in exon 8, resulting in the reduced ability to inhibit target proteases.[22] HAE is inherited as an autosomal dominant trait in almost all cases. While HAE type I patients possess one functional allele, levels of C1-inhibitor normally vary between 5 and 30% of normal levels. Recent studies suggest that in a limited subset of mutations, this could be due to the formation of polymers between functional and dysfunctional C1-inhibitor leading to intracellular retention.[37] [38] Homozygous forms of HAE have long been considered embryonically lethal and only in recent years case reports of consanguineous families with homozygous HAE have sporadically been published. The inheritance pattern in these families is most likely recessive since heterogeneous members of these families are almost always asymptomatic. An unusual biochemical feature found in some patients with homozygous C1-INH deficiency is a significant reduction in C1q levels.[39] [40] [41] [42]

In rare cases, HAE with normal functional and absolute levels of C1-inhibitor (HAE-nC1-INH) occurs. This type of HAE has a very heterogeneous phenotype. Known mutations causing this third type of HAE are summarized in [Table 1]. In up to 25% of HAE-nC1-INH patients a mutation in the gene coding for FXII (HAE-FXII) is found, leading to an increase in autoactivation of FXII. While this mutation is inherited as an autosomal dominant trait, the majority of symptomatic carriers is female,[43] implying an association with estrogen. In patients with HAE type 1 and 2, the influence of estrogen on attack frequency and severity has been described extensively.[44] Previously suggested mechanisms for this observation are the increase of FXII levels, decrease of C1-inhibitor levels, induction of the release of nitric oxide and impairment of the functionality of angiotensin converting enzyme.[44] [45] The specific characteristics of HAE-FXII suggest the influence of estrogen on FXII might be an important factor in the development of angioedema. Another noteworthy difference in the clinical presentation of patients with HAE-FXII is the absence of erythema marginatum as a prodromal symptom.[46]

Table 1

Mutations associated with HAE-nC1-INH

Affected gene

First reported by

Known mutations

Suspected agent

Suspected mechanism

F12

Dewald and Bork[97]

T328K[97]

T328R[97]

c.971_1018 + 24del72[98]

c.892_909dup[99]

Bradykinin

Increased autoactivation of factor XII or increased sensitivity to activation by plasmin

ANGPT1

Bafunno et al[100]

A119S

A8V

Q370H

Vascular endothelium

Disruption of the multimerization of angiopoetin-1, leading to a reduced ability to bind to its receptor on endothelial cells

PLG

Bork et al[101]

K330E

Bradykinin

Direct liberation of bradykinin from high-molecular weight kininogen[102]

KNG1

Bork et al[103]

M379K

Bradykinin

Mechanism remains unclear

MYOF

Ariano et al[104]

R217S

Vascular endothelium

Increased activation of VEGF-mediated intracellular signaling

HS3ST6

Bork et al[105]

T144S

Unclear

Incomplete synthesis of heparan sulfate by heparan sulfate-glucosamine 3-O-sulfotransferase, possibly affecting the interaction between endothelial cell surface and high-molecular weight kininogen

ANGPT1, angiopoietin-1 gene; F12, factor XII gene; HAE-nC1-INH, hereditary angioedema with normal C1-inhibitor; HS3ST6, heparan sulfate-glucosamine 3-O-sulfotransferase 6 gene; KNG1, Kininogen-1 gene; MYOF, myoferlin gene; PLG, plasminogen gene; VEGF, vascular endothelial growth factor



#

Diagnosis

When HAE is suspected, serum levels of C1-inhibitor protein, C1-inhibitor function, and complement component 4 are measured ([Table 2]). While HAE type 1 and 2 are clinically indistinguishable, laboratory tests will make it possible to differentiate between these types: serum of patients with type 1 HAE will show a C1-inhibitor protein level of <50% of normal combined with a decreased functionality of C1-inhibitor, whereas patients with type 2 will have a decrease in C1-inhibitor functionality in the presence of normal or increased levels of C1-inhibitor protein. Even though C4 levels are usually decreased in HAE patients, it is not recommended to use this as a single screening tool due to its limited sensitivity and specificity, estimated at 81 and 85%, respectively.[47] The most recent guideline recommends to repeat C1-inhibitor function, C1-inhibitor protein, and C4 testing in patients to confirm the diagnosis.[8] Genetic sequencing is not recommended as a diagnostic tool for patients with HAE type 1 or 2. However, in case of a high suspicion of HAE with normal C1-inhibitor, assessment for known mutations underlying HAE-nC1-INH is recommended.[8] While C1-inhibitor level and function remain the golden standard for diagnosis of HAE type 1 and 2, analysis can take weeks, making them impractical to distinguish HAE from other forms of angioedema in the acute setting. In patients that are already diagnosed with HAE, there is a need for biomarkers to correctly identify and predict attacks. While recent studies have yielded promising results, none are recommended to be used in daily practice yet.[48] [49] A reduced level of C1-inhibitor protein has a short differential diagnosis. However, if a patient with laboratory findings corresponding with HAE type 1 has a negative family history and onset of symptoms later in life (>30 years), it is important to exclude acquired angioedema with C1-inhibitor deficiency (AAE-C1-INH). This condition has an even lower prevalence than HAE and is estimated to affect 1 in 500,000 individuals. It is associated with plasma cell dyscrasias, lymphoproliferative disorders, and autoimmune diseases that cause a deficiency of C1-inhibitor either through neutralizing antibodies or through increased consumption by massive activation of the complement system.[50] [51] Contrary to HAE, laboratory tests of about 75% of AAE-C1-INH patients show low levels of C1q and in part of the population C1-inhibitor autoantibodies can be identified.[52] [53] AAE-C1-INH requires additional diagnostics to exclude or confirm associated disorders and has different treatment options than HAE, therefore it is important to distinguish between these disorders.[54]

Table 2

Laboratory findings in HAE and AAE-C1-INH

C1-INH level

C1-INH function

C4 level

C1q level

HAE Type 1

N

HAE Type 2

N/↑

N

HAE-nC1-INH

N

N

N

N

AAE-C1-INH

N/↓

Abbreviations: AAE-C1-INH, acquired angioedema with C1-inhibitor deficiency; C1-INH, C1-inhibitor; HAE, hereditary angioedema; HAE-nC1-INH, hereditary angioedema with normal C1-inhibitor; N, normal


HAE patients have a 50% chance to pass the affected gene on to their offspring. However, severity and frequency of symptoms are highly variable even within one family. Notably, in about 25% of patients a de novo mutation occurs.[55] Recent guidelines recommend to test children from HAE patients as early as possible.[8] [56] However, the testing of neonates and infants proves difficult, since antigenic and functional C1-inhibitor levels in peripheral and cord blood of healthy children are lower than in adults.[57] Therefore, tests performed in infants should be repeated after the child is one year old. Genetic testing (preferably of umbilical cord blood) can be considered if the mutation of the parent is known, which is not the case in up to 10% of patients.[8] [56] Prenatal diagnosis and pre-implantation genetic testing are available depending on national laws and practice.[9] [19]


#

Treatment

In contrast to mast cell-mediated forms of angioedema, HAE attacks do not respond to epinephrine, antihistamines, or corticosteroids.[58] In recent years, new improved therapies have drastically changed the landscape of HAE and have even led to a change in treatment goals. In 2021, a global Delphi initiative among HAE specialists concluded that the ultimate goals of treatment should be complete control of the disease and normalization of the patient's life.[59] More highly effective drugs are expected to become available in the near future, making the ambitious treatment goals more realistic for a growing number of patients. Currently available treatment options and therapies in development are summarized in [Table 3].

Table 3

Treatment options for HAE currently available or in development

Drug

Manufacturer (Trade name)

Indication

Mechanism of action

Administration

Regulatory / developmental status

Currently available treatment

 Attenuated androgens (danazol, oxandrolone, stanozolol)

Generic manufacturers

LTP, STP

Induction of APP activity, increased C1-INH synthesis and C1-INH mRNA expression

Oral

Licensed

 Berotralstat

BioCryst (Orladeyo)

LTP

Small molecule plasma kallikrein inhibitor

Oral

Licensed

 Conestat alfa

Pharming (Ruconest)

ODT

Recombinant C1-INH concentrate replacing deficient C1-INH

Intravenous

Licensed

 Ecallantide

Takeda (Kalbitor)

ODT

Plasma kallikrein inhibitor

Subcutaneous

Licensed only in United States

 Icatibant

Takeda (Firazyr)

ODT

Bradykinin B2 receptor antagonist

Subcutaneous

Licensed

 Lanadelumab

Takeda (Takhzyro)

LTP

Monoclonal antibody plasma kallikrein inhibitor

Subcutaneous

Licensed

 Plasma-derived C1-INH concentrate

CSL Behring (Berinert), CSL Behring (Haegarda) Takeda (Cinryze)

ODT, LTP, STP

Replacement of deficient C1-INH

Intravenous or subcutaneous

Licensed

Developmental treatment

 ATN-249

Attune Pharmaceuticals

LTP

Small molecule plasma kallikrein inhibitor

Oral

Phase I

 BMN 331

BioMarin

Curation

AAV5-based gene therapy of C1-INH

Intravenous

Phase 1

 Donidalorsen

Ionis

LTP

Antisense oligonucleotide targeted at prekallikrein

Subcutaneous

Phase 3

 Garadacimab

CSL Behring

LTP

Monoclonal antibody FXIIa inhibitor

Subcutaneous

Phase 3

 KV998086

KalVista

LTP

Small molecule FXIIa inhibitor

Oral

Preclinical

 NTLA-2002

Intellia

Curation

CRISPR-CAS9-based gene editing of prekallikrein

Intravenous

Phase 1

 OTL-105

Orchard Therapeutics and Pharming Group

Curation

Ex vivo autologous hematopoietic stem cell gene therapy to restore C1-INH production

Intravenous

Preclinical

 PHA-022121

Pharvaris

LTP and ODT

Bradykinin B2 receptor antagonist

Oral

Phase 2

 Sebetralstat

KalVista

ODT

Small molecule plasma kallikrein inhibitor

Oral

Phase 3

 STAR-0215

Astria Therapeutics inc.

LTP

Monoclonal antibody plasma kallikrein inhibitor

Subcutaneous

Preclinical

 Unnamed

Spark Therapeutics

Curation

Liver-directed gene therapy

Intravenous

Preclinical

 Unnamed

RegenxBio

Curation

AAV-based gene therapy

Intravenous

Preclinical

Abbreviations: AAV, adeno-associated virus; APP, aminopeptidase P; C1-INH, C1-inhibitor; CRISPR-CAS9, clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9; FXIIa, active Factor XII; HAE, hereditary angioedema; LTP, long-term prophylaxis; mRNA, messenger ribonucleic acid; ODT, on-demand treatment; STP, short-term prophylaxis


HAE treatment consists of a twin-track approach, comprising of symptom relief in the case of an attack on one hand, and the overall decrease in attacks on the other hand. The most recent guideline advises to treat an HAE attack with on demand treatment (ODT) as early as possible, for this is associated with the best treatment response.[60] [61] [62] First line ODT options are plasma-derived or recombinant C1-inhibitor, icatibant or ecallantide.[8] Plasma-derived C1-inhibitor was first administered to an HAE patient in 1979[63] and was approved by the Food and Drug Administration in 2008. Self-administration of intravenous plasma-derived C1-inhibitor concentrate is shown to be safe and effective,[64] though complications may arise if frequent venous access is required.[65] Even though C1-inhibitor has a direct effect on both the intrinsic coagulation and the fibrinolytic pathway, there is no evidence of an elevated risk of thromboembolic events with the use of C1-inhibitor concentrate at recommended doses.[66] A recombinant intravenous C1-inhibitor concentrate called conestat alfa was developed as an alternative for plasma-derived C1-inhibitor.[67] Another frequently used ODT option is icatibant, a competitive selective antagonist of the BKB2R.[68] Icatibant can be self-administered subcutaneously and has been proven to efficiently shorten the time to onset of symptom relief, especially when administered quickly after the first symptoms have emerged.[60] [68] In the United States of America, the subcutaneous direct PKa inhibitor ecallantide is available for ODT. Efficacy has been proven,[69] however, utilization is limited for it can only be administrated by a health care professional with adequate medical support due to the occurrence of anaphylaxis in 3.5% of patients.[70] Before the development of specific therapies, fresh-frozen plasma has been used to replenish deficient C1-inhibitor in case of severe, life-threatening attacks, although not without risks.[71] [72] Nowadays, the use of fresh-frozen plasma is only recommended if no targeted therapies are available,[8] which is still the case in some countries where targeted therapies are unaffordable or not yet registered.[73]

Short-term prophylaxis (STP) is indicated for invasive procedures known to trigger attacks. The only recommended first-line STP in the most recent guideline is intravenous plasma-derived C1-inhibitor.[8] The need for long-term prophylaxis (LTP) is patient-specific and depends on disease activity and burden, as well as patient preferences.[8] The recently updated guideline advises the following first line options for LTP: plasma-derived C1-inhibitor, lanadelumab, and berotralstat.[8] A new subcutaneous form of plasma-derived C1-inhibitor demonstrated a positive effect on HAE-related quality of life.[74] [75] In recent years, two new prophylactic therapies targeting PKa have been licensed by the Food and Drug Administration and the European Medicines Agency. Lanadelumab is an IgG1 type monoclonal antibody directed at PKa with a high efficacy: a subcutaneous dose once every 2 weeks reduced angioedema attacks with more than 90% in a placebo-controlled trail.[76] Berotralstat is an oral PKa inhibitor that reduced angioedema attacks by 44% compared to placebo in a phase 3 study.[77] Attenuated androgens (danazol, oxandrolone, and stanozol) have been used as first-line STP and LTP for more than four decades.[78] They are thought to reduce attack frequency through various mechanisms: induction of C1-inhibitor synthesis in hepatocytes, increased expression of C1-inhibitor mRNA in peripheral blood mononuclear cells,[79] and increased bradykinin degradation by induction of aminopeptidase P activity.[80] While effective in some, long-term use of attenuated androgens comes with considerable side effects in a majority of patients.[81] With the development of effective alternatives, attenuated androgens are now only recommended as second-line LTP.[8] Likewise, antifibrinolytics such as tranexamic acid have long been used as LPT as they prevent plasmin-mediated amplification of FXII activation by inhibiting the activation of plasminogen into plasmin. However, due to insufficient efficacy, they are no longer recommended in the latest international guideline.[8]

New targeted therapies now in development are highly specific to individual components of the contact activation pathway or kallikrein-kinin system. Therapies targeting FXIIa prevent angioedema by hampering contact activation at the very beginning of the cascade. Garadacimab is a IgG4 type recombinant monoclonal antibody directed against FXIIa that is administered subcutaneously. It showed an almost 99% reduction in breakthrough attacks compared to placebo in a phase 2 trial.[82] KalVista has announced to anticipate the start of clinical studies with KV998086, a prophylactic oral FXIIa inhibitor in 2023.[83] [84] Other investigational drugs target PKa or its zymogen prekallikrein (PK), thereby directly preventing the cleavage of high-molecular-weight kininogen and liberation of bradykinin as well as haltering the acceleration of the kallikrein-kinin cascade by preventing the activation of more FXII by kallikrein. Donidalorsen is an antisense oligonucleotide targeted at PK that showed a significant reduction in attack rate of 90% and improvement in angioedema-related quality of life compared to placebo.[85] [86] Notably, the reduction in PKa level does not lead to changes in thrombin formation or fibrinolytic activity.[87] Investigational drugs that target PKa include sebetralstat,[88] [89] ATN-249[91] and STAR-0215.[92] Therapies targeting the BKB2R prevent angioedema by inhibiting the effects of the end-product of the contact activation cascade and kallikrein-kinin system, bradykinin. Oral BKB2R antagonist PHA-022121 is currently in development for both LTP and ODT.[93]

The field of HAE might well be revolutionary changed again in upcoming years, for several gene therapies are currently in development with the goal of curation. One of these is BMN 331, an adeno-associated virus-based gene therapy vector targeting hepatocytes to express more wild-type C1-inhibitor.[94] With NTLA-2002, the Nobel-prize winning CRISPR-Cas9 technique is used to inactivate the KLKB1 gene in hepatocytes, aiming to continuously suppress HAE activity by reducing the level of PK.[95] These gene therapies are currently studied in phase 1/2 trials in patients. Following a different approach, OTL-105 makes use of ex vivo autologous gene therapy to express the SERPING1 gene in hematopoietic stem cells and is currently studied in pre-clinical trials.[96] Furthermore, RegenxBio and Spark Therapeutics have announced to be in the early stages of development of a gene therapy for HAE.


#

Conclusion

While C1-inhibitor is an important regulator in several intravascular pathways, symptoms in HAE patients are caused by insufficient inhibition of the contact activation pathway and kallikrein-kinin system. The subsequent unpredictable and potentially life-threatening angioedema attacks have a pronounced effect on quality of life. However, the future is bright for patients like the 21-year old female in our clinical case. New, highly specific therapies have the potential to drastically reduce their attack rate, improve their safety, and enable them to lead a normal life.


#
#

Conflict of Interest

D.M.C. reports personal fees from Biocryst, personal fees from CSL, personal fees from Ionis pharmaceuticals, personal fees from KalVista, personal fees from Pharvaris, outside the submitted work.

  • References

  • 1 Aygören-Pürsün E, Magerl M, Maetzel A, Maurer M. Epidemiology of Bradykinin-mediated angioedema: a systematic investigation of epidemiological studies. Orphanet J Rare Dis 2018; 13 (01) 73
  • 2 Bork K, Meng G, Staubach P, Hardt J. Hereditary angioedema: new findings concerning symptoms, affected organs, and course. Am J Med 2006; 119 (03) 267-274
  • 3 Christiansen SC, Davis DK, Castaldo AJ, Zuraw BL. Pediatric hereditary angioedema: onset, diagnostic delay, and disease severity. Clin Pediatr (Phila) 2016; 55 (10) 935-942
  • 4 Hofman ZL, Relan A, Hack CE. Hereditary angioedema attacks: local swelling at multiple sites. Clin Rev Allergy Immunol 2016; 50 (01) 34-40
  • 5 Hahn J, Hoess A, Friedrich DT. et al. Unnecessary abdominal interventions in patients with hereditary angioedema. J Dtsch Dermatol Ges 2018; 16 (12) 1443-1449
  • 6 Landerman NS. Hereditary angioneurotic edema. I. Case reports and review of the literature. J Allergy 1962; 33: 316-329
  • 7 Minafra FG, Gonçalves TR, Alves TM, Pinto JA. The mortality from hereditary angioedema worldwide: a review of the real-world data literature. Clin Rev Allergy Immunol 2022; 62 (01) 232-239
  • 8 Maurer M, Magerl M, Betschel S. et al. The international WAO/EAACI guideline for the management of hereditary angioedema-The 2021 revision and update. Allergy 2022; 77 (07) 1961-1990
  • 9 Caballero T, Farkas H, Bouillet L. et al; C-1-INH Deficiency Working Group. International consensus and practical guidelines on the gynecologic and obstetric management of female patients with hereditary angioedema caused by C1 inhibitor deficiency. J Allergy Clin Immunol 2012; 129 (02) 308-320
  • 10 Zotter Z, Csuka D, Szabó E. et al. The influence of trigger factors on hereditary angioedema due to C1-inhibitor deficiency. Orphanet J Rare Dis 2014; 9: 44
  • 11 Caballero T, Maurer M, Longhurst HJ, Aberer W, Bouillet L, Fabien V. IOS Study Group. Triggers and prodromal symptoms of angioedema attacks in patients with hereditary angioedema. J Investig Allergol Clin Immunol 2016; 26 (06) 383-386
  • 12 Magerl M, Doumoulakis G, Kalkounou I. et al. Characterization of prodromal symptoms in a large population of patients with hereditary angio-oedema. Clin Exp Dermatol 2014; 39 (03) 298-303
  • 13 Reshef A, Prematta MJ, Craig TJ. Signs and symptoms preceding acute attacks of hereditary angioedema: results of three recent surveys. Allergy Asthma Proc 2013; 34 (03) 261-266
  • 14 Leibovich-Nassi I, Reshef A, Somech R, Golander H. Prodromes as predictors of hereditary angioedema attacks. Allergy 2022; 77 (04) 1309-1312
  • 15 Fouche AS, Saunders EF, Craig T. Depression and anxiety in patients with hereditary angioedema. Ann Allergy Asthma Immunol 2014; 112 (04) 371-375
  • 16 Caballero T, Aygören-Pürsün E, Bygum A. et al. The humanistic burden of hereditary angioedema: results from the Burden of Illness Study in Europe. Allergy Asthma Proc 2014; 35 (01) 47-53
  • 17 Banerji A, Davis KH, Brown TM. et al. Patient-reported burden of hereditary angioedema: findings from a patient survey in the United States. Ann Allergy Asthma Immunol 2020; 124 (06) 600-607
  • 18 Savarese L, Mormile I, Bova M. et al. Psychology and hereditary angioedema: a systematic review. Allergy Asthma Proc 2021; 42 (01) e1-e7
  • 19 Fijen LM, Petersen RS, Levi M, Lakeman P, Henneman L, Cohn DM. Patient perspectives on reproductive options for hereditary angioedema: a cross-sectional survey study. J Allergy Clin Immunol Pract 2022; 10 (09) 2483-2486.e1
  • 20 Levi M, Cohn DM. The role of complement in hereditary angioedema. Transfus Med Rev 2019; 33 (04) 243-247
  • 21 Zeerleder S, Levi M. Hereditary and acquired C1-inhibitor-dependent angioedema: from pathophysiology to treatment. Ann Med 2016; 48 (04) 256-267
  • 22 Busse PJ, Christiansen SC. Hereditary angioedema. N Engl J Med 2020; 382 (12) 1136-1148
  • 23 Bossi F, Fischetti F, Regoli D. et al. Novel pathogenic mechanism and therapeutic approaches to angioedema associated with C1 inhibitor deficiency. J Allergy Clin Immunol 2009; 124 (06) 1303-10.e4
  • 24 Kaplan AP, Joseph K. Pathogenic mechanisms of bradykinin mediated diseases: dysregulation of an innate inflammatory pathway. Adv Immunol 2014; 121: 41-89
  • 25 Wuillemin WA, Minnema M, Meijers JC. et al. Inactivation of factor XIa in human plasma assessed by measuring factor XIa-protease inhibitor complexes: major role for C1-inhibitor. Blood 1995; 85 (06) 1517-1526
  • 26 Levi M, Roem D, Kamp AM, de Boer JP, Hack CE, ten Cate JW. Assessment of the relative contribution of different protease inhibitors to the inhibition of plasmin in vivo. Thromb Haemost 1993; 69 (02) 141-146
  • 27 van Geffen M, Cugno M, Lap P, Loof A, Cicardi M, van Heerde W. Alterations of coagulation and fibrinolysis in patients with angioedema due to C1-inhibitor deficiency. Clin Exp Immunol 2012; 167 (03) 472-478
  • 28 Joseph K, Tholanikunnel TE, Kaplan AP. Treatment of episodes of hereditary angioedema with C1 inhibitor: serial assessment of observed abnormalities of the plasma bradykinin-forming pathway and fibrinolysis. Ann Allergy Asthma Immunol 2010; 104 (01) 50-54
  • 29 Relan A, Bakhtiari K, van Amersfoort ES, Meijers JC, Hack CE. Recombinant C1-inhibitor: effects on coagulation and fibrinolysis in patients with hereditary angioedema. BioDrugs 2012; 26 (01) 43-52
  • 30 Reshef A, Zanichelli A, Longhurst H, Relan A, Hack CE. Elevated D-dimers in attacks of hereditary angioedema are not associated with increased thrombotic risk. Allergy 2015; 70 (05) 506-513
  • 31 de Maat S, Joseph K, Maas C, Kaplan AP. Blood clotting and the pathogenesis of types I and II hereditary angioedema. Clin Rev Allergy Immunol 2021; 60 (03) 348-356
  • 32 Matsushita M, Thiel S, Jensenius JC, Terai I, Fujita T. Proteolytic activities of two types of mannose-binding lectin-associated serine protease. J Immunol 2000; 165 (05) 2637-2642
  • 33 Davis III AE, Mejia P, Lu F. Biological activities of C1 inhibitor. Mol Immunol 2008; 45 (16) 4057-4063
  • 34 Triggianese P, Chimenti MS, Toubi E. et al. The autoimmune side of hereditary angioedema: insights on the pathogenesis. Autoimmun Rev 2015; 14 (08) 665-669
  • 35 Gösswein T, Kocot A, Emmert G. et al. Mutational spectrum of the C1INH (SERPING1) gene in patients with hereditary angioedema. Cytogenet Genome Res 2008; 121 (3-4): 181-188
  • 36 Ponard D, Gaboriaud C, Charignon D. et al. SERPING1 mutation update: mutation spectrum and C1 Inhibitor phenotypes. Hum Mutat 2020; 41 (01) 38-57 DOI: 10.1002/humu.23917.
  • 37 Yasuno S, Ansai O, Hayashi R, Nakamura S, Shimomura Y. Evidence for a dominant-negative effect of a missense mutation in the SERPING1 gene responsible for hereditary angioedema type I. J Dermatol 2021; 48 (08) 1243-1249
  • 38 Haslund D, Ryø LB, Seidelin Majidi S. et al. Dominant-negative SERPING1 variants cause intracellular retention of C1 inhibitor in hereditary angioedema. J Clin Invest 2019; 129 (01) 388-405
  • 39 Mete Gökmen N, Rodríguez-Alcalde C, Gülbahar O, Lopez-Trascasa M, Onay H, López-Lera A. Novel homozygous variants in the SERPING1 gene in two Turkish families with hereditary angioedema of recessive inheritance. Immunol Cell Biol 2020; 98 (08) 693-699
  • 40 Blanch A, Roche O, Urrutia I, Gamboa P, Fontán G, López-Trascasa M. First case of homozygous C1 inhibitor deficiency. J Allergy Clin Immunol 2006; 118 (06) 1330-1335
  • 41 López-Lera A, Favier B, de la Cruz RM, Garrido S, Drouet C, López-Trascasa M. A new case of homozygous C1-inhibitor deficiency suggests a role for Arg378 in the control of kinin pathway activation. J Allergy Clin Immunol 2010; 126 (06) 1307-10.e3
  • 42 Bafunno V, Divella C, Sessa F. et al. De novo homozygous mutation of the C1 inhibitor gene in a patient with hereditary angioedema. J Allergy Clin Immunol 2013; 132 (03) 748-750.e3
  • 43 Bork K, Machnig T, Wulff K, Witzke G, Prusty S, Hardt J. Clinical features of genetically characterized types of hereditary angioedema with normal C1 inhibitor: a systematic review of qualitative evidence. Orphanet J Rare Dis 2020; 15 (01) 289
  • 44 Kulkarni M, Travers JB, Rohan C. High estrogen states in hereditary angioedema: a spectrum. Clin Rev Allergy Immunol 2021; 60 (03) 396-403
  • 45 Gompel A, Fain O, Boccon-Gibod I, Gobert D, Bouillet L. Exogenous hormones and hereditary angioedema. Int Immunopharmacol 2020; 78: 106080
  • 46 Bork K, Wulff K, Hardt J, Witzke G, Staubach P. Hereditary angioedema caused by missense mutations in the factor XII gene: clinical features, trigger factors, and therapy. J Allergy Clin Immunol 2009; 124 (01) 129-134
  • 47 Tarzi MD, Hickey A, Förster T, Mohammadi M, Longhurst HJ. An evaluation of tests used for the diagnosis and monitoring of C1 inhibitor deficiency: normal serum C4 does not exclude hereditary angio-oedema. Clin Exp Immunol 2007; 149 (03) 513-516
  • 48 Bindke G, Gehring M, Wieczorek D, Kapp A, Buhl T, Wedi B. Identification of novel biomarkers to distinguish bradykinin-mediated angioedema from mast cell-/histamine-mediated angioedema. Allergy 2022; 77 (03) 946-955
  • 49 Porebski G, Kwitniewski M, Reshef A. Biomarkers in hereditary angioedema. Clin Rev Allergy Immunol 2021; 60 (03) 404-415
  • 50 Malbran A, Hammer CH, Frank MM, Fries LF. Acquired angioedema: observations on the mechanism of action of autoantibodies directed against C1 esterase inhibitor. J Allergy Clin Immunol 1988; 81 (06) 1199-1204
  • 51 Melamed J, Alper CA, Cicardi M, Rosen FS. The metabolism of C1 inhibitor and C1q in patients with acquired C1-inhibitor deficiency. J Allergy Clin Immunol 1986; 77 (02) 322-326
  • 52 Gobert D, Paule R, Ponard D. et al. A nationwide study of acquired C1-inhibitor deficiency in France: Characteristics and treatment responses in 92 patients. Medicine (Baltimore) 2016; 95 (33) e4363
  • 53 Pólai Z, Balla Z, Andrási N. et al. A follow-up survey of patients with acquired angioedema due to C1-inhibitor deficiency. J Intern Med 2021; 289 (04) 547-558
  • 54 Levi M, Cohn D, Zeerleder S, Dziadzio M, Longhurst H. Long-term effects upon rituximab treatment of acquired angioedema due to C1-inhibitor deficiency. Allergy 2019; 74 (04) 834-840
  • 55 Pappalardo E, Cicardi M, Duponchel C. et al. Frequent de novo mutations and exon deletions in the C1inhibitor gene of patients with angioedema. J Allergy Clin Immunol 2000; 106 (06) 1147-1154
  • 56 Farkas H, Martinez-Saguer I, Bork K. et al; HAWK. International consensus on the diagnosis and management of pediatric patients with hereditary angioedema with C1 inhibitor deficiency. Allergy 2017; 72 (02) 300-313
  • 57 Nielsen EW, Johansen HT, Holt J, Mollnes TE. C1 inhibitor and diagnosis of hereditary angioedema in newborns. Pediatr Res 1994; 35 (02) 184-187
  • 58 Agostoni A, Aygören-Pürsün E, Binkley KE. et al. Hereditary and acquired angioedema: problems and progress: proceedings of the third C1 esterase inhibitor deficiency workshop and beyond. J Allergy Clin Immunol 2004; 114 (suppl 3): S51-S131
  • 59 Maurer M, Aygören-Pürsün E, Banerji A. et al. Consensus on treatment goals in hereditary angioedema: a global Delphi initiative. J Allergy Clin Immunol 2021; 148 (06) 1526-1532
  • 60 Maurer M, Aberer W, Bouillet L. et al; IOS Investigators. Hereditary angioedema attacks resolve faster and are shorter after early icatibant treatment. PLoS One 2013; 8 (02) e53773
  • 61 Banta E, Horn P, Craig TJ. Response to ecallantide treatment of acute attacks of hereditary angioedema based on time to intervention: results from the EDEMA clinical trials. Allergy Asthma Proc 2011; 32 (04) 319-324
  • 62 Craig TJ, Rojavin MA, Machnig T, Keinecke HO, Bernstein JA. Effect of time to treatment on response to C1 esterase inhibitor concentrate for hereditary angioedema attacks. Ann Allergy Asthma Immunol 2013; 111 (03) 211-215
  • 63 Gadek JE, Hosea SW, Gelfand JA. et al. Replacement therapy in hereditary angioedema: successful treatment of acute episodes of angioedema with partly purified C1 inhibitor. N Engl J Med 1980; 302 (10) 542-546
  • 64 Levi M, Choi G, Picavet C, Hack CE. Self-administration of C1-inhibitor concentrate in patients with hereditary or acquired angioedema caused by C1-inhibitor deficiency. J Allergy Clin Immunol 2006; 117 (04) 904-908
  • 65 Riedl MA, Bygum A, Lumry W. et al; Berinert Registry investigators. Safety and usage of C1-inhibitor in hereditary angioedema: Berinert Registry Data. J Allergy Clin Immunol Pract 2016; 4 (05) 963-971
  • 66 Burnham K, Reinert JP. Thromboembolic risk of C1 esterase inhibitors: a systematic review on current evidence. Expert Rev Clin Pharmacol 2020; 13 (07) 779-786
  • 67 Craig TJ, Bewtra AK, Bahna SL. et al. C1 esterase inhibitor concentrate in 1085 hereditary angioedema attacks–final results of the I.M.P.A.C.T.2 study. Allergy 2011; 66 (12) 1604-1611
  • 68 Lumry WR, Li HH, Levy RJ. et al. Randomized placebo-controlled trial of the bradykinin B2 receptor antagonist icatibant for the treatment of acute attacks of hereditary angioedema: the FAST-3 trial. Ann Allergy Asthma Immunol 2011; 107 (06) 529-537
  • 69 Levy RJ, Lumry WR, McNeil DL. et al. EDEMA4: a phase 3, double-blind study of subcutaneous ecallantide treatment for acute attacks of hereditary angioedema. Ann Allergy Asthma Immunol 2010; 104 (06) 523-529
  • 70 Craig TJ, Li HH, Riedl M. et al. Characterization of anaphylaxis after ecallantide treatment of hereditary angioedema attacks. J Allergy Clin Immunol Pract 2015; 3 (02) 206-212.e4
  • 71 Jaffe CJ, Atkinson JP, Gelfand JA, Frank MM. Hereditary angioedema: the use of fresh frozen plasma for prophylaxis in patients undergoing oral surgery. J Allergy Clin Immunol 1975; 55 (06) 386-393
  • 72 Pickering RJ, Good RA, Kelly JR, Gewurz H. Replacement therapy in hereditary angioedema. Successful treatment of two patients with fresh frozen plasma. Lancet 1969; 1 (7590): 326-330
  • 73 Wentzel N, Panieri A, Ayazi M. et al. Fresh frozen plasma for on-demand hereditary angioedema treatment in South Africa and Iran. World Allergy Organ J 2019; 12 (09) 100049
  • 74 Craig T, Zuraw B, Longhurst H. et al; COMPACT Investigators. Long-term outcomes with subcutaneous c1-inhibitor replacement therapy for prevention of hereditary angioedema attacks. J Allergy Clin Immunol Pract 2019; 7 (06) 1793-1802.e2
  • 75 Lumry WR, Craig T, Zuraw B. et al. Health-related quality of life with subcutaneous C1-inhibitor for prevention of attacks of hereditary angioedema. J Allergy Clin Immunol Pract 2018; 6 (05) 1733-1741.e3
  • 76 Banerji A, Riedl MA, Bernstein JA. et al; HELP Investigators. Effect of Lanadelumab compared with placebo on prevention of hereditary angioedema attacks: a randomized clinical trial. JAMA 2018; 320 (20) 2108-2121
  • 77 Zuraw B, Lumry WR, Johnston DT. et al. Oral once-daily berotralstat for the prevention of hereditary angioedema attacks: a randomized, double-blind, placebo-controlled phase 3 trial. J Allergy Clin Immunol 2021; 148 (01) 164-172.e9
  • 78 Gelfand JA, Sherins RJ, Alling DW, Frank MM. Treatment of hereditary angioedema with danazol. Reversal of clinical and biochemical abnormalities. N Engl J Med 1976; 295 (26) 1444-1448
  • 79 Pappalardo E, Zingale LC, Cicardi M. Increased expression of C1-inhibitor mRNA in patients with hereditary angioedema treated with Danazol. Immunol Lett 2003; 86 (03) 271-276
  • 80 Drouet C, Désormeaux A, Robillard J. et al. Metallopeptidase activities in hereditary angioedema: effect of androgen prophylaxis on plasma aminopeptidase P. J Allergy Clin Immunol 2008; 121 (02) 429-433
  • 81 Bork K, Bygum A, Hardt J. Benefits and risks of danazol in hereditary angioedema: a long-term survey of 118 patients. Ann Allergy Asthma Immunol 2008; 100 (02) 153-161
  • 82 Craig T, Magerl M, Levy DS. et al. Prophylactic use of an anti-activated factor XII monoclonal antibody, garadacimab, for patients with C1-esterase inhibitor-deficient hereditary angioedema: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2022; 399 (10328): 945-955
  • 83 KalVista. Factor XIIa. 2022 . Accessed June 24, 2022, at: https://www.kalvista.com/products-pipeline/factor-xiia
  • 84 Clermont A. Identification of novel potent oral Factor XIIa inhibitors that block kallikrein kinin system activation in a preclinical model of angioedema. Conference session presented at: EAACI Hybrid Congress; 1–3 July 2022 2022; Prague
  • 85 Fijen LM, Riedl MA, Bordone L. et al. Inhibition of Prekallikrein for hereditary angioedema. N Engl J Med 2022; 386 (11) 1026-1033
  • 86 OASIS-HAE. A Study to Evaluate the Safety and Efficacy of Donidalorsen (ISIS 721744 or IONIS-PKK-LRx) in Participants with Hereditary Angioedema (HAE),. at: https://ClinicalTrials.gov/show/NCT05139810
  • 87 Fijen LM, Petersen RS, Meijers JCM, Bordone L, Levi M, Cohn DM. The influence of plasma prekallikrein oligonucleotide antisense therapy on coagulation and fibrinolysis assays: a post-hoc analysis. Thromb Haemost 17 Aug 2022 (e-pub ahead of print). DOI: 10.1055/a-1926-2367.
  • 88 Maetzel A, Smith MD, Duckworth EJ. et al. KVD900, an oral on-demand treatment for hereditary angioedema: phase 1 study results. J Allergy Clin Immunol 2022; 149 (06) 2034-2042
  • 89 A Phase III, Crossover Trial Evaluating the Efficacy and Safety of KVD900 for On-Demand Treatment of Angioedema Attacks in Adolescent and Adult Patients With Hereditary Angioedema (HAE). Accessed 5 July 2022, at: https://ClinicalTrials.gov/show/NCT05259917
  • 90 A Trial to Evaluate the Efficacy and Safety of Different Doses of KVD824 for Prophylactic Treatment of HAE Type I or II,. Accessed 5 July 2022, at: https://ClinicalTrials.gov/show/NCT05055258
  • 91 Hofman ZLM, Clark CC, Hack CE, de Maat S, Maas C. Detecting oral kallikrein-targeting therapy through triggered contact activation: A phase I study. J Allergy Clin Immunol 2020; 146 (06) 1446-1449.e5
  • 92 Bista P, Nichols A. STAR-0215 Is a Long-Acting Monoclonal Antibody Plasma Kallikrein Inhibitor in Development for Treatment of HAE. Poster presentation presented at: American Academy of Allergy, Asthma & Immunology Annual Scientific Virtual Meeting. 2022 ; Phoenix, AZ. https://astriatx.com/pdfs/Poster-02-25-22-Astria-AAAAI.pdf
  • 93 Crabbé R. PHA-022121: a novel and potent bradykinin 2 receptor antagonist for oral treatment of hereditary angioedema. J Allergy Clin Immunol 2021; 147 (02) AB242
  • 94 A Gene Therapy Study of BMN 331 in Subjects With Hereditary Angioedema,. at: https://ClinicalTrials.gov/show/NCT05121376
  • 95 NTLA-2002 in Adults With Hereditary Angioedema (HAE). , Accessed 5 July 2022, at: https://ClinicalTrials.gov/show/NCT05120830
  • 96 Orchard therapeutics. Our Portfolio and Focus. 2022 . Accessed 5 July 2022, at: https://www.orchard-tx.com/approach/pipeline/
  • 97 Dewald G, Bork K. Missense mutations in the coagulation factor XII (Hageman factor) gene in hereditary angioedema with normal C1 inhibitor. Biochem Biophys Res Commun 2006; 343 (04) 1286-1289
  • 98 Bork K, Wulff K, Meinke P, Wagner N, Hardt J, Witzke G. A novel mutation in the coagulation factor 12 gene in subjects with hereditary angioedema and normal C1-inhibitor. Clin Immunol 2011; 141 (01) 31-35
  • 99 Kiss N, Barabás E, Várnai K. et al. Novel duplication in the F12 gene in a patient with recurrent angioedema. Clin Immunol 2013; 149 (01) 142-145
  • 100 Bafunno V, Firinu D, D'Apolito M. et al. Mutation of the angiopoietin-1 gene (ANGPT1) associates with a new type of hereditary angioedema. J Allergy Clin Immunol 2018; 141 (03) 1009-1017
  • 101 Bork K, Wulff K, Steinmüller-Magin L. et al. Hereditary angioedema with a mutation in the plasminogen gene. Allergy 2018; 73 (02) 442-450
  • 102 Dickeson SK, Kumar S, Sun MF. et al. A mechanism for hereditary angioedema caused by a lysine 311-to-glutamic acid substitution in plasminogen. Blood 2022; 139 (18) 2816-2829
  • 103 Bork K, Wulff K, Rossmann H. et al. Hereditary angioedema cosegregating with a novel kininogen 1 gene mutation changing the N-terminal cleavage site of bradykinin. Allergy 2019; 74 (12) 2479-2481
  • 104 Ariano A, D'Apolito M, Bova M. et al. A myoferlin gain-of-function variant associates with a new type of hereditary angioedema. Allergy 2020; 75 (11) 2989-2992
  • 105 Bork K, Wulff K, Möhl BS. et al. Novel hereditary angioedema linked with a heparan sulfate 3-O-sulfotransferase 6 gene mutation. J Allergy Clin Immunol 2021; 148 (04) 1041-1048

Address for correspondence

Remy S. Petersen, MD
Department of Vascular Medicine, Amsterdam Cardiovascular Science, University of Amsterdam
Meibergdreef 9 1105 AZ Amsterdam
the Netherlands   

Publication History

Article published online:
23 November 2022

© 2022. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

  • References

  • 1 Aygören-Pürsün E, Magerl M, Maetzel A, Maurer M. Epidemiology of Bradykinin-mediated angioedema: a systematic investigation of epidemiological studies. Orphanet J Rare Dis 2018; 13 (01) 73
  • 2 Bork K, Meng G, Staubach P, Hardt J. Hereditary angioedema: new findings concerning symptoms, affected organs, and course. Am J Med 2006; 119 (03) 267-274
  • 3 Christiansen SC, Davis DK, Castaldo AJ, Zuraw BL. Pediatric hereditary angioedema: onset, diagnostic delay, and disease severity. Clin Pediatr (Phila) 2016; 55 (10) 935-942
  • 4 Hofman ZL, Relan A, Hack CE. Hereditary angioedema attacks: local swelling at multiple sites. Clin Rev Allergy Immunol 2016; 50 (01) 34-40
  • 5 Hahn J, Hoess A, Friedrich DT. et al. Unnecessary abdominal interventions in patients with hereditary angioedema. J Dtsch Dermatol Ges 2018; 16 (12) 1443-1449
  • 6 Landerman NS. Hereditary angioneurotic edema. I. Case reports and review of the literature. J Allergy 1962; 33: 316-329
  • 7 Minafra FG, Gonçalves TR, Alves TM, Pinto JA. The mortality from hereditary angioedema worldwide: a review of the real-world data literature. Clin Rev Allergy Immunol 2022; 62 (01) 232-239
  • 8 Maurer M, Magerl M, Betschel S. et al. The international WAO/EAACI guideline for the management of hereditary angioedema-The 2021 revision and update. Allergy 2022; 77 (07) 1961-1990
  • 9 Caballero T, Farkas H, Bouillet L. et al; C-1-INH Deficiency Working Group. International consensus and practical guidelines on the gynecologic and obstetric management of female patients with hereditary angioedema caused by C1 inhibitor deficiency. J Allergy Clin Immunol 2012; 129 (02) 308-320
  • 10 Zotter Z, Csuka D, Szabó E. et al. The influence of trigger factors on hereditary angioedema due to C1-inhibitor deficiency. Orphanet J Rare Dis 2014; 9: 44
  • 11 Caballero T, Maurer M, Longhurst HJ, Aberer W, Bouillet L, Fabien V. IOS Study Group. Triggers and prodromal symptoms of angioedema attacks in patients with hereditary angioedema. J Investig Allergol Clin Immunol 2016; 26 (06) 383-386
  • 12 Magerl M, Doumoulakis G, Kalkounou I. et al. Characterization of prodromal symptoms in a large population of patients with hereditary angio-oedema. Clin Exp Dermatol 2014; 39 (03) 298-303
  • 13 Reshef A, Prematta MJ, Craig TJ. Signs and symptoms preceding acute attacks of hereditary angioedema: results of three recent surveys. Allergy Asthma Proc 2013; 34 (03) 261-266
  • 14 Leibovich-Nassi I, Reshef A, Somech R, Golander H. Prodromes as predictors of hereditary angioedema attacks. Allergy 2022; 77 (04) 1309-1312
  • 15 Fouche AS, Saunders EF, Craig T. Depression and anxiety in patients with hereditary angioedema. Ann Allergy Asthma Immunol 2014; 112 (04) 371-375
  • 16 Caballero T, Aygören-Pürsün E, Bygum A. et al. The humanistic burden of hereditary angioedema: results from the Burden of Illness Study in Europe. Allergy Asthma Proc 2014; 35 (01) 47-53
  • 17 Banerji A, Davis KH, Brown TM. et al. Patient-reported burden of hereditary angioedema: findings from a patient survey in the United States. Ann Allergy Asthma Immunol 2020; 124 (06) 600-607
  • 18 Savarese L, Mormile I, Bova M. et al. Psychology and hereditary angioedema: a systematic review. Allergy Asthma Proc 2021; 42 (01) e1-e7
  • 19 Fijen LM, Petersen RS, Levi M, Lakeman P, Henneman L, Cohn DM. Patient perspectives on reproductive options for hereditary angioedema: a cross-sectional survey study. J Allergy Clin Immunol Pract 2022; 10 (09) 2483-2486.e1
  • 20 Levi M, Cohn DM. The role of complement in hereditary angioedema. Transfus Med Rev 2019; 33 (04) 243-247
  • 21 Zeerleder S, Levi M. Hereditary and acquired C1-inhibitor-dependent angioedema: from pathophysiology to treatment. Ann Med 2016; 48 (04) 256-267
  • 22 Busse PJ, Christiansen SC. Hereditary angioedema. N Engl J Med 2020; 382 (12) 1136-1148
  • 23 Bossi F, Fischetti F, Regoli D. et al. Novel pathogenic mechanism and therapeutic approaches to angioedema associated with C1 inhibitor deficiency. J Allergy Clin Immunol 2009; 124 (06) 1303-10.e4
  • 24 Kaplan AP, Joseph K. Pathogenic mechanisms of bradykinin mediated diseases: dysregulation of an innate inflammatory pathway. Adv Immunol 2014; 121: 41-89
  • 25 Wuillemin WA, Minnema M, Meijers JC. et al. Inactivation of factor XIa in human plasma assessed by measuring factor XIa-protease inhibitor complexes: major role for C1-inhibitor. Blood 1995; 85 (06) 1517-1526
  • 26 Levi M, Roem D, Kamp AM, de Boer JP, Hack CE, ten Cate JW. Assessment of the relative contribution of different protease inhibitors to the inhibition of plasmin in vivo. Thromb Haemost 1993; 69 (02) 141-146
  • 27 van Geffen M, Cugno M, Lap P, Loof A, Cicardi M, van Heerde W. Alterations of coagulation and fibrinolysis in patients with angioedema due to C1-inhibitor deficiency. Clin Exp Immunol 2012; 167 (03) 472-478
  • 28 Joseph K, Tholanikunnel TE, Kaplan AP. Treatment of episodes of hereditary angioedema with C1 inhibitor: serial assessment of observed abnormalities of the plasma bradykinin-forming pathway and fibrinolysis. Ann Allergy Asthma Immunol 2010; 104 (01) 50-54
  • 29 Relan A, Bakhtiari K, van Amersfoort ES, Meijers JC, Hack CE. Recombinant C1-inhibitor: effects on coagulation and fibrinolysis in patients with hereditary angioedema. BioDrugs 2012; 26 (01) 43-52
  • 30 Reshef A, Zanichelli A, Longhurst H, Relan A, Hack CE. Elevated D-dimers in attacks of hereditary angioedema are not associated with increased thrombotic risk. Allergy 2015; 70 (05) 506-513
  • 31 de Maat S, Joseph K, Maas C, Kaplan AP. Blood clotting and the pathogenesis of types I and II hereditary angioedema. Clin Rev Allergy Immunol 2021; 60 (03) 348-356
  • 32 Matsushita M, Thiel S, Jensenius JC, Terai I, Fujita T. Proteolytic activities of two types of mannose-binding lectin-associated serine protease. J Immunol 2000; 165 (05) 2637-2642
  • 33 Davis III AE, Mejia P, Lu F. Biological activities of C1 inhibitor. Mol Immunol 2008; 45 (16) 4057-4063
  • 34 Triggianese P, Chimenti MS, Toubi E. et al. The autoimmune side of hereditary angioedema: insights on the pathogenesis. Autoimmun Rev 2015; 14 (08) 665-669
  • 35 Gösswein T, Kocot A, Emmert G. et al. Mutational spectrum of the C1INH (SERPING1) gene in patients with hereditary angioedema. Cytogenet Genome Res 2008; 121 (3-4): 181-188
  • 36 Ponard D, Gaboriaud C, Charignon D. et al. SERPING1 mutation update: mutation spectrum and C1 Inhibitor phenotypes. Hum Mutat 2020; 41 (01) 38-57 DOI: 10.1002/humu.23917.
  • 37 Yasuno S, Ansai O, Hayashi R, Nakamura S, Shimomura Y. Evidence for a dominant-negative effect of a missense mutation in the SERPING1 gene responsible for hereditary angioedema type I. J Dermatol 2021; 48 (08) 1243-1249
  • 38 Haslund D, Ryø LB, Seidelin Majidi S. et al. Dominant-negative SERPING1 variants cause intracellular retention of C1 inhibitor in hereditary angioedema. J Clin Invest 2019; 129 (01) 388-405
  • 39 Mete Gökmen N, Rodríguez-Alcalde C, Gülbahar O, Lopez-Trascasa M, Onay H, López-Lera A. Novel homozygous variants in the SERPING1 gene in two Turkish families with hereditary angioedema of recessive inheritance. Immunol Cell Biol 2020; 98 (08) 693-699
  • 40 Blanch A, Roche O, Urrutia I, Gamboa P, Fontán G, López-Trascasa M. First case of homozygous C1 inhibitor deficiency. J Allergy Clin Immunol 2006; 118 (06) 1330-1335
  • 41 López-Lera A, Favier B, de la Cruz RM, Garrido S, Drouet C, López-Trascasa M. A new case of homozygous C1-inhibitor deficiency suggests a role for Arg378 in the control of kinin pathway activation. J Allergy Clin Immunol 2010; 126 (06) 1307-10.e3
  • 42 Bafunno V, Divella C, Sessa F. et al. De novo homozygous mutation of the C1 inhibitor gene in a patient with hereditary angioedema. J Allergy Clin Immunol 2013; 132 (03) 748-750.e3
  • 43 Bork K, Machnig T, Wulff K, Witzke G, Prusty S, Hardt J. Clinical features of genetically characterized types of hereditary angioedema with normal C1 inhibitor: a systematic review of qualitative evidence. Orphanet J Rare Dis 2020; 15 (01) 289
  • 44 Kulkarni M, Travers JB, Rohan C. High estrogen states in hereditary angioedema: a spectrum. Clin Rev Allergy Immunol 2021; 60 (03) 396-403
  • 45 Gompel A, Fain O, Boccon-Gibod I, Gobert D, Bouillet L. Exogenous hormones and hereditary angioedema. Int Immunopharmacol 2020; 78: 106080
  • 46 Bork K, Wulff K, Hardt J, Witzke G, Staubach P. Hereditary angioedema caused by missense mutations in the factor XII gene: clinical features, trigger factors, and therapy. J Allergy Clin Immunol 2009; 124 (01) 129-134
  • 47 Tarzi MD, Hickey A, Förster T, Mohammadi M, Longhurst HJ. An evaluation of tests used for the diagnosis and monitoring of C1 inhibitor deficiency: normal serum C4 does not exclude hereditary angio-oedema. Clin Exp Immunol 2007; 149 (03) 513-516
  • 48 Bindke G, Gehring M, Wieczorek D, Kapp A, Buhl T, Wedi B. Identification of novel biomarkers to distinguish bradykinin-mediated angioedema from mast cell-/histamine-mediated angioedema. Allergy 2022; 77 (03) 946-955
  • 49 Porebski G, Kwitniewski M, Reshef A. Biomarkers in hereditary angioedema. Clin Rev Allergy Immunol 2021; 60 (03) 404-415
  • 50 Malbran A, Hammer CH, Frank MM, Fries LF. Acquired angioedema: observations on the mechanism of action of autoantibodies directed against C1 esterase inhibitor. J Allergy Clin Immunol 1988; 81 (06) 1199-1204
  • 51 Melamed J, Alper CA, Cicardi M, Rosen FS. The metabolism of C1 inhibitor and C1q in patients with acquired C1-inhibitor deficiency. J Allergy Clin Immunol 1986; 77 (02) 322-326
  • 52 Gobert D, Paule R, Ponard D. et al. A nationwide study of acquired C1-inhibitor deficiency in France: Characteristics and treatment responses in 92 patients. Medicine (Baltimore) 2016; 95 (33) e4363
  • 53 Pólai Z, Balla Z, Andrási N. et al. A follow-up survey of patients with acquired angioedema due to C1-inhibitor deficiency. J Intern Med 2021; 289 (04) 547-558
  • 54 Levi M, Cohn D, Zeerleder S, Dziadzio M, Longhurst H. Long-term effects upon rituximab treatment of acquired angioedema due to C1-inhibitor deficiency. Allergy 2019; 74 (04) 834-840
  • 55 Pappalardo E, Cicardi M, Duponchel C. et al. Frequent de novo mutations and exon deletions in the C1inhibitor gene of patients with angioedema. J Allergy Clin Immunol 2000; 106 (06) 1147-1154
  • 56 Farkas H, Martinez-Saguer I, Bork K. et al; HAWK. International consensus on the diagnosis and management of pediatric patients with hereditary angioedema with C1 inhibitor deficiency. Allergy 2017; 72 (02) 300-313
  • 57 Nielsen EW, Johansen HT, Holt J, Mollnes TE. C1 inhibitor and diagnosis of hereditary angioedema in newborns. Pediatr Res 1994; 35 (02) 184-187
  • 58 Agostoni A, Aygören-Pürsün E, Binkley KE. et al. Hereditary and acquired angioedema: problems and progress: proceedings of the third C1 esterase inhibitor deficiency workshop and beyond. J Allergy Clin Immunol 2004; 114 (suppl 3): S51-S131
  • 59 Maurer M, Aygören-Pürsün E, Banerji A. et al. Consensus on treatment goals in hereditary angioedema: a global Delphi initiative. J Allergy Clin Immunol 2021; 148 (06) 1526-1532
  • 60 Maurer M, Aberer W, Bouillet L. et al; IOS Investigators. Hereditary angioedema attacks resolve faster and are shorter after early icatibant treatment. PLoS One 2013; 8 (02) e53773
  • 61 Banta E, Horn P, Craig TJ. Response to ecallantide treatment of acute attacks of hereditary angioedema based on time to intervention: results from the EDEMA clinical trials. Allergy Asthma Proc 2011; 32 (04) 319-324
  • 62 Craig TJ, Rojavin MA, Machnig T, Keinecke HO, Bernstein JA. Effect of time to treatment on response to C1 esterase inhibitor concentrate for hereditary angioedema attacks. Ann Allergy Asthma Immunol 2013; 111 (03) 211-215
  • 63 Gadek JE, Hosea SW, Gelfand JA. et al. Replacement therapy in hereditary angioedema: successful treatment of acute episodes of angioedema with partly purified C1 inhibitor. N Engl J Med 1980; 302 (10) 542-546
  • 64 Levi M, Choi G, Picavet C, Hack CE. Self-administration of C1-inhibitor concentrate in patients with hereditary or acquired angioedema caused by C1-inhibitor deficiency. J Allergy Clin Immunol 2006; 117 (04) 904-908
  • 65 Riedl MA, Bygum A, Lumry W. et al; Berinert Registry investigators. Safety and usage of C1-inhibitor in hereditary angioedema: Berinert Registry Data. J Allergy Clin Immunol Pract 2016; 4 (05) 963-971
  • 66 Burnham K, Reinert JP. Thromboembolic risk of C1 esterase inhibitors: a systematic review on current evidence. Expert Rev Clin Pharmacol 2020; 13 (07) 779-786
  • 67 Craig TJ, Bewtra AK, Bahna SL. et al. C1 esterase inhibitor concentrate in 1085 hereditary angioedema attacks–final results of the I.M.P.A.C.T.2 study. Allergy 2011; 66 (12) 1604-1611
  • 68 Lumry WR, Li HH, Levy RJ. et al. Randomized placebo-controlled trial of the bradykinin B2 receptor antagonist icatibant for the treatment of acute attacks of hereditary angioedema: the FAST-3 trial. Ann Allergy Asthma Immunol 2011; 107 (06) 529-537
  • 69 Levy RJ, Lumry WR, McNeil DL. et al. EDEMA4: a phase 3, double-blind study of subcutaneous ecallantide treatment for acute attacks of hereditary angioedema. Ann Allergy Asthma Immunol 2010; 104 (06) 523-529
  • 70 Craig TJ, Li HH, Riedl M. et al. Characterization of anaphylaxis after ecallantide treatment of hereditary angioedema attacks. J Allergy Clin Immunol Pract 2015; 3 (02) 206-212.e4
  • 71 Jaffe CJ, Atkinson JP, Gelfand JA, Frank MM. Hereditary angioedema: the use of fresh frozen plasma for prophylaxis in patients undergoing oral surgery. J Allergy Clin Immunol 1975; 55 (06) 386-393
  • 72 Pickering RJ, Good RA, Kelly JR, Gewurz H. Replacement therapy in hereditary angioedema. Successful treatment of two patients with fresh frozen plasma. Lancet 1969; 1 (7590): 326-330
  • 73 Wentzel N, Panieri A, Ayazi M. et al. Fresh frozen plasma for on-demand hereditary angioedema treatment in South Africa and Iran. World Allergy Organ J 2019; 12 (09) 100049
  • 74 Craig T, Zuraw B, Longhurst H. et al; COMPACT Investigators. Long-term outcomes with subcutaneous c1-inhibitor replacement therapy for prevention of hereditary angioedema attacks. J Allergy Clin Immunol Pract 2019; 7 (06) 1793-1802.e2
  • 75 Lumry WR, Craig T, Zuraw B. et al. Health-related quality of life with subcutaneous C1-inhibitor for prevention of attacks of hereditary angioedema. J Allergy Clin Immunol Pract 2018; 6 (05) 1733-1741.e3
  • 76 Banerji A, Riedl MA, Bernstein JA. et al; HELP Investigators. Effect of Lanadelumab compared with placebo on prevention of hereditary angioedema attacks: a randomized clinical trial. JAMA 2018; 320 (20) 2108-2121
  • 77 Zuraw B, Lumry WR, Johnston DT. et al. Oral once-daily berotralstat for the prevention of hereditary angioedema attacks: a randomized, double-blind, placebo-controlled phase 3 trial. J Allergy Clin Immunol 2021; 148 (01) 164-172.e9
  • 78 Gelfand JA, Sherins RJ, Alling DW, Frank MM. Treatment of hereditary angioedema with danazol. Reversal of clinical and biochemical abnormalities. N Engl J Med 1976; 295 (26) 1444-1448
  • 79 Pappalardo E, Zingale LC, Cicardi M. Increased expression of C1-inhibitor mRNA in patients with hereditary angioedema treated with Danazol. Immunol Lett 2003; 86 (03) 271-276
  • 80 Drouet C, Désormeaux A, Robillard J. et al. Metallopeptidase activities in hereditary angioedema: effect of androgen prophylaxis on plasma aminopeptidase P. J Allergy Clin Immunol 2008; 121 (02) 429-433
  • 81 Bork K, Bygum A, Hardt J. Benefits and risks of danazol in hereditary angioedema: a long-term survey of 118 patients. Ann Allergy Asthma Immunol 2008; 100 (02) 153-161
  • 82 Craig T, Magerl M, Levy DS. et al. Prophylactic use of an anti-activated factor XII monoclonal antibody, garadacimab, for patients with C1-esterase inhibitor-deficient hereditary angioedema: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2022; 399 (10328): 945-955
  • 83 KalVista. Factor XIIa. 2022 . Accessed June 24, 2022, at: https://www.kalvista.com/products-pipeline/factor-xiia
  • 84 Clermont A. Identification of novel potent oral Factor XIIa inhibitors that block kallikrein kinin system activation in a preclinical model of angioedema. Conference session presented at: EAACI Hybrid Congress; 1–3 July 2022 2022; Prague
  • 85 Fijen LM, Riedl MA, Bordone L. et al. Inhibition of Prekallikrein for hereditary angioedema. N Engl J Med 2022; 386 (11) 1026-1033
  • 86 OASIS-HAE. A Study to Evaluate the Safety and Efficacy of Donidalorsen (ISIS 721744 or IONIS-PKK-LRx) in Participants with Hereditary Angioedema (HAE),. at: https://ClinicalTrials.gov/show/NCT05139810
  • 87 Fijen LM, Petersen RS, Meijers JCM, Bordone L, Levi M, Cohn DM. The influence of plasma prekallikrein oligonucleotide antisense therapy on coagulation and fibrinolysis assays: a post-hoc analysis. Thromb Haemost 17 Aug 2022 (e-pub ahead of print). DOI: 10.1055/a-1926-2367.
  • 88 Maetzel A, Smith MD, Duckworth EJ. et al. KVD900, an oral on-demand treatment for hereditary angioedema: phase 1 study results. J Allergy Clin Immunol 2022; 149 (06) 2034-2042
  • 89 A Phase III, Crossover Trial Evaluating the Efficacy and Safety of KVD900 for On-Demand Treatment of Angioedema Attacks in Adolescent and Adult Patients With Hereditary Angioedema (HAE). Accessed 5 July 2022, at: https://ClinicalTrials.gov/show/NCT05259917
  • 90 A Trial to Evaluate the Efficacy and Safety of Different Doses of KVD824 for Prophylactic Treatment of HAE Type I or II,. Accessed 5 July 2022, at: https://ClinicalTrials.gov/show/NCT05055258
  • 91 Hofman ZLM, Clark CC, Hack CE, de Maat S, Maas C. Detecting oral kallikrein-targeting therapy through triggered contact activation: A phase I study. J Allergy Clin Immunol 2020; 146 (06) 1446-1449.e5
  • 92 Bista P, Nichols A. STAR-0215 Is a Long-Acting Monoclonal Antibody Plasma Kallikrein Inhibitor in Development for Treatment of HAE. Poster presentation presented at: American Academy of Allergy, Asthma & Immunology Annual Scientific Virtual Meeting. 2022 ; Phoenix, AZ. https://astriatx.com/pdfs/Poster-02-25-22-Astria-AAAAI.pdf
  • 93 Crabbé R. PHA-022121: a novel and potent bradykinin 2 receptor antagonist for oral treatment of hereditary angioedema. J Allergy Clin Immunol 2021; 147 (02) AB242
  • 94 A Gene Therapy Study of BMN 331 in Subjects With Hereditary Angioedema,. at: https://ClinicalTrials.gov/show/NCT05121376
  • 95 NTLA-2002 in Adults With Hereditary Angioedema (HAE). , Accessed 5 July 2022, at: https://ClinicalTrials.gov/show/NCT05120830
  • 96 Orchard therapeutics. Our Portfolio and Focus. 2022 . Accessed 5 July 2022, at: https://www.orchard-tx.com/approach/pipeline/
  • 97 Dewald G, Bork K. Missense mutations in the coagulation factor XII (Hageman factor) gene in hereditary angioedema with normal C1 inhibitor. Biochem Biophys Res Commun 2006; 343 (04) 1286-1289
  • 98 Bork K, Wulff K, Meinke P, Wagner N, Hardt J, Witzke G. A novel mutation in the coagulation factor 12 gene in subjects with hereditary angioedema and normal C1-inhibitor. Clin Immunol 2011; 141 (01) 31-35
  • 99 Kiss N, Barabás E, Várnai K. et al. Novel duplication in the F12 gene in a patient with recurrent angioedema. Clin Immunol 2013; 149 (01) 142-145
  • 100 Bafunno V, Firinu D, D'Apolito M. et al. Mutation of the angiopoietin-1 gene (ANGPT1) associates with a new type of hereditary angioedema. J Allergy Clin Immunol 2018; 141 (03) 1009-1017
  • 101 Bork K, Wulff K, Steinmüller-Magin L. et al. Hereditary angioedema with a mutation in the plasminogen gene. Allergy 2018; 73 (02) 442-450
  • 102 Dickeson SK, Kumar S, Sun MF. et al. A mechanism for hereditary angioedema caused by a lysine 311-to-glutamic acid substitution in plasminogen. Blood 2022; 139 (18) 2816-2829
  • 103 Bork K, Wulff K, Rossmann H. et al. Hereditary angioedema cosegregating with a novel kininogen 1 gene mutation changing the N-terminal cleavage site of bradykinin. Allergy 2019; 74 (12) 2479-2481
  • 104 Ariano A, D'Apolito M, Bova M. et al. A myoferlin gain-of-function variant associates with a new type of hereditary angioedema. Allergy 2020; 75 (11) 2989-2992
  • 105 Bork K, Wulff K, Möhl BS. et al. Novel hereditary angioedema linked with a heparan sulfate 3-O-sulfotransferase 6 gene mutation. J Allergy Clin Immunol 2021; 148 (04) 1041-1048

Zoom Image
Fig. 1 Degradation of bradykinin. Bradykinin binds to the bradykinin B2 receptor (B2R) and is transformed to des-arg9-bradykinin by carboxypeptidase N and M (CPN/CPM), which in turn binds to the bradykinin B1 receptor (B1R). Bradykinin is principally degraded by angiotensin converting enzyme (ACE), while angiotensin converting enzyme 2 (ACE2) has a higher affinity for des-arg9-bradykinin. Both bradykinin and des-arg9-bradykinin are broken down into inactive parts by aminopeptidase P (APP), dipeptidyl peptidase IV (DPP-IV), and neutral endopeptidase (NEP).
Zoom Image
Fig. 2 Functions of C1-inhibitor. C1-inhibitor (C1-INH) is a crucial regulator of several intravascular cascades, including the contact-activation system and kallikrein-kinin pathway. The contact-activation system starts when factor XII (FXII) is converted in active factor XII (FXIIa), illustrated by a solid arrow. The activation of plasma prekallikrein (PK) in plasma kallikrein (PKa) is subsequently stimulated by FXIIa, as illustrated by a dotted arrow. Kallikrein, in turn, induces further activation of FXII and facilitates the cleavage of high-molecular weight kininogen (HK) into cleaved high-molecular weight kininogen (cHK) and bradykinin, which in turn binds to the bradykinin B2 receptor (B2R), resulting in the efflux of fluid to extravasal tissue. In the coagulation system, C1-INH inhibits both the activation of factor XI (FXI) and the function of active FXI (FXIa). In the fibrinolytic pathway, kallikrein induces the activation of plasminogen (Plg) to plasmin (Pl) directly and indirectly through the stimulation of urokinase plasminogen activator (uPA). C1-INH inhibits these functions of kallikrein as well as the direct effects of plasmin on fibrin degradation. In the complement system, both the classical pathway, involving complement 1 complex (C1 complex), complement 2 (C2), and complement 4 (C4, as well as the lectin pathway involving mannose-associated serine protease 1 and 2 (MASP-1/2) are inhibited by C1-INH.