3,054
Views
0
CrossRef citations to date
0
Altmetric
Clinical feature - Review

The importance of recognizing and managing a rare form of angioedema: hereditary angioedema due to C1-inhibitor deficiency

ORCID Icon &
Pages 639-650 | Received 04 Dec 2020, Accepted 16 Mar 2021, Published online: 06 Jul 2021

ABSTRACT

The majority of angioedema cases encountered in clinical practice are histamine-mediated (allergic); however, some cases are bradykinin-related (non-allergic) and do not respond to standard anti-allergy medications. Among bradykinin-related angioedema, hereditary angioedema (HAE) is a rare, but chronic and debilitating condition. The majority of HAE is caused by deficiency (type 1) or abnormal function (type 2) of the naturally occurring protein, C1-inhibitor (C1-INH)—a major inhibitor of proteases in the contact (kallikrein-bradykinin cascade), fibrinolytic pathway, and complement systems. Failure to recognize HAE and initiate appropriate intervention can lead to years of pain, disability, impaired quality of life (QoL) and, in cases of laryngeal involvement, it can be life-threatening. HAE must be considered in the differential diagnosis of non-urticarial angioedema, particularly for patients with a history of recurrent angioedema attacks, family history of HAE, symptom onset in childhood/adolescence, prodromal signs/symptoms before swellings, recurrent/painful abdominal symptoms, and upper airway edema. Management strategies for HAE include on-demand treatment for acute attacks, short-term prophylaxis prior to attack-triggering events/procedures, and long-term or routine prophylaxis for attack prevention. Patients should be evaluated at least annually to assess need for routine prophylaxis. HAE specific medications like plasma-derived and recombinant C1-INH products, kallikrein inhibitors, and bradykinin B2 receptor antagonists, have improved management of HAE. While the introduction of intravenous C1-INH represented a major breakthrough in routine HAE prophylaxis, some patients fail to achieve adequate control and others have psychological barriers or experience complications related to intravenous administration. Subcutaneous (SC) C1-INH, SC monoclonal antibody (mAb)-based therapies, and an oral kallikrein inhibitor offer effective alternatives for HAE attack prevention and may facilitate self-administration. HAE management should be individualized, with QoL improvement being a key goal. This can be achieved with broader availability of existing options for routine prophylaxis, including greater global availability of C1-INH(SC), mAb-based therapy, oral treatments, and multiple on-demand therapies.

1. Introduction

Angioedema is commonly encountered by general medicine practitioners. Etiologies of angioedema vary and differential diagnosis can be critical for instituting effective treatment. Typically characterized by tissue swelling resulting from increased vascular permeability in deeper layers of skin and mucous membranes [Citation1–3], the majority of angioedema cases are histamine- (mast cell) mediated; however, the etiology can also be bradykinin-related (). Histamine-mediated, or allergic angioedema, is a response to external allergens, such as food, insect stings, and drugs, or it may be idiopathic [Citation5]. Allergic angioedema may be associated with urticaria, flushing, generalized pruritus, bronchospasm, throat tightness, and hypotension, and respond to treatment with conventional anti-allergy medications (eg, antihistamines, glucocorticoids, and epinephrine) [Citation6]. In patients presenting with acute allergic angioedema, attention should be given to the signs and symptoms of anaphylaxis and prompt administration of epinephrine should be provided if appropriate. Bradykinin-related, or non-allergic angioedema, may be acquired (ie, related to drugs such as angiotensin-converting enzyme [ACE] inhibitors or underlying lymphoreticular dyscrasias) or inherited (hereditary angioedema [HAE]) [Citation7–9].

Table 1. Typical clinical and laboratory characteristics of various types of angioedema disorders.a

HAE is a relatively rare form of non-allergic angioedema, and it is associated with major morbidity and potential mortality [Citation10,Citation11]. While HAE may have similar manifestations as other types of angioedema, there is a notable absence of urticaria [Citation12,Citation13]. In addition, HAE requires a different treatment approach than allergic angioedema since antihistamines, glucocorticoids, and epinephrine are not effective for HAE [Citation6]. Unfortunately, misdiagnosis of HAE is prevalent [Citation14]. Historically, misunderstanding of symptoms and low awareness of HAE has contributed to misdiagnosis and delayed diagnoses [Citation12]. Given the unique nature of HAE in relation to other types of angioedema, the potential for adverse consequences with incorrect or delayed diagnosis, and the availability of highly effective treatment options for HAE, it is imperative to consider HAE in the differential diagnosis of angioedema [Citation10,Citation12].

This article presents an illustrative case study of HAE, provides an overview of epidemiology and pathophysiology, reviews guidelines for diagnosis and management, and describes current and emerging treatment options.

2. Case

A 14-year-old female was admitted to the hospital with severe abdominal pain and nausea. She was evaluated by the surgical service with a suspicion of acute appendicitis. Abdominal ultrasound was nonspecific except for some edematous loops of bowel. D-Dimer was elevated. While being prepped for the operating room, her mother mentions that her father suffered from recurrent severe abdominal pain and had been diagnosed with a rare condition that causes swelling. The astute resident considers HAE in the differential diagnosis and orders a dose of intravenous (IV) plasma-derived C1-inhibitor (C1-INH) at 20 units/kg along with a C4 level, C1-INH functional activity, and C1-INH level. Within 30 minutes her abdominal pain is resolving, and continues to subside over the next several hours. The operation is canceled. Results for the C4, C1-INH level, and C1-INH function all come back low, confirming a diagnosis of type-I HAE.

3. Overview of HAE

3.1. Epidemiology and types of HAE

HAE is a genetic disorder that affects anywhere between 1:100,000 to 1:10,000 individuals [Citation11,Citation15,Citation16]. HAE is characterized by recurrent episodes of localized subcutaneous (SC) or submucosal nonpruritic swelling [Citation15,Citation17]. Endotypic variants of HAE include C1-INH-HAE type 1, which is characterized by a deficiency of the serine protease inhibitor, C1-INH, and accounts for approximately 85% of cases [Citation15,Citation18,Citation19]. Less common, C1-INH-HAE type 2 is characterized by abnormal C1-INH function in the presence of normal or elevated C1-INH levels, and accounts for approximately 15% of cases. Clinically these endotypes are identical in presentation and response to therapy. They are distinguished biochemically by a quantitative C1-INH level. A third, rare, type of HAE is characterized by recurrent angioedema in the absence of urticaria despite normal C1-INH; this variation has been associated with various genetic mutations, including mutations in genes encoding for factor XII [Citation20–22], angiopoetin-1 [Citation23], kininogen-1 [Citation22,Citation24], plasminogen [Citation25], myoferlin [Citation26], and heparan sulfate glucosamine 3-O-sulfotransferase-6 [Citation27]; many cases are considered idiopathic in the absence of relevant genetic mutation findings. It should be noted that other than factor XII, genetic screening for these other known mutations is not widely available to clinicians, and a specific mutation does not have to be identified in order to diagnose a patients with HAE with normal C1-INH. Given the lower prevalence of this latter type of HAE and dearth of clinical information, for the remainder of this review the term ‘HAE’ denotes C1-INH-HAE types 1 and 2.

3.2. Pathophysiology of HAE

C1-inhibitor is a naturally occurring protein that serves as a major inhibitor of proteases in the contact system (ultimately regulating the downstream production of bradykinin) and the complement system. C1-INH also inhibits proteases in the coagulation system and the fibrinolytic system, although to a lesser degree [Citation28,Citation29] ().

Figure 1. Pathway of contact system activation and interaction with fibrinolytic system. Contact system activation starts with the activation of factor XII. Activated factor XII converts plasma prekallikrein into plasma kallikrein. Kallikrein cleaves high molecular weight kininogen to produce bradykinin. Bradykinin causes vasodilatation and increased vascular permeability, leading to angioedema. The fibrinolytic system can also lead to bradykinin formation and vascular leakage via factor XII activation by plasmin. Kallikrein regulates the fibrinolytic system by cleaving pro-urokinase plasminogen activator into urokinase-type plasminogen activator, causing activation of plasminogen to plasmin. C1 inhibitor (C1-INH) regulates these pathways via inhibition (bold crosses) [Citation30].

Figure 1. Pathway of contact system activation and interaction with fibrinolytic system. Contact system activation starts with the activation of factor XII. Activated factor XII converts plasma prekallikrein into plasma kallikrein. Kallikrein cleaves high molecular weight kininogen to produce bradykinin. Bradykinin causes vasodilatation and increased vascular permeability, leading to angioedema. The fibrinolytic system can also lead to bradykinin formation and vascular leakage via factor XII activation by plasmin. Kallikrein regulates the fibrinolytic system by cleaving pro-urokinase plasminogen activator into urokinase-type plasminogen activator, causing activation of plasminogen to plasmin. C1 inhibitor (C1-INH) regulates these pathways via inhibition (bold crosses) [Citation30].

In patients with HAE, it is believed that angioedema attacks occur when the contact system is activated; active plasma kallikrein cleaves high-molecular weight kininogen, leading to release of the inflammatory mediator bradykinin [Citation28,Citation29]. The reduced levels of functional C1-INH in HAE types 1 and 2 result from mutations in the SERPING1 gene, which codes for the C1-INH protein [Citation29,Citation31]. In patients with type 1 HAE, mutations result in a failure to synthesize or secrete C1-INH, whereas in type 2 HAE, mutations result in the synthesis of dysfunctional C1-INH proteins that are unable to interact with and inhibit targeted proteases. Thus, the contact system in patients with HAE is disinhibited due to the lack of sufficient functional C1-INH. One study has suggested that lower baseline levels of functional C1-INH correlate significantly with disease severity [Citation32], although such a correlation was not found in a later study [Citation33].

3.3 Clinical manifestations and burden of HAE

Areas of the body that are most commonly affected by angioedema include the extremities, the GI tract, face, the trunk, and the genitals [Citation3,Citation17,Citation18]. The majority of patients with HAE experience recurrent abdominal attacks that are often associated with pain, vomiting, and diarrhea, and cause significant disability and distress [Citation3,Citation34]. Severe headaches, visual disturbances, and other neurological manifestations can also occur [Citation3,Citation35,Citation36]. Laryngeal involvement has the potential to result in death [Citation37,Citation38]. While laryngeal attacks may account for a small proportion of overall HAE attacks, in a survey of patients with HAE, approximately half of patients reported having had at least one laryngeal attack [Citation3]. For all types of HAE, attacks are generally unpredictable, may be acutely painful and disfiguring, and are potentially life-threatening [Citation19,Citation39,Citation40]. Attacks are often spontaneous but can be triggered by physical or emotional factors such as trauma, medical or dental procedures, infections, psychological stress, and hormonal variations including estrogen exposure [Citation41]. Many, but not all, HAE attacks are preceded by “prodromal” symptoms, which may include rash (erythema marginatum), nausea, fatigue, tight/pricking sensation in the skin, and irritability [Citation42,Citation43].

HAE may negatively impact patients’ quality of life, both during and in between episodes [Citation44–47], and is associated with elevated rates of anxiety and depression [Citation40,Citation48–50]. Attacks have been associated with missed time from work, school, and social activities, and decreased productivity [Citation40,Citation48,Citation51–54].

4. Guidelines for diagnosis and general management of HAE

4.1. Diagnosis of HAE

Prompt diagnosis and appropriate management of HAE are critical to optimize health and psychosocial outcomes; however, diagnosis of HAE is often delayed [Citation55,Citation56]. HAE symptoms may initially develop in childhood with a relatively lower frequency and severity of attacks [Citation57,Citation58]; however, patients may not seek treatment until attacks become more frequent and/or severe, which typically coincides with puberty. Consequences of delayed diagnosis, whether due to patients not seeking care or providers missing the diagnosis, may result in patients undergoing unnecessary medical procedures and spending years suffering without appropriate or effective treatment. Further, in the case of laryngeal involvement, consequences can be fatal [Citation1,Citation59,Citation60]. Young children may be at particular risk due to the smaller diameter of their airways [Citation58]. Families with a history of HAE, as well as providers who are aware of such family history, need to be proactive in surveilling for HAE in family members since a first attack could be a potentially fatal laryngeal attack. HAE guidelines recommend that family members of individuals with HAE-C1-INH type 1 or 2 be screened for C1-INH function, C1-INH protein, and C4 plasma levels [Citation1,Citation61]. Laboratory testing for HAE in young infants typically has not been advised given a high degree of variability in C4 and C1-INH antigenic levels during the first year of life [Citation61], although some evidence suggest that C1-INH functional assays may have diagnostic value in situations where testing is considered necessary in patients younger than one year of age [Citation62]. It should be noted that HAE symptoms typically begin in childhood or adolescence, rather than infancy; median age of onset is 12 years [Citation1].

Differential diagnosis between allergic (histamine-mediated) and non-allergic (bradykinin-mediated) angioedema is critical since the underlying mechanism guides appropriate treatment [Citation1,Citation7]. Bradykinin-induced angioedema can further be classified into inherited (ie, HAE) and acquired angioedema. Although both HAE and acquired angioedema (AAE) may be due to a deficiency or dysfunction of C1-inhibitor, AAE may be due to drugs (ACEI) or a complement consumptive process, which in many cases can be related to a B-cell dyscrasia (eg, monoclonal gammopathy of uncertain significance) or less commonly an autoimmune process [Citation7]. Recurrent, unexplained angioedema can sometimes be the initial presenting symptom of an underlying lymphoproliferative disease [Citation63], thus patients with diagnosed AAE should be carefully evaluated to rule out underlying malignant pathology.

There should be a high index of suspicion of HAE for patients who have a history of recurrent, otherwise unexplained angioedema attacks [Citation1]. Clinical features that may be helpful in differential diagnosis of angioedema are summarized in , and a differential diagnosis decision tree is provided in . The presence of urticaria with angioedema attacks is typically indicative of a histamine-related pathophysiology, rather than HAE. Diagnosis of HAE is also supported by the following: family history, symptom onset in childhood/adolescence, prodromal signs/symptoms before swellings, recurrent/painful abdominal symptoms, upper airway edema, absence of urticaria, and lack of response to conventional anti-allergy treatments [Citation1]. Of note, up to 25% of HAE may develop as the result of a de novo genetic mutation; therefore, a diagnosis of HAE should not be excluded based on lack of family history of angioedema [Citation7]. The onset of symptoms after age 40 should raise suspicion of AAE.

Figure 2. Diagnostic workup for patients with unexplained recurrent angioedema [Citation27,Citation42,Citation64]. Each evaluation step is shown as an oval with possible outcomes indicated with arrows. Eventual diagnoses are shown in boxes. C1-INH testing must include C1-INH functional activity if the antigenic level is normal. Response to antihistamines requires regular and sustained dosing (at least long enough for 3 expected attacks) using high dose antihistamines (at least 4 times the standard dose of second-generation non-sedating antihistamines). C1-INH, C1-inhibitor; HAE-C1-INH, hereditary angioedema due to C1-inhibitor deficiency; HAE-ANGPT1, hereditary angioedema with a specific angiopoietin-1 gene mutation (ANGPT1); HAE-FXII, hereditary angioedema with F12 mutation; HAE-HS3OST6, hereditary angioedema with a mutation coding for the heparan sulfate glucosamine 3-O-sulfotransferase-6 (3-OST-6); HAE-KNG1, hereditary angioedema with a specific kininogegn-1 gene mutation (KNG1); HAE-Myoferlin, hereditary angioedema with a specific myoferlin gene mutation (MYOF); HAE-PLG, hereditary angioedema with a specific plasminogen gene mutation (PLG); HAE-U, hereditary angioedema, unknown mutation

Figure 2. Diagnostic workup for patients with unexplained recurrent angioedema [Citation27,Citation42,Citation64]. Each evaluation step is shown as an oval with possible outcomes indicated with arrows. Eventual diagnoses are shown in boxes. C1-INH testing must include C1-INH functional activity if the antigenic level is normal. Response to antihistamines requires regular and sustained dosing (at least long enough for 3 expected attacks) using high dose antihistamines (at least 4 times the standard dose of second-generation non-sedating antihistamines). C1-INH, C1-inhibitor; HAE-C1-INH, hereditary angioedema due to C1-inhibitor deficiency; HAE-ANGPT1, hereditary angioedema with a specific angiopoietin-1 gene mutation (ANGPT1); HAE-FXII, hereditary angioedema with F12 mutation; HAE-HS3OST6, hereditary angioedema with a mutation coding for the heparan sulfate glucosamine 3-O-sulfotransferase-6 (3-OST-6); HAE-KNG1, hereditary angioedema with a specific kininogegn-1 gene mutation (KNG1); HAE-Myoferlin, hereditary angioedema with a specific myoferlin gene mutation (MYOF); HAE-PLG, hereditary angioedema with a specific plasminogen gene mutation (PLG); HAE-U, hereditary angioedema, unknown mutation

Table 2. Clinical features useful for a differential diagnosis of angioedema [modified from Citation7]

International and US guidelines recommend that all patients suspected of having HAE should undergo prompt laboratory evaluation to support diagnosis [Citation1,Citation17,Citation61]. Laboratory evaluation for patients suspected of having HAE include blood levels of complement C4, C1-INH protein, and C1-INH function; these tests should be repeated to confirm diagnosis of HAE 1 or 2 if any of these levels are abnormally low [Citation1]. outlines laboratory findings typical of the various types of angioedema. C1q levels are typically normal in most types of bradykinin-mediated angioedema; however, low levels can be indicative of AAE, especially in the patient presenting with symptoms after the age of 40 or those with lymphoproliferative or autoimmune disease [Citation61]. Laboratory analysis of complement levels C1q and C4 can be affected by handling of samples, giving spuriously low levels. C1-inhibitor function can be assayed by a commonly available enzyme-linked immunosorbent assay (ELISA) or by a chromogenic method. The ELISA may tend to overestimate function giving a “gray zone” result, a problem not present with the chromogenic assay. The chromogenic assay is unfortunately not as readily available as the ELISA [Citation65]. Laboratory values may also be altered by recent use of C1-INH or 17-alpha alkylated androgenic steroids. Genetic testing for SERPING1 mutations is not essential for the diagnosis of HAE-C1-INH types 1 and 2 [Citation66].

4.2. General approach to the management of HAE

Broadly, management strategies for patients with C1-INH-HAE include treatment of HAE attacks on an episode-by-episode basis (acute, on-demand treatment), pre-treatment prior to an event or procedure that may trigger an attack (short-term prophylaxis), or administration of ongoing therapy to prevent the development of HAE attacks (long-term or routine prophylaxis) [Citation1,Citation67,Citation68].

Clinical guidelines recommend that all patients with HAE should have access to on-demand treatment. Further, all patients should have sufficient medication for treatment of 2 attacks and should carry on-demand medication at all times [Citation1,Citation69–74]. Short-term prophylaxis is recommended before procedures that may induce an attack [Citation1]. Long-term, routine prophylaxis should be considered for patients who have frequent HAE attacks (≥1/month) or a history of laryngeal attacks, for patients whose condition has resulted in excessive loss of work or school, significant anxiety, impaired quality of life, and/or for patients for whom on-demand therapy is not controlling the disease adequately [Citation1,Citation69,Citation75,Citation76]. It is recommended that patients be evaluated for long-term prophylaxis at least once a year [Citation1]. An increasing trend toward home-based treatment has been associated with increased quality of life and may facilitate timely treatment, increase patients’ self-confidence in managing their HAE, and provide patients with greater flexibility and convenience, thus enabling them to lead more normal lives [Citation1,Citation77–81]. Home-based treatment may enhance adherence to routine HAE prophylaxis, possibly because of the added convenience and reduced requirement to travel to physician’s offices or infusion centers [Citation82].

A growing expert consensus recognizes that management of HAE should focus on individualizing patient care, with a key management goal being the improvement of quality of life [Citation1,Citation69,Citation74,Citation83]. Ideally, patients with this condition should be followed by a clinician with knowledge and experience in treating patients with HAE, one who is familiar with all available treatment options, given the complexity and variability of HAE and its management [Citation73,Citation74].

5. Treatment for HAE

5.1. Changes in HAE treatment paradigm

5.1.1. Historical overview of HAE treatments

In the past decade there has been a great expansion in the availability of new HAE-specific medications in the United States (US) and globally. Historically, available treatments for HAE included on-demand treatment with fresh frozen plasma and prophylaxis with attenuated androgens and antifibrinolytics [Citation18,Citation58,Citation67]. Although attenuated androgens effectively reduce attack frequency in most patients and are well tolerated by some patients, their use is limited by potentially serious side effects and poor efficacy in some patients, and efficacy is diminished when used at lower doses [Citation67,Citation84–86]. Antifibrinolytic agents are no longer recommended for long-term prophylaxis because of insufficient evidence of efficacy [Citation70]. These older treatment modalities have largely been displaced following the introduction of newer HAE-specific medications ().

Table 3. Available treatments for hereditary angioedema in the US

5.1.2. On-demand treatments

Intravenous C1-INH concentrate, (BERINERT®; CSL Behring, LLC; Kankakee, IL), was approved by the US FDA in 2009 for acute treatment of HAE attacks [Citation87], although this product had been available in Europe since the early 1970s [Citation96]. Additional HAE-pathway specific treatments for on-demand treatment were subsequently approved by the US FDA including a SC kallikrein inhibitor, ecallantide (KALBITOR®; Dyax Corp, now part of Takeda; Lexington, MA) [Citation90] in 2009; a SC bradykinin B2 receptor antagonist, icatibant (FIRAZYR®; Shire Orphan Therapies LLC, now part of Takeda; Lexington, MA) [Citation89] in 2011; and IV recombinant C1-INH (conestat alfa; RUCONEST®; Pharming Healthcare Inc.; Warren, NJ) in 2014 [Citation88]. The IV C1-INH concentrate (CINRYZE®; ViroPharma Biologics LLC, a Takeda Company; Lexington, MA) is approved for on-demand treatment in the European Union, but not the US.

5.1.3. Long-term prophylaxis

5.1.3.1. Prophylaxis with plasma-derived IV C1-INH and unmet needs

Plasma-derived IV C1-INH concentrate (CINRYZE®; ViroPharma Biologics LLC, a Takeda Company; Lexington, MA) was approved for routine prophylaxis in patients with HAE in October 2008 [Citation92]. HAE guidelines are placing greater emphasis on expanding eligibility for prophylaxis and striving to improve quality of life. Until recently, C1-INH replacement therapy for prophylaxis was available only as an IV administered formulation, C1-INH(IV), (CINRYZE®; ViroPharma Biologics LLC, a Takeda Company; Lexington, MA). A limitation of C1-INH(IV) prophylaxis is frequent breakthrough attacks in some patients, especially at the original approved dose of 1000 IU twice weekly [Citation44,Citation97–99]. Doses up to 2500 IU are now allowed [Citation92]. In addition to concerns regarding breakthrough attacks, routine prophylaxis with this IV formulation presents challenges related to IV access, safety risks of thrombosis and infection from indwelling port use, and issues related to patient convenience [Citation73,Citation98,Citation100].

An online survey of 50 adults with HAE, 47 of whom were using C1-INH(IV) for prophylaxis, found that 30% of patients who administered via a peripheral vein were somewhat dissatisfied or not at all satisfied with their treatment, with 62% of patients reporting difficulty in finding usable veins or administering the infusion [Citation98]. Further, complications such as blockage, thrombosis, and infection were experienced by almost half of patients who administered their C1-INH(IV) through a central port catheter. The concerns associated with C1-INH(IV) underscore that additional options for routine prophylaxis of HAE are needed.

5.1.3.2. C1-INH(SC)

In June 2017, the US FDA approved the first SC formulation of C1-INH (C1-INH[SC]; HAEGARDA®; CSL Behring, LLC; Marburg, Germany). C1-INH(SC) is indicated for the routine prevention of HAE attacks in adolescents and adults [Citation93,Citation101]. This relatively new C1-INH treatment option may provide greater convenience, facilitate self-administration, and potentially enhance adherence. The prospective, multicenter, open-label, crossover Clinical Study for Optimal Management of Preventing Angioedema with Low-Volume Subcutaneous C1-INH Replacement Therapy (COMPACT phase 2 study) showed that administration of C1-INH(SC) achieved constant trough C1-INH functional activity levels above 40% at 3000 and 6000 IU, a threshold expected to provide clinical protection against HAE attacks [Citation102]. A post hoc analysis found that, compared with standard C1-INH(IV) prophylaxis, simulated C1-INH functional activity time profiles were associated with a lower peak-to-trough ratio and more consistent exposures. Patients with a low body weight who are administered C1-INH at a fixed dose are predicted to achieve a relatively high C1-INH level compared with higher body weight patients. Therefore, body weight-adjusted dosing is predicted to achieve the same level of activity across the range of body weights.

Clinical efficacy of C1-INH(SC) was evaluated in a prospective, international, multicenter, randomized, double-blind, placebo-controlled, dose-ranging phase 3 trial (COMPACT) [Citation103]. Patients ≥12 years of age with C1-INH-HAE type 1 or 2 who experienced at least 2 attacks per month were randomly assigned to 1 of 4 treatment sequences: C1-INH(SC) 40 IU/kg (n = 45) or 60 IU/kg (n = 45) followed or preceded by a placebo phase in crossover fashion; each treatment was self-administered subcutaneously twice-weekly over a 16-week period. The mean (95% CI) rate of time-normalized attacks per month was significantly lower for patients randomized to C1-INH(SC) versus placebo; for the FDA approved dose of 60 IU/kg, there were 0.52 (0.00 to 1.04) attacks per month versus 4.03 (3.51 to 4.55) for placebo (within-patient difference, −3.51 [−4.21 to −2.81]; P < 0.001). The majority of patients receiving C1-INH(SC) 60 IU/kg had a response, with 90% (95% CI, 77% to 96%), 83% (68–91%), and 58% (42–72%) of patients having a ≥ 50%, ≥ 70%, and ≥ 90% reduction in HAE attacks, respectively. Notably, fewer patients had laryngeal attacks during treatment with C1-INH(SC) 60 IU/kg (0 patients) than while receiving placebo (25 patients). In a long-term, open-label extension of the COMPACT study, C1-INH(SC) demonstrated substantial and sustained efficacy over a median treatment duration of 53 weeks. Nineteen of 23 patients (83%) treated with C1-INH(SC) 60 IU/kg for more than 2 years were attack-free during the final six weeks of the study [Citation104].

Results from a post hoc analysis of data from the COMPACT study suggest that, compared with placebo, C1-INH(SC) may have important benefits with regard to anxiety and work productivity [Citation54]. Another post hoc analysis of COMPACT data indicated that patients using C1-INH(IV) as routine prophylaxis prior to the COMPACT study experienced an approximately 50% further mean reduction (>70% median reduction) in HAE attack rate while using C1-INH(SC) (40 or 60 IU/kg) for prophylaxis during the study [Citation105].

In the double-blind COMPACT study, adverse events were reported by similar percentages of patients while using C1-INH(SC) or placebo injections. The most common events in the placebo-controlled and open-label studies were injection site reactions, the great majority of which were mild and transient. There were no anaphylactic reactions or evidence of inhibitory anti-C1-INH antibodies during these studies [Citation103,Citation104].

5.1.3.3. Lanadelumab-flyo

A recombinant, human monoclonal antibody long-acting inhibitor of kallikrein formulated for SC administration, lanadelumab-flyo (TAKHZYROTM; DX-2930; Shire, now part of Takeda; Lexington, MA) [Citation106] was approved by the FDA in August 2018 for prophylaxis to prevent HAE attacks [Citation94]. The recommended dose of lanadelumab is 300 mg every 2 weeks; dosing every 4 weeks can be considered in patients who are well-controlled (eg, attack free) for more than six months [Citation94]. In a 26-week, phase 3, multicenter, randomized, double-blind, placebo-controlled trial in patients 12 years of age or older with HAE (HELP study; NCT02586805), all lanadelumab treatment regimens were more effective than placebo in reducing the number of HAE attacks during the 26-week treatment period. When given at the FDA approved dose (300 mg SC given every 2 weeks), lanadelumab reduced the monthly HAE attack rate by 87% versus placebo (mean rate ratio 0.13 [95% CI, 0.07–0.24]; P < 0.001) [Citation107]. Post hoc analyses of the HELP study found that attack prevention was evident within the initial 2 weeks of treatment, and sustained, even among patients who had a high baseline attack frequency [Citation108]. A phase 3 long-term/extension study was conducted (NCT02741596; N = 212; median [range] 33 [1.4, 34.2] months of therapy), and reported that efficacy was sustained without emergence of any new safety concerns [Citation109,Citation110].

Injection site reactions (pain, erythema, bruising at injection site) were the most common adverse reactions in the pivotal clinical trial, reported in 52% of lanadelumab-treated patients and 34% of placebo-treated patients. Headache was reported by 33%, 21%, and 11% of patients receiving lanadelumab 300 mg every 2 weeks, 300 mg every 4 weeks, and 150 mg every 4 weeks, respectively, and reported by 22% of patients receiving placebo [Citation94]. Low-titer neutralizing anti-drug antibodies were identified in two lanadelumab patients in the phase 3 double-blind trial and six (2.8%) patients in the extension study; the presence of the antibodies had no noticeable clinical impact [Citation107,Citation110].

5.1.3.4. Berotralstat

Berotralstat (ORLADEYOTM; BioCryst Pharmaceuticals, Inc.; Durham, NC), approved by the FDA in December 2020, is a small molecule plasma kallikrein inhibitor and the first oral drug developed specifically for HAE prophylaxis. The recommended dose is one 150 mg capsule daily [Citation95]. The safety and efficacy of berotralstat were established in the phase 3 Angioedema Prophylaxis-2 (APeX-2; NCT03485911) trial, a double-blind, parallel-group study in patients with HAE-C1-INH aged 12 years or older [Citation111]. At the FDA-approved dose of 150 mg once daily, the rate of HAE attacks was reduced significantly relative to placebo (1.31 vs 2.35 attacks per month; P < 0.001) during a 24-week treatment period. Gastrointestinal adverse events including abdominal pain, vomiting, and diarrhea occurred primarily during the first month of treatment were more common among patients taking berotralstat 150 mg as compared to those taking placebo (50% vs 36%); the median duration of these events in the berotralstat group was 2 days, as compared to 1 day in the placebo group. Back pain was also reported more frequently in the berotralstat group.

5.1.3.5. Recombinant C1-INH (conestat alfa)

RUCONEST® (conestat alfa; recombinant C1-INH; Pharming Healthcare Inc.) is currently FDA-approved only as on-demand treatment in HAE [Citation88]. However, some published clinical study data suggest efficacy with this product when used as short-term or long-term prophylaxis [Citation97,Citation112].

5.2. HAE management in special populations

5.2.1. Pregnant and lactating patients

Increased estrogen exposure during pregnancy (as well as menstruation and use of combination oral contraceptives), can increase the severity of HAE symptoms in some, but not all, women adding to the challenge of disease management [Citation113,Citation114]. HAE management guidelines recommend C1-INH products during pregnancy for prophylaxis and as on-demand treatment when necessary [Citation1,Citation61]. Available data gathered from women exposed to C1-INH during pregnancy suggest that C1-INH use appears to be generally safe for both mother and baby [Citation113,Citation115,Citation116]. Androgens cross the placental barrier and can lead to virilization of a female fetus, thus are absolutely contraindicated during pregnancy [Citation1,Citation61,Citation117]. Surprisingly, the labor and delivery process does not generally trigger HAE attacks [Citation1].

Breastfeeding can trigger HAE attacks but is still encouraged because of the benefits to the baby [Citation1]. C1-INH is the preferred therapy choice in breastfeeding women. It is unknown whether androgens are excreted in breast milk, but their use during breastfeeding is not advised [Citation117].

5.2.2. Pediatric patients

Due to the rarity of HAE and the tendency of clinical trials to focus largely on adult patients, there is a relative lack of data overall in this age group. Yet, several HAE medications have been granted FDA approval for use in pediatric patients of any age (BERINERT®/C1-INH[IV]) or in age groups defined as either ‘adolescents’ or ‘12 years and older’ (RUCONEST®/recombinant C1-INH; CINRYZE®/C1-INH[IV], HAEGARDA®/C1-INH[SC]; TAKHZYROTM/lanadelumab-flyo; ORLADEYOTM/berotralstat) (). Guidelines currently recommend C1-INH as first-line therapy in pediatric patients as both on-demand and prophylactic therapy [Citation1,Citation61]. Guidelines stress that androgens should not be used for prophylaxis in patients younger than 16 [Citation61]. The smaller airway diameter in children may contribute to a particularly rapid progression to asphyxiation in laryngeal attacks, so rapid and effective intervention is critical [Citation1,Citation118].

5.3. Emerging and future therapies for HAE prophylaxis

In light of recent and ongoing advancements in medical therapies for HAE, more individualized management goals are being developed. Early recognition of HAE and early introduction of appropriate therapy, including prophylaxis, are management goals that aim to improve patients’ quality of life to the greatest extent possible.

Novel treatments and treatment approaches for HAE continue to be actively investigated. Molecules under clinical development at the time of manuscript writing are summarized in and include a SC factor XIIa-inhibitory monoclonal antibody, a SC compound that reduces production of prekallikrein, an oral bradykinin B2 receptor antagonist, and an oral plasma kallikrein inhibitor.

Table 4. Products in clinical development for HAE prophylaxis

6. Summary and conclusions

HAE is a lifelong, disabling, and potentially life-threatening condition. The root cause of HAE is related to an absence of functional C1-INH. HAE should be included in the differential diagnosis of patients presenting with angioedema given that misdiagnosis could lead to unnecessary interventions or even potentially life-threatening outcomes. Once HAE is identified, therapy (both acute treatment and prophylaxis) should be individualized considering unique patient factors such as attack history, proximity to a medical/infusion center, occupation/student status, impact of HAE on patients’ quality of life, and other patient circumstances and preferences/expectations [Citation84].

Guidelines recommend that the need for long-term prophylaxis be evaluated at least annually in each patient with HAE [Citation1,Citation84]. Ideally, patients with HAE should follow-up with a clinician who has expertise/experience with the management of HAE [Citation73,Citation74].

Acknowledgments

Medical writing and editorial assistance were provided by Katie Singh, of Churchill Communications, Maplewood, New Jersey.

Disclosure statement

JJ has received funds for sponsored research from CSL Behring, Takeda, and BioCryst Pharmaceuticals, Inc. and consulting fees and speaker honoraria from CSL Behring and Takeda.

TN has no conflicting interests to disclose.

A reviewer on this manuscript has disclosed that they have received fees for consultation from Takeda. The other peer reviewers on this manuscript have no other relevant financial relationships or otherwise to disclose.

Additional information

Funding

Medical writing support funding was provided by CSL Behring.

References

  • Maurer M, Magerl M, Ansotegui I, et al. The international WAO/EAACI guideline for the management of hereditary angioedema—The 2017 revision and update. Allergy. 2018;73(8):1575–1596.
  • Cicardi M, Bergamaschini B, Marasini B, et al. Hereditary angioedema: an appraisal of 104 cases. Am J Med Sci. 1982;284(1):2–9.
  • Bork K, Meng G, Staubach P, et al. Hereditary angioedema: new findings concerning symptoms, affected organs, and course. Am J Med. 2006;119(3):267–274.
  • Lumry W, Templeton T, Omert L, et al. Advances in hereditary angioedema: the prevention of angioedema attacks with subcutaneous C1-inhibitor replacement therapy. J Infus Nurs. 2020;43(3):134–145.
  • Bernstein JA, Cremonesi P, Hoffmann TK, et al. Angioedema in the emergency department: a practical guide to differential diagnosis and management. Int J Emerg Med. 2017;10(1):15.
  • Long BJ, Koyfman A, Gottlieb M. Evaluation and management of angioedema in the emergency department. West J Emerg Med. 2019;20(4):587–600.
  • Bova M, De Feo G, Parente R, et al. Hereditary and acquired angioedema: heterogeneity of pathogenesis and clinical phenotypes. Int Arch Allergy Immunol. 2018;175(3):126–135.
  • Nzeako UC. Diagnosis and management of angioedema with abdominal involvement: a gastroenterology perspective. World J Gastroenterol. 2010;16(39): 4913–4921.
  • Moellman JJ, Bernstein JA, Lindsell C, et al. A consensus parameter for the evaluation and management of angioedema in the emergency department. Acad Emerg Med. 2014;21(4):469–484.
  • Lenschow M, Bas M, Johnson F, et al. A score for the differential diagnosis of bradykinin- and histamine-induced head and neck swellings. Eur Arch Otorhinolaryngol. 2018;275(7):1767–1773.
  • Aygören-Pürsün E, Magerl M, Maetzel A, et al. Epidemiology of Bradykinin-mediated angioedema: a systematic investigation of epidemiological studies. Orphanet J Rare Dis. 2018;13(1):73.
  • Henao MP, Kraschnewski JL, Kelbel T, et al. Diagnosis and screening of patients with hereditary angioedema in primary care. Ther Clin Risk Manag. 2016;12:701–711.
  • Nzeako UC, Frigas E, Tremaine WJ. Hereditary angioedema: a broad review for clinicians. Arch Intern Med. 2001;161(20):2417–2429.
  • Zanichelli A, Longhurst HJ, Maurer M, et al. Misdiagnosis trends in patients with hereditary angioedema from the real-world clinical setting. Ann Allergy Asthma Immunol. 2016;117(4):394–398.
  • Bernstein JA. HAE update: epidemiology and burden of disease. Allergy Asthma Proc. 2013;34(1):3–6.
  • Cicardi M, Agostoni A. Hereditary angioedema. N Engl J Med. 1996;334(25):1666–1667.
  • Cicardi M, Aberer W, Banerji A, et al. Classification, diagnosis, and approach to treatment for angioedema: consensus report from the Hereditary Angioedema International Working Group. Allergy. 2014;69(5):602–616.
  • Bork K. Pasteurized and nanofiltered, plasma-derived C1 esterase inhibitor concentrate for the treatment of hereditary angioedema. Immunotherapy. 2014;6(5):533–551.
  • 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(3 Suppl):S51–S131.
  • Bork K, Gul D, Hardt J, et al. Hereditary angioedema with normal C1 inhibitor: clinical symptoms and course. Am J Med. 2007;120(11):987–992.
  • Bork K, Wulff K, Meinke P, et al. A novel mutation in the coagulation factor 12 gene in subjects with hereditary angioedema and normal C1-inhibitor. Clin Immunol. 2011;141(1):31–35.
  • Bova M, Suffritti C, Bafunno V, et al. Impaired control of the contact system in hereditary angioedema with normal C1-inhibitor. Allergy. 2020;75(6):1394–1403.
  • 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(3):1009–1017.
  • 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.
  • Bork K, Wulff K, Steinmüller-Magin L, et al. Hereditary angioedema with a mutation in the plasminogen gene. Allergy. 2018;73(2):442–450.
  • 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.
  • 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;S0091-6749(21)00094-4.
  • Kaplan AP, Joseph K. Complement, kinins, and hereditary angioedema: mechanisms of plasma instability when C1 inhibitor is absent. Clin Rev Allergy Immunol. 2016;51(2):207–215.
  • Zuraw BL, Christiansen SC. HAE pathophysiology and underlying mechanisms. Clin Rev Allergy Immunol. 2016;51(2):216–229.
  • Longhurst HJ, Bork K. Hereditary angioedema: an update on causes, manifestations and treatment. Br J Hosp Med (Lond). 2019;80(7):391–398.
  • Germenis AE, Speletas M. Genetics of hereditary angioedema revisited. Clin Rev Allergy Immunol. 2016;51(2):170–182.
  • Kelemen Z, Moldovan D, Mihaly E, et al. Baseline level of functional C1-inhibitor correlates with disease severity scores in hereditary angioedema. Clin Immunol. 2010;134(3):354–358.
  • Bafunno V, Bova M, Loffredo S, et al. Mutational spectrum of the c1 inhibitor gene in a cohort of Italian patients with hereditary angioedema: description of nine novel mutations. Ann Hum Genet. 2014;78(2):73–82.
  • Bork K, Staubach P, Eckardt AJ, et al. Symptoms, course, and complications of abdominal attacks in hereditary angioedema due to C1 inhibitor deficiency. Am J Gastroenterol. 2006;101(3):619–627.
  • Testori A, Melamed I. Neurological manifestations of hereditary angioedema. Ann Allergy Asthma Immunol. 2017;118(1):119–120.
  • Bonnaud I, Rouaud V, Guyot M, et al. Exceptional stroke-like episodes in a patient with type 1 autosomal angioedema. Neurology. 2012;78(8):598.
  • Bork K, Hardt J, Witzke G. Fatal laryngeal attacks and mortality in hereditary angioedema due to C1-INH deficiency. J Allergy Clin Immunol. 2012;130(3):692–697.
  • Bork K, Brehler R, Witzke G, et al. Blindness, tetraspasticity, and other signs of irreversible brain damage in hereditary angioedema. Ann Allergy Asthma Immunol. 2017;118(4):520–521.
  • Banerji A. The burden of illness in patients with hereditary angioedema. Ann Allergy Asthma Immunol. 2013;111(5):329–336.
  • Lumry WR, Castaldo AJ, Vernon MK, et al. The humanistic burden of hereditary angioedema: impact on health-related quality of life, productivity, and depression. Allergy Asthma Proc. 2010;31(5):407–414.
  • Busse PJ, Christiansen SC. Hereditary angioedema. N Engl J Med. 2020;382(12):1136–1148.
  • Zuraw BL, Christiansen SC. How we manage persons with hereditary angioedema. Br J Haematol. 2016;173(6):831–843.
  • Caballero T, Maurer M, Longhurst HJ, et al. Triggers and prodromal symptoms of angioedema attacks in patients with hereditary angioedema. J Investig Allergol Clin Immunol. 2016;26(6):283–386.
  • Banerji A, Busse P, Christiansen SC, et al. Current state of hereditary angioedema management: a patient survey. Allergy Asthma Proc. 2015;36(3):213–217.
  • Aygören-Pürsün E, Bygum A, Beusterien K, et al. Estimation of EuroQol 5-dimensions health status utility values in hereditary angioedema. Patient Prefer Adherence. 2016;10:1699–1707.
  • Jindal NL, Harniman E, Prior N, et al. Hereditary angioedema: health-related quality of life in Canadian patients as measured by the SF-36. Allergy Asthma Clin Immunol. 2017;13:4.
  • Kuman Tunçel Ö, Gökmen NM, Demir E, et al. The impact of hereditary angioedema on quality of life and family planning decisions. Int J Psychiatry Med. 2019;54(6):377–394.
  • 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(1):47–53.
  • Fouche AS, Saunders EF, Craig T. Depression and anxiety in patients with hereditary angioedema. Ann Allergy Asthma Immunol. 2014;112(4):371–375.
  • Kessel A, Farkas H, Kivity S, et al. The relationship between anxiety and quality of life in children with hereditary angioedema. Pediatr Allergy Immunol. 2017;28(7):692–698.
  • Nordenfelt P, Dawson S, Wahlgren CF, et al. Quantifying the burden of disease and perceived health state in patients with hereditary angioedema in Sweden. Allergy Asthma Proc. 2014;35(2):185–190.
  • Wilson DA, Bork K, Shea EP, et al. Economic costs associated with acute attacks and long-term management of hereditary angioedema. Ann Allergy Asthma Immunol. 2010;104(4):314–320.
  • Aygören-Pürsün E, Bygum A, Beusterien K, et al. Socioeconomic burden of hereditary angioedema: results from the hereditary angioedema burden of illness study in Europe. Orphanet J Rare Dis. 2014;9:99.
  • 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(5):1733–1741.e3.
  • Zanichelli A, Magerl M, Longhurst H, et al. Hereditary angioedema with C1 inhibitor deficiency: delay in diagnosis in Europe. Allergy Asthma Clin Immunol. 2013;9(1):29.
  • Bygum A. Hereditary angio-oedema in Denmark: a nationwide survey. Br J Dermatol. 2009;161(5):1153–1158.
  • Frank MM, Zuraw B, Banerji A, et al. Management of children with hereditary angioedema due to C1 inhibitor deficiency. Pediatrics. 2016;138(5).
  • Farkas H. Pediatric hereditary angioedema due to C1-inhibitor deficiency. Allergy Asthma Clin Immunol. 2010;6(1):18.
  • Berger J, Carroll MP, Champoux E, et al. Extremely delayed diagnosis of type II hereditary angioedema: case report and review of the literature. Mil Med. 2018;183(11–12):e765–e767.
  • 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.
  • Busse PJ, Christiansen SC, Riedl MA, et al. US HAEA medical advisory board 2020 guidelines for the management of hereditary angioedema. J Allergy Clin Immunol Pract. 2021;9:132–150.
  • Farkas H, Martinez-Saguer I, Bork K, et al. International consensus on the diagnosis and management of pediatric patients with hereditary angioedema with C1 inhibitor deficiency. Allergy. 2017;72(2):300—313.
  • Abdel-Samad NN, Kokai JS. A case of acquired angioedema with low C1 inhibitor (C1-INH) associated with splenic marginal zone lymphoma. Am J Case Rep. 2019;20:1476–1481.
  • Bork K, Machnig T, Wulff K, et al. 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(1):289.
  • Li HH, Busse P, Lumry WR, et al. Comparison of chromogenic and ELISA functional C1 inhibitor tests in diagnosing hereditary angioedema. J Allergy Clin Immunol Pract. 2015;3(2):200–205.
  • Cicardi M, Zuraw BL. Angioedema due to bradykinin dysregulation. J Allergy Clin Immunol Pract. 2018;6(4):1132–1141.
  • Gower RG. HAE update: determining optimal patient specific therapy. Allergy Asthma Proc. 2013;34(1):7–12.
  • Bork K. A decade of change: recent developments in pharmacotherapy of hereditary angioedema (HAE). Clin Rev Allergy Immunol. 2016;51(2):183–192.
  • Cicardi M, Bork K, Caballero T, et al. Evidence-based recommendations for the therapeutic management of angioedema owing to hereditary C1 inhibitor deficiency: consensus report of an International Working Group. Allergy. 2012;66(2):147–157.
  • Craig TJ, Aygören-Pürsün E, Bork K, et al. WAO guideline for the management of hereditary angioedema. World Allergy Organ J. 2012;5(12):182–199.
  • Lang DM, Aberer W, Bernstein JA, et al. International consensus on hereditary and acquired angioedema. Ann Allergy Asthma Immunol. 2012;109:395–402.
  • Wahn V, Aberer W, Eberl W, et al. Hereditary angioedema (HAE) in children and adolescents—a consensus on therapeutic strategies. Eur J Pediatr. 2012;171:1339–1348.
  • Zuraw BL, Banerji A, Bernstein JA, et al. US hereditary angioedema association medical advisory board 2013 recommendations for the management of hereditary angioedema due to C1 inhibitor deficiency. J Allergy Clin Immunol Pract. 2013;1(5):458–467.
  • Betschel S, Badiou J, Binkley K, et al. Canadian hereditary angioedema guideline. Allergy Asthma Clin Immunol. 2014;10:50.
  • Craig T, Riedl M, Dykewicz MS, et al. When is prophylaxis for hereditary angioedema necessary? Ann Allergy Asthma Immunol. 2009;102(5):366–372.
  • Longhurst H, Zinser E. Prophylactic therapy for hereditary angioedema. Immunol Allergy Clin North Am. 2017;37(3):557–570.
  • Bygum A, Andersen KE, Mikkelsen CS. Self-administration of intravenous C1-inhibitor therapy for hereditary angioedema and associated quality of life benefits. Eur J Dermatol. 2009;19(2):147–151.
  • Longhurst HJ, Farkas H, Craig T, et al. HAE international home therapy consensus document. Allergy Asthma Clin Immunol. 2010;6(1):22.
  • Boysen HB, Bouillet L, Aygören-Pürsün E. Challenges of C1-inhibitor concentrate self-administration. Int Arch Allergy Immunol. 2013;161 Suppl 1:21–25.
  • Cicardi M, Craig TJ, Martinez-Saguer I, et al. Review of recent guidelines and consensus statements on hereditary angioedema therapy with focus on self-administration. Int Arch Allergy Immunol. 2013;161 Suppl 1:3–9.
  • Li HH. Self-administered C1 esterase inhibitor concentrates for the management of hereditary angioedema: usability and patient acceptance. Patient Prefer Adherence. 2016;10:1727–1737.
  • Gregory C, Landmesser LM, Corrigan L, et al. Feasibility of home infusion and self-administration of nanofiltered C1 esterase inhibitor for routine prophylaxis in patients with hereditary angioedema and characterization of a training and support program. J Infus Nurs. 2014;37(1):29–34.
  • Farkas H, Martinez-Saguer I, Bork K, et al. International consensus on the diagnosis and management of pediatric patients with hereditary angioedema with C1 inhibitor deficiency. Allergy. 2017;72(2):300–313.
  • Gower RG, Lumry WR, Davis-Lorton MA, et al. Current options for prophylactic treatment of hereditary angioedema in the United States: patient-based considerations. Allergy Asthma Proc. 2012;33(3):235–240.
  • Zuraw BL, Davis DK, Castaldo AJ. Tolerability and effectiveness of 17-α-alkylated androgen therapy for hereditary angioedema: a re-examination. J Allergy Clin Immunol Pract. 2016;4(5):948–955.e15.
  • Tse KY, Zuraw BL, Chen Q, et al. Anabolic androgen use in the management of hereditary angioedema: not so cheap after all. Ann Allergy Asthma Immunol. 2017;118(4):456–460.e1.
  • BERINERT® [package insert]. Kankakee, IL: CSL Behring, LLC; 2019.
  • RUCONEST® [package insert]. Warren, NJ: Pharming Healthcare Inc.; 2020.
  • FIRAZYR® [package insert]. Lexington, MA: Shire Orphan Therapies LLC, now part of Takeda; 2015.
  • KALBITOR® [package insert]. Burlington, MA: Dyax Corp, now part of Takeda; 2015.
  • DANOCRINE® prescribing information. Bridgewater, NJ: Sanofi-Aventis US LLC. December 2011.
  • CINRYZE® [package insert]. Lexington, MA: ViroPharma Biologics LLC, a Takeda Company; 2021.
  • HAEGARDA® [package insert]. Kankakee, IL: CSL Behring, LLC; 2017.
  • TAKHZYROTM [package insert]. Lexington, MA: Dyax Corp, now part of Takeda; 2018.
  • ORLADEYOTM [package insert]. Durham, NC: BioCryst Pharmaceuticals, Inc.; 2020.
  • Brackertz D, Kueppers F. Hereditary angioneurotic oedema. Lancet. 1973;2(7830):680.
  • Riedl MA, Grivcheva-Panovska V, Moldovan D, et al. Recombinant human C1 esterase inhibitor for prophylaxis of hereditary angio-oedema: a phase 2, multicentre, randomised, double-blind, placebo-controlled crossover trial. Lancet. 2017;390(10102):1595–1602.
  • Riedl MA, Banerji A, Busse PJ, et al. Patient satisfaction and experience with intravenously administered C1-inhibitor concentrates in the United States. Ann Allergy Asthma Immunol. 2017;119(1):59–64.
  • Zuraw BL, Kalfus I. Safety and efficacy of prophylactic nanofltered C1-inhibitor in hereditary angioedema. Am J Med. 2012;125(9):938.e1–7.
  • Yoo SM, Khan DA. Implantable venous access device associated complications in patients with hereditary angioedema. J Allergy Clin Immunol Pract. 2013;1(5):524–525.
  • US Food and Drug Administration. FDA approves first subcutaneous C1 Esterase Inhibitor to treat rare genetic disease. 2017 Jun [cited 2020 Feb 7]. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-first-subcutaneous-c1-esterase-inhibitor-treat-rare-genetic-disease.
  • Zuraw BL, Cicardi M, Longhurst HJ, et al. Phase II study results of a replacement therapy for hereditary angioedema with subcutaneous C1-inhibitor concentrate. Allergy. 2015;70(10):1319–1328.
  • Longhurst H, Cicardi M, Craig T, et al. COMPACT Investigators. Prevention of hereditary angioedema attacks with a subcutaneous C1 inhibitor. N Engl J Med. 2017;376(12):1131–1140.
  • Craig T, Zuraw B, Longhurst H, et al. for the COMPACT Investigators. Long-term outcomes with subcutaneous C1-inhibitor replacement therapy for prevention of hereditary angioedema attacks. J Allergy Clin Immunol Pract. 2019;7(6):1793–1802.
  • Craig T, Lumry W, Cicardi M, et al. Treatment effect of switching from intravenous to subcutaneous C1-inhibitor for prevention of hereditary angioedema attacks: COMPACT subgroup findings. J Allergy Clin Immunol Pract. 2019;7(6):2035–2038.
  • Banerji A, Busse P, Shennak M, et al. Inhibiting plasma kallikrein for hereditary angioedema prophylaxis. N Engl J Med. 2017;376(8):717–728.
  • Banerji A, Riedl MA, Bernstein JA, et al. Effect of lanadelumab compared with placebo on prevention of hereditary angioedema attacks: a randomized clinical trial. JAMA. 2018;320(20):2108–2121. 10.1001/jama.2018.16773. Erratum in: JAMA. 2019 Apr 23;321(16):1636.
  • Riedl MA, Maurer M, Bernstein JA, et al. HELP Investigators. Lanadelumab demonstrates rapid and sustained prevention of hereditary angioedema attacks. Allergy. 2020;75(11):2879–2887.
  • Riedl MA, Bernstein JA, Craig T, et al. An open-label study to evaluate the long-term safety and efficacy of lanadelumab for prevention of attacks in hereditary angioedema: design of the HELP study extension. Clin Transl Allergy. 2017;7:36.
  • Banerji A, Hao J, Yu M, et al. Long-term efficacy and safety of lanadelumab: final results from the HELP open-label extension study [abstract P150]. Allergy Asthma Immunol. 2020;125:S21.
  • 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. 2020;S0091-6749(20):31484–31486.
  • Valerieva A, Staevska M, Jesenak M, et al. Recominant huima C1 esterase as short-term prophylaxis in patients with hereditary angioedema. J Allergy Clin Immunol Pract. 2020;8(2):799–802.
  • Martinez-Saguer I, Rusicke E, Aygören-Pürsün E, et al. Characterization of acute hereditary angioedema attacks during pregnancy and breast-feeding and their treatment with C1 inhibitor concentrate. Am J Obstet Gynecol. 2010;203(2):131.e1–7.
  • Bouillet L, Longhurst H, Boccon-Gibod I, et al. Disease expression in women with hereditary angioedema. Am J Obstet Gynecol. 2008;199(5):484.e1–484.e4.
  • Fox J, Vegh AB, Martinez-Saguer I, et al. Safety of a C1-inhibitor concentrate in pregnancy women with hereditary angioedema. Allergy Asthma Proc. 2017;38(3):216–221.
  • Levy DS, Farkas H, Riedl MA, et al. Long-term efficacy and safety of subcutaneous C1-inhibitor in women with hereditary angioedema: subgroup analysis from an open-label extension of a phase 3 trial. Allergy Asthma Clin Immunol. 2020;16:8.
  • Caballero T, Canabal J, Rivero-Paparoni D, et al. Management of hereditary angioedema in pregnant women: a review. Int J Women’s Health. 2014;6:839–848.
  • Bork K, Hardt J, Schicketanz KH, et al. Clinical studies of sudden upper airway obstruction in patients with hereditary angioedema due to C1 esterase inhibitor deficiency. Arch Intern Med. 2003;163(10):1229–1235.
  • CSL Behring. CSL312 (Garadacimab) in the prevention of hereditary angioedema attacks. [ cited 2021 Mar 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT04656418?term=Garadacimab&draw=2&rank=1.
  • Ionis Pharmaceuticals Inc. A study to assess the clinical efficacy of IONIS-PKK-LRx in participants with hereditary angioedema. [ cited 2021 Mar 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT04030598?term=ionis-pkk&draw=2&rank=3.
  • Ionis Pharmaceuticals Inc. An extension study of IONIS-PKK-LRx in participants with hereditary angioedema. [ cited 2021 Mar 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT04307381?term=Ionis-PKK&draw=2&rank=2.
  • Pharvaris Netherlands B.V. Dose-ranging study of oral PHA-022121 for acute treatment of angioedema attacks in patients with hereditary angioedema (RAPIDe-1). [ cited 2021 Mar 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT04618211?term=PHA121&draw=2&rank=1.
  • KalVista Pharmaceuticals. A phase II, cross-over clinical trial evaluating the efficacy and safety of KVD900 in the on-demand treatment of angioedema attacks in adult subjects with hereditary angioedema Type I or II. [ cited 2021 Mar 3]. Available from: https://clinicaltrials.gov/ct2/show/NCT04208412?term=KVD900&draw=2&rank=1.
  • KalVista Pharmaceuticals. KVD824 for HAE. [ cited 2021 Mar 9]. Available from: https://www.kalvista.com/products-pipeline/kvd824-hae.