ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Hereditary angioedema (due to C1 inhibitor deficiency): General care and long-term prophylaxis

Hereditary angioedema (due to C1 inhibitor deficiency): General care and long-term prophylaxis
Literature review current through: Jan 2024.
This topic last updated: Jun 29, 2023.

INTRODUCTION — Hereditary angioedema (HAE) with C1 inhibitor deficiency is a rare genetic disorder in which mutations in the SERPING1 gene result in deficiency (type I) or dysfunction (type II) of C1 inhibitor (C1-INH) protein [1,2]. Terminology is evolving, and the abbreviations HAE-C1-INH and C1-INH-HAE are both used for this disorder in the literature [2,3]. HAE is characterized by recurrent episodes of angioedema without pruritus or urticaria, which most often affect the skin or mucosal tissues of the upper respiratory and gastrointestinal tracts. Although swelling resolves spontaneously in two to five days in the absence of treatment, angioedema is often temporarily debilitating, and laryngeal angioedema may cause fatal asphyxiation.

In patients with sufficiently frequent or severe episodes of angioedema, regularly administered prophylaxis is required to maintain an acceptable quality of life. This is referred to as long-term prophylaxis, in contrast to short-term prophylaxis, which is premedication given briefly before a specific medical or dental procedure. Short-term prophylaxis is discussed separately. (See "Hereditary angioedema: Short-term prophylaxis before procedures or stressful events to prevent angioedema episodes".)

The general management of adults and children with HAE-C1-INH and the various long-term prophylactic therapies that are used to prevent attacks will be reviewed here. The management of HAE attacks as well as the clinical manifestations, pathogenesis, and diagnosis of this condition are discussed separately:

(See "Hereditary angioedema: Acute treatment of angioedema attacks".)

(See "Hereditary angioedema: Epidemiology, clinical manifestations, exacerbating factors, and prognosis".)

(See "Hereditary angioedema (due to C1 inhibitor deficiency): Pathogenesis and diagnosis".)

There is another form of HAE in which patients have normal complement studies called HAE with normal C1-INH. This disorder affects mainly females, while males are often asymptomatic carriers. Mutations in at least four genes (coding for coagulation factor XII, plasminogen, angiopoietin 1, and kininogen 1) have been associated with some patients with HAE with normal C1-INH. This disorder is also characterized by the tendency of estrogens in exacerbating the swelling. HAE with normal C1-INH is reviewed separately. (See "Hereditary angioedema with normal C1 inhibitor".)

GENERAL CARE — General care measures for HAE include education, testing of family members, identification and avoidance of possible triggers, and planning for HAE attacks. The first international consensus document was published in 2010 based upon the available literature and practices worldwide [4]. Two updated international consensus documents/guidelines have since been released [5,6]. Recommendations have also been published by an expert American group [7]. The approach in this review is consistent with these publications.

Education — The most important intervention for a patient who has been newly diagnosed with HAE is education about the disease. Both the affected individual and his/her family members should be involved. Patients can access detailed, patient-oriented information through the United States Hereditary Angioedema Association and HAE International. The Angioedema Centers of Reference and Excellence (ACARE) program has been launched for developing and accrediting an interactive network of HAE centers. Patients should be taught about attack triggers, potential risks, and the benefits of early treatment of HAE attacks. The genetics of HAE and the implications regarding family members should be discussed. Finally, the entire range of treatment options should be explained.

Testing of family members — First-degree relatives of the patient should be offered diagnostic testing for HAE. The disease is transmitted in an autosomal dominant fashion with variable but typically high penetrance. However, the patient may be the first affected member in the family since approximately 25 percent of cases result from new mutations.

The importance of testing first-degree family members is illustrated by a cohort study that demonstrated that the risk of death from asphyxiation was greater in patients with undiagnosed HAE compared with those who had been formally diagnosed and educated about the condition [8]. It should be explained to patients that one-half of the children of a person with HAE can be expected to have the abnormal gene, although not everyone with the abnormal gene has symptoms. Some people with the abnormal gene have minimal or no symptoms, while others have symptoms of variable severity, and there is no way to predict how severely an individual will be affected. Another advantage of testing is that it may help avoid diagnostic confusion and inappropriate treatment if and when the person does develop symptoms.

Complement studies (C4, C1-INH level, and C1-INH function) are the recommended tests for screening family members. Additional issues surrounding testing are discussed separately. (See "Hereditary angioedema (due to C1 inhibitor deficiency): Pathogenesis and diagnosis", section on 'Testing family members'.)

Trigger avoidance — Triggers vary among patients, although the following measures are important for most:

Avoidance of trauma, particularly iatrogenic trauma to the face and upper respiratory tract

Recognition and prompt treatment of upper airway and dental infections

Avoidance of medications that can worsen the severity or frequency of angioedema episodes (estrogens, angiotensin-converting enzyme inhibitors, others)

Triggers are reviewed in more detail separately. (See "Hereditary angioedema: Epidemiology, clinical manifestations, exacerbating factors, and prognosis", section on 'Triggers and exacerbating factors'.)

Planning for acute treatment — All patients should be equipped with the following:

A medical information bracelet or necklace identifying the condition.

A written plan for treatment of HAE attacks for use in emergency department care. This document (also available in a wallet card) should briefly explain the patient's diagnosis, outline the indicated treatment for HAE attacks, and provide contact information for the supervising clinician (form 1 and form 2). (See "Hereditary angioedema: Acute treatment of angioedema attacks".)

Whenever possible, the ability to self-administer an effective drug outside of a medical facility. Patients should have a sufficient amount of medicine available and be trained in self-administration.

Baseline laboratory studies — Baseline studies are recommended to assess hepatic and other organ function (due to the potential need for chronic medications) and to screen for bloodborne illnesses (due to the possible need for blood products). The following tests are suggested: liver function tests (including alanine aminotransferase, total bilirubin); albumin; creatine kinase; blood urea nitrogen; creatinine; complete blood count and differential; pregnancy testing (if applicable); urinalysis; and screening tests for human immunodeficiency virus (HIV), hepatitis B, and hepatitis C [4]. A cholesterol panel should be obtained if the patient may be treated with androgens.

Vaccinations — Patients should be vaccinated against hepatitis B in case they require blood-derived products in the future [4].

Gynecologic and obstetric care — An international expert panel produced guidelines on the gynecologic and obstetric care of females with HAE [9].

Well-tolerated options for contraception include progesterone-based methods, such as progesterone-only oral contraceptives, other progesterone delivering methods, and intrauterine devices [10].

The effect of pregnancy on a given patient's frequency and pattern of HAE attacks is difficult to predict, although abdominal attacks usually become more frequent [11]. Disease activity during one pregnancy does not necessarily predict disease activity during subsequent pregnancies [12]. C1-INH is the preferred agent for prophylaxis and acute treatment of HAE attacks during pregnancy.

Labor and delivery generally do not precipitate HAE attacks, and the rate of cesarean deliveries is not elevated compared with the overall population. However, C1-INH concentrate should be available during delivery in case acute symptoms develop and for at least 48 hours afterward.

Lactation can cause an increase in HAE attacks in some females. C1-INH concentrate is preferred during lactation because the side effects of lanadelumab, berotralstat, tranexamic acid, and androgens on infants are not known.

The impact of menopause on HAE severity is variable.

Other issues, such as the management of polycystic ovary syndrome, infertility, and breast, uterine, and cervical cancers, are also discussed in the guidelines [9].

ACCESS TO "ON-DEMAND" THERAPIES — All patients should have access to therapies to treat acute episodes of angioedema, irrespective of whether they are receiving long-term prophylactic treatment. Options for on-demand therapy include plasma-derived C1-INH concentrate (human), recombinant C1-INH concentrate (human), icatibant, and ecallantide, as reviewed in detail separately. (See "Hereditary angioedema: Acute treatment of angioedema attacks", section on 'Overview'.)

APPROACH TO LONG-TERM PROPHYLAXIS — The consensus guidelines provide several options for long-term prophylaxis to prevent attacks of HAE-C1-INH, with which the authors agree, although practice differs somewhat around the world [1,4].

Overview of available therapies — The therapies used for long-term prophylaxis in patients with HAE-C1-INH are various preparations of C1 inhibitor concentrate, lanadelumab, berotralstat, antifibrinolytics, and attenuated androgens. Each has advantages and disadvantages, as summarized in the table (table 1). C1-INH concentrate, lanadelumab, and berotralstat are first-line therapies where available. Antifibrinolytics and attenuated androgens are alternatives that are less costly and are given orally, but they either have significant side effects (androgens) or are less effective (antifibrinolytics) than the first-line therapies.

C1-INH concentrate – Regular injections of plasma-derived C1 inhibitor concentrate (pdC1-INH) are effective and well tolerated by nearly all patients. pdC1-INH prophylaxis can be administered by intravenous (IV) or subcutaneous (SC) injection. When administered IV, repeated injections can lead to problems with venous access, and the placement of indwelling ports is discouraged because of risk of infection and thrombosis [13]. (See 'Intravenous plasma-derived C1 inhibitor' below.)

An SC preparation of pdC1-INH was approved for long-term prophylaxis in the United States in 2017 (Haegarda [brand name]). SC administration provides more consistent plasma levels compared with IV administration. It is also helpful for patients with difficult venous access. It is anticipated to become available in other countries soon. (See 'Subcutaneous C1 inhibitor' below.)

Recombinant human C1 inhibitor (rhC1-INH) is not approved for long-term prophylaxis, although, in a clinical trial, prophylaxis with rhC1-INH provided clinically relevant reductions in frequency of hereditary angioedema attacks and was well tolerated [14]. If shortages of pdC1-INH arise, rhC1-INH could be used instead. The suggested dose is 50 international units per kilogram twice weekly initially, rounding up to use the whole vial and avoid wasting drug and subsequently adjusted up or down to achieve control of symptoms. Use of rhC1-INH for acute treatment of HAE attacks is reviewed separately. (See "Hereditary angioedema: Acute treatment of angioedema attacks", section on 'Recombinant C1 inhibitor'.)

LanadelumabLanadelumab is a fully human monoclonal antibody to kallikrein, which became available in the United States in 2018. It is injected at a dose of 300 mg SC every two weeks and was shown to be safe and effective. In patients who experience no attacks after six months of treatment with lanadelumab, the dose may be reduced to 300 mg every four weeks. (See 'Lanadelumab' below.)

BerotralstatBerotralstat is a synthetic small molecule developed to inhibit plasma kallikrein, which can be administered orally as once-daily capsules (150 mg). It is effective and safe for prophylaxis of HAE attacks and was approved in United States in 2020. (See 'Berotralstat' below.)

Attenuated androgens – Long-term androgen therapy is usually effective but can have significant adverse effects. It is least problematic in postpubertal males, particularly when used at a low dose. It may be tolerated by females too, if the doses required to control symptoms are sufficiently low. It should not be used in growing children or pregnant or lactating females, unless there are extenuating circumstances. (See 'Attenuated androgens' below.)

Antifibrinolytics – Antifibrinolytic agents include tranexamic acid and epsilon aminocaproic acid (also called aminocaproic acid). These are less predictably effective for preventing HAE episodes compared with pdC1-INH concentrate, lanadelumab, or androgens, and some patients have problems tolerating them. However, antifibrinolytics can be useful for long-term prophylaxis in growing children and possibly in females who are pregnant or planning to become pregnant in the near future in whom androgens should not be used and pdC1-INH is not available. Once a female is pregnant or lactating, pdC1-INH concentrate is preferred for prophylaxis. Consensus guidelines do not emphasize the use of antifibrinolytics, but they remain an option [7]. (See 'Antifibrinolytics' below.)

Indications for long-term prophylaxis — The long‐term prophylaxis of HAE refers to the use of regular medication to reduce the burden of the disease by preventing/attenuating attacks in patients with confirmed HAE. Long-term prophylaxis is given to decrease the number, severity, and length of HAE attacks. Not all patients require it, and the decision to start a patient on long-term prophylaxis should be individualized based on maximizing quality of life [7]. Data are limited, but the authors estimate that from 20 (HF) to 70 percent (BZ) of their patient panels are on long-term prophylactic therapies. In contrast, in countries where on-demand therapies are less available, long-term prophylaxis is required by most symptomatic patients.

The decision to administer long-term prophylaxis to a specific patient should take into account the frequency and severity of attacks, access to emergency treatment, comorbid conditions, patient preference and experience, and failure to attain adequate control with appropriate on-demand therapies [5,15]. The therapeutic strategy should always be individualised. Patients should be evaluated for long‐term prophylaxis at every visit, or at least once a year. Patients receiving ongoing long‐term prophylaxis should be assessed regularly for efficacy and safety of the therapy, and dose and/or treatment interval should be adapted according to the clinical response. Trigger avoidance should be reviewed periodically to see if patients can avoid exacerbating factors more effectively. All patients using long‐term prophylaxis should also have access to on‐demand medication. (See 'Access to "on-demand" therapies' above.)

The decision about when it is appropriate to initiate long-term prophylaxis is approached somewhat differently around the world:

United States guidelines suggest that the decision to initiate long-term prophylaxis be individualized based upon multiple parameters, including attack frequency, comorbidities, patient preferences, and access to emergency care [7].

In some countries, regulatory authorities have stipulated that patients are eligible for long-term prophylaxis with twice-weekly injections of pdC1-INH if they are requiring four or more on-demand treatments per month, an approach that was adapted from hemophilia, where prophylactic injections of factor VIII are initiated when the number of on-demand approaches the number of prophylactic injections [16].

Goal of long-term prophylaxis — Long-term prophylaxis is deemed successful when symptoms are controlled to a level that is acceptable to that patient.

CHOICE OF AGENT IN SPECIFIC GROUPS — The options for long-term prophylaxis are intravenous (IV) or subcutaneous (SC) plasma-derived C1-INH (pdC1-INH) concentrate, lanadelumab (a monoclonal antibody to plasma kallikrein), berotralstat (an orally available plasma kallikrein inhibitor), attenuated androgens, and antifibrinolytics. Head-to-head trials are lacking. Within this group of therapies, pdC1-INH, lanadelumab, and berotralstat, which are all relatively costly, have either better efficacy or fewer adverse effects and are thus considered preferable to androgens and antifibrinolytics. The choice of which long-term prophylactic agent to use is influenced both by patient characteristics (age, sex, pregnancy/lactation), as well as regulatory requirements in different countries (table 2).

Prepubescent children — Prepubertal children can usually be managed with on-demand therapies alone because HAE symptoms are generally less severe prior to puberty. The expanded availability of these therapies for HAE attacks has greatly reduced the number of children who require long-term prophylaxis. However, for those who do require preventative therapy, several options are available and should be considered. The decision about which of these agents to use, if available, should be based on safety, tolerability, and patient preference.

SC or IV pdC1-INH or oral tranexamic acid is commonly used. Long-term prophylaxis with IV or SC pdC1-INH is approved down to the age of six years. Tranexamic acid is easiest to administer, while pdC1-INH is more effective.

Lanadelumab, which is approved for children aged two years and above in the United States, is a newer option. However, long-term safety data are lacking, and the role of plasma kallikrein in normal development has not been extensively studied.

We avoid androgens in children who require long-term prophylaxis.

Before the newer therapies became available, management options in children consisted of tranexamic acid and androgens, while C1-INH was usually reserved for life-threatening HAE attacks. The experience with these therapies remains relevant in resource-limited settings. One group published outcomes of a cohort of 48 children (23 boys and 25 girls) followed from diagnosis until age 18 years in which tranexamic acid was the initial agent given for long-term prophylaxis [17]. Overall, a relatively low percentage of children (19 percent) required long-term prophylaxis initially, compared with adult populations. However, by the age of 18 years, 43 percent required prophylaxis, with most needing to start long-term prophylaxis around the time of puberty. Some patients were adequately controlled with intermittent use of long-term prophylaxis during times when more attacks were anticipated (eg, during school exams, flu season, winter months, family crises, or puberty). If symptoms were not controlled by tranexamic acid, it was stopped and replaced with low-dose danazol (starting at 100 mg every other day). Only 2 of 48 patients required significant use of pdC1-INH concentrate. Using this approach, serious side effects of danazol were avoided in children of both sexes. Androgen therapy should be administered in consultation with an endocrinology specialist.

Postpubescent and adult males — The options preferred for pubertal and adult male patients are regular injections of pdC1-INH, lanadelumab, berotralstat, androgens, or tranexamic acid.

The approach of the American author (BZ) is to present all four treatment options, review the advantages and disadvantages of each, and make a joint decision about which therapy should be tried initially (table 1).

In more resource-limited settings, tranexamic acid and androgens are commonly used, and pdC1-INH is reserved for males with contraindications to or inadequate response to these therapies. If tranexamic acid is chosen as an initial prophylactic agent, a trial of full-dose therapy is undertaken for a few weeks. If tranexamic acid does not result in sufficient improvement, it should be stopped and androgen therapy initiated. Once attacks have subsided on androgens, most patients do not require high doses to keep the disease under control. Prophylaxis can also be given intermittently during times of anticipated stress to reduce overall exposure.

People not considering pregnancy in the near future — The agents that are preferred in females who are not considering pregnancy in the near future and who require preventative therapy are pdC1-INH, lanadelumab, berotralstat, and tranexamic acid. Attenuated androgens can also be used, provided symptoms can be controlled with lower doses. (See 'Gynecologic and obstetric care' above.)

The approach of the American author (BZ) is to present all treatment options, review the advantages and disadvantages of each, and make a joint decision about which therapy should be tried initially. Androgens are generally avoided.

Pregnant and lactating people — If long-term prophylaxis is required in a pregnant or lactating patient, the best-studied approach is regular injections of pdC1-INH concentrate [9]. If pdC1-INH injections are not tolerated (rare) or not available, tranexamic acid or fresh frozen plasma on demand may be used instead [12]. Lanadelumab and berotralstat have not been studied in pregnancy. (See 'Adverse effects and safety' below.)

SPECIFIC AGENTS — The agents that can be used for long-term prophylaxis are reviewed here, with mechanisms of action, availability, efficacy, adverse effects, contraindications, and monitoring [18]. Monitoring C4 or C1-INH antigenic levels for the purpose of assessing disease control is not indicated or useful, except possibly in the case of subcutaneous (SC) plasma-derived C1-INH (pdC1-INH). (See 'Subcutaneous C1 inhibitor' below.)

Prophylactic treatments may be divided into targeted therapies (eg, various forms of C1-INH replacement therapy, lanadelumab, and berotralstat), which were specifically developed for use in HAE-C1-INH or related disorders and are generally more effective, and nontargeted therapies (eg, androgens and antifibrinolytics) that were originally developed for other disorders and have proven useful in the clinical management of HAE.

C1 inhibitor replacement therapies

Intravenous plasma-derived C1 inhibitor — Plasma-derived C1 inhibitor concentrate (pdC1-INH) can be injected intravenously (IV) at regular intervals (continuous replacement therapy) to prevent attacks of angioedema. Two human pdC1-INH are available: Cinryze (brand name) and Berinert (brand name). pdC1-INH concentrate is available throughout Europe, the United Kingdom, Canada, and Argentina [4,19].

Indications — Continuous replacement therapy with IV pdC1-INH is a first-line option for patients requiring long-term prophylaxis [20,21]. In the United States, pdC1-INH is a first-line, long-term prophylactic agent for HAE-C1-INH without the need to have failed or experienced side effects from other medications such as androgens or antifibrinolytics. In some other countries, pdC1-INH may be restricted to patients who have had adverse effects to androgens or antifibrinolytics, were not adequately controlled on these agents, or who do not wish to take these agents. pdC1-INH concentrate is the treatment of choice for long-term prophylaxis during pregnancy [9]. (See 'Choice of agent in specific groups' above.)

Dosing and efficacy — Dosing of pdC1-INH Cinryze (brand name) for prophylaxis is 1000 units IV every three to four days. This dose may be adjusted over time to determine the lowest effective dose for each patient.

Small randomized trials and other studies have demonstrated that pdC1-INH concentrate is highly effective for long-term prophylaxis when given either on demand or on a regular schedule [20-24]. Earlier estimates of efficacy may be understated since the patients enrolled in these studies were refractory to other treatments and represented the severe end of the disease spectrum.

The first randomized, controlled trial of pdC1-INH for long-term prophylaxis studied six severely affected HAE-C1-INH patients who demonstrated significant decreases in attack frequency when receiving pdC1-INH 25 units per kilogram every third day for 17 days compared with placebo in a crossover design [20]. The phase-III trial included 24 patients randomized to pdC1-INH (1000 units every three to four days) or placebo injections for 12 weeks, after which the groups crossed over for another 12 weeks [21]. Subjects receiving pdC1-INH experienced approximately one-half as many attacks as those in the placebo group, and attacks were significantly shorter and less severe.

An open-label extension study of pdC1-INH (dose: 1000 units every three to four days) in 146 HAE patients for up to 2.6 years found an overall 93.7 percent reduction in the number of angioedema attacks compared with historical controls [24]. Just over one-third of patients experienced no attacks on prophylactic pdC1-INH. Another 7.5 percent of patients continued to experience frequent attacks, despite using the pdC1-INH, and no predictive marker for nonresponsiveness was found. No evidence for loss of effectiveness of the pdC1-INH over time was observed, which has also been noted in other studies [25].

Adverse effects — Adverse effects of IV pdC1-INH are rare and include headache and fever [26,27]. Other concerns are rare thrombotic events, hypersensitivity reactions [26,28], and the possibility of disease transmission.

Rare thrombotic events have been reported in association with long-term prophylaxis with pdC1-INH, particularly in patients with indwelling injection ports or those receiving higher-than-usual doses. A retrospective analysis of the US Food and Drug Administration (FDA) event reporting system database identified an association between thrombotic events and use of Cinryze (brand name), especially in patients with ports, although no conclusions about causality or frequency could be made from the data [29]. In a registry of patients receiving Berinert (brand name), there was one thrombotic event considered related to pdC1-INH, although this patient also had an indwelling port [27]. However, there are no data to suggest that pdC1-INH at registered doses is associated with an increased risk of thromboembolism in patients without indwelling ports [30]. A retrospective study of patients treated for many years with pdC1-INH found no evidence for an increased risk of thromboembolism during treatment with pdC1-INH, even in patients with multiple predisposing factors [31]. In addition, animal studies suggest that high-dose C1-INH therapy is antithrombotic rather than prothrombotic [32]. However, because of the uncertainty surrounding this issue, pdC1-INH product inserts include a warning that patients with risk factors for thromboembolic events (eg, ports, previous thrombosis, underlying atherosclerosis, oral contraceptives, androgens, obesity, or immobility) should be monitored for thrombotic events. In addition, the use of ports should be avoided in patients with HAE.

Treatment with any plasma-derived product, including pdC1-INH, confers some risk of disease transmission. Before the introduction of virucidal methods (from 1980 to 1985), a high percentage of HAE patients were infected with hepatitis C virus through a lyophilized preparation manufactured by Immuno Vienna (later Baxter). Production of the Baxter pdC1-INH product was subsequently discontinued in 2004. In response to this, pdC1-INH was subsequently vapor heated to inactivate viruses, and the source of plasma was changed to carefully screened donors and continually monitored for pathogens [33]. Since vapor-heated pdC1-INH became standard, only hepatitis G transmission has been reported, and the pathogenic significance of hepatitis G infection is unclear [34]. (See "Human pegivirus-1 infection".)

The preparations in use in most of the world are Berinert and Cinryze, which are virus inactivated by pasteurization and nanofiltered. Postmarketing surveillance for Berinert since 1985 has revealed no case of viral transmission, despite the administration of hundreds of millions of units [27,35-38]. Processing methods also remove prions [38]. Still, patients must be informed that there is a theoretical risk of transmission of unidentified pathogens.

Monitoring — The efficacy of pdC1-INH therapy should be assessed clinically by sufficient reduction of HAE attacks. Usefulness for monitoring C4 or C1-INH antigenic levels has not been tested. For patients receiving C1-INH concentrate, studies have not been undertaken to correlate successful therapy with specific C1-INH levels, and attacks may subside at levels of C1-INH that are still significantly below normal.

Periodic testing for hepatitis C and human immunodeficiency virus (HIV) is recommended for patients regularly receiving pdC1-INH.

Subcutaneous C1 inhibitor — The administration of a concentrated formulation of pdC1-INH SC is easier for most patients and also appears to be more effective for preventing attacks compared with IV administration. The SC formulation contains 1500 international units in 3 mL of water compared with the IV formulation, which contains 500 international units in 10 mL. In addition, SC administration results in more consistent plasma levels between doses compared with IV [39]. SC pdC1-INH is available for prophylaxis in the United States (Haegarda [brand name]) and in Europe (Sc Berinert 2000/3000 [brand name]) [40,41].

A SC formulation of Cinryze (brand name) is in development [42]. Although not yet available, this product would further expand options for SC therapy.

Dosing and efficacy — The dose of SC pdC1-INH for prophylaxis of HAE attacks in adolescents and adults is 60 international units per kilogram body weight twice weekly (every three or four days). It is injected SC into the abdominal area. Pending additional information about the use of higher doses, patients who do not achieve the desired level of control with the recommended dose should use the shortest interval between doses (ie, three days) to maximize efficacy.

Efficacy was demonstrated in a randomized, multicenter trial of 90 patients (12 years of age and older), in which participants self-administered one of two doses of SC pdC1-INH (either 40 or 60 international units per kilogram body weight) twice weekly or placebo for 16 weeks each in a crossover design [43]. The primary endpoint was number of episodes of angioedema, which was reduced 89 and 95 percent with the lower and higher dose, respectively, relative to placebo. For patients taking the higher dose, there were 3.51 fewer attacks per month compared with placebo (95% CI -4.21 to -2.81). The predominant adverse effect was mild injection-site reactions that did not lead to discontinuation. Earlier studies of twice-weekly injections of pdC1-INH demonstrated that SC injection resulted in higher and more sustained plasma levels relative to twice-weekly IV administration [39,43]. Higher levels of plasma C1-INH function correlated with a lower risk of attacks, with a functional level of approximately 40 percent being an inflection point for the relative risk of attacks. The study also showed that postinjection functional C1-INH plasma levels varied among patients, such that individualized dose adjustment may be needed for optimal disease control. In the open-label extension trial, the mean C1-INH functional trough level was 66 percent, and 83 percent of the 23 subjects receiving pdC1-INH for 30 months were attack free in the last six months [40]. Longer-duration studies are needed to assess long-term safety.

Compared with intravenous — Although studies direction comparing SC with IV pdC1-INH are not available, SC administration provides the advantage of more consistent plasma levels, and preliminary evidence suggests it provides better symptom control. An analysis of a subset of patients from a larger study examined attack rates in individuals who changed from IV to a SC preparation of pdC1-INH [44]. Although the design was not optimized for this assessment, the majority of patients changing from various doses of IV to 40 or 60 international units/kg of the SC preparation experienced clinically meaningful reductions in HAE attacks.

Adverse effects and monitoring — Adverse effects occurring in more than 4 percent of subjects treated with SC C1-INH in initial trials were injection-site reactions, hypersensitivity, nasopharyngitis, and dizziness. SC C1-INH carries a similar theoretical risk for disease transmission as IV pdC1-INH concentrate. Thromboembolic events have not been reported.

Higher levels of functional C1-INH correlated with lower attack risk in patients receiving SC C1-INH, in contrast to IV C1-INH, which does not result in a sustained increase in plasma levels of C1-INH function at clinically effective doses. As with other C1-INH formulations, periodic testing for HIV and hepatitis C should be done. (See 'Monitoring' above.)

Plasma kallikrein inhibitors — Two agents are available that inhibit plasma kallikrein: lanadelumab and berotralstat.

Lanadelumab — A fully human monoclonal antibody to plasma kallikrein, lanadelumab (brand name: Takhzyro), is approved by the US FDA for prophylaxis in patients 2 years of age and older and by the European Medicines Agency (EMA) for patients 12 years of age and older [45,46]. This agent limits the production of excess bradykinin by reducing plasma kallikrein activity (figure 1). It is injected SC, and its duration of action, which is significantly longer than other prophylactic therapies, allows dosing once or twice a month.

Dosing and efficacy — The recommended initial dose of lanadelumab is 300 mg injected SC every two weeks.

In the study on which approval was based, patients discontinued other prophylactic therapies completely before starting lanadelumab, but, in clinical practice, this may be less than ideal as it takes over two months to reach a steady state of lanadelumab. A conservative empiric approach is to continue the patient's current therapy for at least two weeks and possibly longer for those with frequent or severe attacks and to gradually reduce the dose of the previous prophylactic agent over an additional two weeks for a total of one month of overlap.

If a patient is stably attack free (eg, for more than six months), especially in patients with lower body weights, the dosing interval can be lengthened to 300 mg every four weeks.

The dose should not be increased, because higher doses have not been studied.

In a phase-Ib study, two doses of lanadelumab given 14 days apart reduced attack rates by between 88 and 100 percent [47]. A phase-III randomized trial involving 125 HAE patients showed a reduction in attacks ranging from 73 to 87 percent, depending on the dose and timing. Monthly attack rates were 1.64, 0.42, and 0.21 in subjects receiving placebo, 300 mg every four weeks, and 300 mg every two weeks, respectively [48].

Adverse effects and safety — Injection-site reactions were the most common adverse effect with lanadelumab, occurring in 45 to 56 percent of subjects receiving active drug compared with 34 percent of those receiving placebo. Increased aspartate and alanine transaminase levels occurred in 2 percent of active drug recipients versus 0 percent in the placebo group. They were asymptomatic and transient and did not require drug discontinuation. Lanadelumab can increase activated partial thromboplastin time but has not been associated with abnormal bleeding.

There are no known contraindications to use of lanadelumab. However, lanadelumab has not been studied in pregnancy or lactation, and, although animal studies in pregnancy are reassuring, we would avoid use in pregnant or lactating people until more information is available [49].

Berotralstat — Berotralstat (BCX7353) is an orally administered plasma kallikrein inhibitor that was approved by the US FDA in late 2020 for prevention of HAE attacks in adult and pediatric patients 12 years of age and older (figure 1) [50,51]. The drug binds to plasma kallikrein and inhibits its proteolytic activity. The dose is 150 mg once daily, taken with food. It will likely become available in other countries in the near future.

Safety and drug interactions — Higher doses can increase the risk of QT prolongation and are not recommended. A lower-strength dose of 110 mg is also available and is indicated in patients with moderate-to-severe hepatic impairment (Child-Pugh B or C) and in those taking breast cancer resistance protein (BCRP; eg, cyclosporine) or P-glycoprotein (P-gp) inhibiting medications (table 3). Berotralstat should not be used in patients taking P-gp inducers. The most common side effects are gastrointestinal symptoms (abdominal pain, vomiting, diarrhea, and gastroesophageal reflux) and back pain, all of which occurred in ≥10 percent of patients but generally subsided with continued use [52]. No specific laboratory monitoring is recommended by the manufacturer. Berotralstat has not been studied in pregnancy or lactation, although animal studies are reassuring [52].

Efficacy — Several trials have addressed efficacy and safety of berotralstat:

In the multicenter phase-II Angioedema Prophylaxis 1 (APeX-1) trial, 77 patients with a history of at least two attacks per month were randomized to placebo or four different doses of active drug at doses of 62.5, 125, 250, and 350 mg once daily for 28 days [53,54]. Once-daily oral administration of a dose of 125 mg or more resulted in a significantly lower rate of HAE attacks compared with placebo.

In a phase-III trial of 121 adolescents and adults with HAE (APeX-2), patients were randomized to either berotralstat 150 or 110 mg or placebo for 24 weeks to determine optimal dosing [55]. Patients had a median of 2.9 attacks per month after discontinuing other prophylactic medications. Response to berotralstat was defined as ≥50 percent reduction in that patient's HAE attack rate compared with baseline. Berotralstat demonstrated a significant reduction in attack rate at both 150 and 110 mg (1.31 and 1.65 attacks per month, respectively, compared with 2.35 in the placebo group). A response was seen in 58, 51, and 25 percent of patients receiving 150 mg, 110 mg, and placebo respectively. On the higher dose, 50 percent of patients achieved at least a 75 percent reduction in attacks, and 23 percent had at least a 90 percent reduction.

In a similar trial in Japan, APeX-J, orally administered, once-daily berotralstat 150 mg significantly reduced the frequency of HAE attacks and was safe and well tolerated [56].

APeX-S (NCT03472040) is a long-term safety study of berotralstat being conducted simultaneously with the APeX-2 study to evaluate the long-term effects of the 110 and 150 mg doses over a 48-week period.

Nontargeted therapies — Nontargeted therapies refer to medications that were originally developed for other disorders and have proven useful in the clinical management of HAE.

Attenuated androgens — Synthetic 17-alpha-alkylated androgens ("attenuated" or anabolic androgens) include danazol, stanozolol, tibolone, and methyltestosterone. The precise mechanism of action of attenuated androgens is unknown [57-59]. The choice of androgen is based largely upon availability since differences among drugs have not been well characterized. Danazol is widely available throughout the world. Stanozolol is not produced commercially in the United States, although it is available by prescription and can be compounded by individual pharmacies. Tibolone may be less virilizing than other agents [60-62]. Methyltestosterone is another alternative [63].

Efficacy — In a double-blind, placebo-controlled study of nine patients, high doses of androgen therapy (danazol, 600 mg daily) dramatically reduced the risk of at least one acute episode during a one-month treatment period from 94 to 2 percent and reduced attack rates from monthly to one every 10 months [64]. Other small, controlled studies and multiple observational studies also concluded that androgens were effective at preventing attacks [62,65-71]. A retrospective survey of 650 HAE patients revealed substantial variability in the efficacy of androgens [72].

Dosing — Most patients with HAE are controlled with doses ranging from 50 to 200 mg of danazol, either daily or every other day (or an equivalent dose of another androgen [eg, stanozolol 1 to 2 mg daily or every other day]). We suggest either of the following two ways of administering androgens for long-term prophylaxis: starting with a high dose and then tapering or starting with a low dose and gradually increasing (table 1) [4].

Initial high dose – This protocol is based on the strategy of initiating androgen therapy with high doses that are subsequently tapered as tolerated. This approach is favored for rapid control of attacks. Using danazol as an example, therapy is begun with 400 to 600 mg daily until new attacks have ceased (usually a few weeks). The dose can then be tapered over several weeks to 200 mg daily, although the guidelines suggest tapering more slowly (eg, by 100 mg daily per month or by one-third of the dose per month if there are no breakthroughs). When 200 mg daily is attained, therapy is tapered by 50 mg every two months and by every three months when below 100 mg per day. The minimum dose is 50 mg daily for five days every week for most patients. For those in whom a breakthrough attack occurs, remission can be reinduced by resuming the last effective dose and then using a higher maintenance dose.

The equivalent dosing for stanozolol is 4 to 6 mg daily for the first month and then gradual tapering to the minimal effective dose, which is generally in the range of 0.5 to 2 mg daily.

Initial low dose – This regimen involves the initiation of therapy with low doses, with increases in the dose as necessary. This approach minimizes medication side effects. Using danazol as an example, therapy is initiated at a dose of 100 mg daily for males and 50 mg daily for females. If there is no response after two weeks, the dose is increased to 200 mg daily (males)/100 mg daily (females) for two to four weeks.

General observations regarding the use of attenuated androgens for long-term prophylaxis include the following:

In postpubertal females, androgenic effects may be troublesome, and regulation of periods and use of other hormones can become issues. We suggest that treatment decisions be made with the cooperation of the patient's obstetrician/gynecologist.

Some patients have reported that increasing the dose of androgen in response to prodromal symptoms or at the first sign of swelling seems to help prevent progression to more severe symptoms. As an example, a patient taking 200 mg of danazol daily for prophylaxis could increase the dose to 400 mg three times daily and maintain the higher dose until there is some indication of clinical improvement or an attack ensues. Although there are anecdotal reports of efficacy, this intervention has not been studied, and opponents of the practice counter that androgens do not act quickly enough to be effective when used in this manner [4,73].

Adverse effects and contraindications — At the doses typically required (ie, 200 mg danazol daily or less), androgen therapy is usually tolerated by postpubertal males and by some female patients as well. However, most patients do experience some adverse effects, and there are important contraindications [72].

Adverse effects with long-term androgen administration may include weight gain, lipid abnormalities, arterial hypertension, virilization, abnormalities in serum transaminases, menstrual irregularities, postmenopausal bleeding, diminished libido, vasomotor symptoms, and depression [4,65,69,71-76]. These adverse effects can be controlled or minimized by using the lowest effective dose. Elevations of transaminases are most often seen with prolonged administration of higher doses (eg, danazol 400 mg or more daily or stanozolol 4 to 16 mg daily). There are also rare reports of hepatocellular adenomas and carcinoma with years of use [77,78]. Because of these concerns, daily doses greater than 200 mg per day are not recommended for long-term use [12]. Although androgen-related adverse effects are more common with higher doses, they are not always dose dependent, and some patients experience side effects at low doses [76]. A yearly measurement of alpha-fetoprotein and liver ultrasound was also suggested by the 2010 consensus guidelines as a possible means of detecting liver neoplasms, although not all experts agree that this is helpful.

Contraindications to long-term androgen use include childhood, pregnancy and lactation, liver disease, nephrotic syndrome, hypercholesterolemia, hypertension, mood disorders, and breast or prostate cancer, although rare exceptions are made.

Androgens should not be used in children prior to Tanner stage 5 (table 4). The main concern about androgen use in prepubertal children is the potential of these agents to cause premature epiphyseal closure and decreased growth [79]. However, findings of the only retrospective study conducted among pediatric patients suggest that treatment with the lowest effective doses of danazol does not influence growth [80].

Monitoring — The efficacy of androgen therapy should be assessed clinically by sufficient reduction of attacks. Monitoring C4 or C1-INH antigenic levels is not necessary or useful.

Laboratory values – The 2010 consensus guidelines recommend obtaining a complete blood count, alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, lipid profile, and urinalysis (to detect hematuria that can result from androgen use) every six months in patients taking long-term androgen therapy [4]. A yearly measurement of alpha-fetoprotein was also suggested by the 2010 consensus guidelines as a possible means of detecting liver neoplasms, although not all experts agree that this is helpful.

Ultrasonography – The risk of hepatic neoplasms may be increased with the long-term use of androgen therapy, both for patients with HAE and other diseases [78,81,82]. This risk appears to be small, although there are insufficient data to quantitate magnitude. In addition, it is not clear how best to detect focal neoplastic lesions, which may not necessarily result in elevations in serum transaminases [83]. As a result, not all of the authors of this review agree with the below suggestions.

In the absence of consensus, the 2010 guidelines suggest that ultrasonography of the liver be performed every six months for adult patients receiving more than 200 mg/day of danazol (or the equivalent of another androgen) and for all prepubertal patients receiving maintenance androgens [4]. Yearly screening may be sufficient for adults receiving 200 mg or less of danazol daily.

Antifibrinolytics — Antifibrinolytics interfere with the functions of plasminogen and plasmin, leading to decreased production of bradykinin, although the mechanism of action in angioedema remains poorly defined (figure 1) [84]. The only antifibrinolytic medication in widespread use for HAE is tranexamic acid. Another agent, epsilon aminocaproic acid (EACA), was shown to reduce the frequency of HAE attacks in a randomized trial in the 1970s [85]. However, EACA is less potent and has more gastrointestinal side effects compared with tranexamic acid, although some patients do well with it [36]. Antifibrinolytics effectively prevent attacks in a smaller proportion of patients compared with pdC1-INH.

In settings in which C1-INH replacement therapy is not available or access is limited, antifibrinolytics are generally preferable to androgens for long-term prophylaxis in pregnant people, children, and patients who do not tolerate androgens. Androgens should be avoided in patients with increased risk for thrombosis. Because pregnancy itself is a relative risk factor for thrombosis, C1-INH replacement therapy is a better choice for prophylaxis in pregnancy. However, one of the authors (HF) has some experience with using tranexamic acid successfully in pregnancy, particularly if the patient has taken the drug successfully before pregnancy, and it could be considered if C1-INH replacement therapy is not feasible. Of note, Cinryze (brand name) has been associated with a small number of cases of thrombosis as well. (See 'Adverse effects' above.)

Experience with tranexamic acid is greatest in Europe and Asia, where it has been available for decades [17,86,87]. In the United States, EACA has been available for many years, and an oral preparation of tranexamic acid became available in 2009 when it was approved by the US FDA for the treatment of menorrhagia. Antifibrinolytics are not US FDA approved for HAE, although this does not preclude their use.

Dosing and efficacy — In both children and adults, tranexamic acid is administered for long-term prophylaxis at a dose of 1 to 3 grams per day or at a total daily amount of 25 mg/kg, divided into two to three doses per day. It is available in a 500 mg dose in much of the world and in a 650 mg dose in the United States.

In children, we usually start with 250 mg twice daily for a few weeks and gradually increase if tolerated to 500 mg twice or three times daily.

In adolescent and adults, we start with 500 mg (or 650 mg of the United States preparation) two to three times daily for a few weeks and gradually increase if tolerated to 1 gram three times daily. Only approximately one-third of patients will achieve adequate control with doses below 3 grams per day. However, the dose of tranexamic acid must be reduced for patients with kidney impairment.

In observational studies of tranexamic acid, attack frequency and severity are reduced somewhat in approximately 70 percent of patients, although efficacy seems higher in some case series [36,88,89]. Overall, there is a general agreement that a sufficient response is seen in less than 30 percent. Still, tranexamic acid has been used successfully for long-term prophylaxis in some pediatric patients [90]. (See 'Prepubescent children' above.)

The usual prophylactic dose of epsilon aminocaproic acid (EACA) in adults is 1 to 2 grams per dose, given three times per day (ie, 3 to 6 grams daily). For children, the suggested dose is 0.05 g/kg per dose, given two times per day.

Adverse effects and contraindications — Antifibrinolytics carry the potential risk of intravascular thrombosis, although the true prothrombotic effects of these medications have not been quantified [91,92]. These agents should not be used in patients with increased thrombotic risk or history of ischemic events.

Additional side effects associated with these agents include the following:

Tranexamic acid uncommonly causes gastrointestinal distress (eg, nausea, vomiting, diarrhea), headache, hypotension, and anal pruritus, which can occur early in therapy [17,36]. There are also reports of retinal damage with prolonged use of higher doses of tranexamic acid [93].

Epsilon aminocaproic acid (EACA) is associated with several adverse effects at higher doses than are indicated for HAE [85,88]. However, at the doses typically used in HAE patients, gastrointestinal distress is typically the main adverse effect, and some patients tolerate it quite well.

Monitoring — The therapeutic success of tranexamic acid is determined clinically, and each patient should be maintained on the lowest dose that adequately prevents attacks.

Laboratory values – Doses are adjusted for decreased kidney function, so blood urea nitrogen and serum creatinine should be measured yearly. The 2010 consensus algorithm suggests that creatinine kinase, tests of liver and kidney function, and urinalysis be performed every six months, although some experts believe yearly evaluation is sufficient.

Ophthalmologic exam – Annual assessment of ocular pressure has been suggested for patients on long-term therapy, based on the theoretical concern that retinal thrombosis could block outflow of aqueous humor. Periodic ophthalmologic exams for patients on long-term therapy are suggested by some drug information databases because of early reports of ophthalmologic tumors in animals treated with high doses of tranexamic acid, although this has never been reported in humans. The authors have not performed ophthalmologic exams as part of monitoring therapy.

INVESTIGATIONAL THERAPIES — Several additional drugs for long-term prophylaxis of HAE are in human trials.  

Garadacimab (previously known as CSL312) is a fully human, immunoglobulin G4 (IgG4) type recombinant monoclonal antibody, which binds to the catalytic site of FXIIa, blocking its proteolytic activity (figure 1). Efficacy, pharmacokinetics, and safety of subcutaneously (SC) administered CSL312 as prophylaxis to prevent HAE attacks were investigated in a multicenter, phase-II study. The number of attacks was reduced compared with placebo. Mean percentage reductions with three doses of garadacimab were 89, 99, and 91 versus placebo, and the drug was well tolerated [94].

KVD824 is a small molecule that inhibits plasma kallikrein, limiting the generation of bradykinin. In a phase-I trial, 84 healthy volunteers received KVD824 either as a single dose of up to 1280 mg or as multiple doses of up to 640 mg each. The trial investigated the safety, tolerability, and pharmacokinetics of the treatment at each of the doses and also studied the effect of food on pharmacokinetics. Participants tolerated all doses well. Another study is underway in the United Kingdom assessing single and multiple doses of KVD824 in healthy volunteers. Phase-II Clinical Trial of KVD824 as an Oral Prophylactic Treatment for Hereditary Angioedema is in planning.

IONIS-PKK-LRx (donidalorsen) is a ligand-conjugated antisense oligonucleotide designed to inhibit prekallikrein activity, which is under development as a prophylactic agent for bradykinin-mediated angioedema. It is intended for receptor-mediated delivery to hepatocytes. In a phase-I study, IONIS-PKK-LRx was well tolerated and reduced plasma kallikrein activity and levels in a dose-dependent manner [95]. In a pilot compassionate-use study, subcutaneous (SC) injections in two patients with different types of severe bradykinin-mediated angioedema over a several-month period was accompanied by a reduction in the angioedema attack rate [96]. In a small phase-II study of 20 patients (14 received active drug), monthly attack rate was 0.23 (95% CI 0.08-0.39) among patients receiving donidalorsen and 2.21 (95% CI 0.58-3.85) among those receiving placebo [97]. The medication was administered as a monthly SC injection.

ALN-F12 is a double-stranded small interfering ribonucleic acid (siRNA) inhibitor of FXII conjugated to an N-acetylgalactosamine ligand, which suppresses the synthesis of the FXII protein and reduces vascular permeability. RNA interference-mediated reduction of FXII was shown to mitigate excess bradykinin stimulation in an animal model and in an ex vivo human plasma assay [98].

PHA-022121 is a novel small-molecule antagonist of the bradykinin 2 (B2) receptor that is administered orally. PHA-022121 inhibited the bradykinin-induced changes in blood pressure at all doses tested one hour after administration. Based on preclinical experimental data and modelling, this compound is predicted to be effective for prevention of HAE attacks [99].

RESOURCES FOR PATIENTS — Patients can access detailed, patient-oriented information through the United States Hereditary Angioedema Association and the International Patient Organization for C1 Inhibitor Deficiencies.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Hereditary angioedema and other forms of nonhistaminergic angioedema".)

SUMMARY AND RECOMMENDATIONS

General care – General measures for patients with hereditary angioedema (HAE) include education, testing of family members, identification and avoidance of possible triggers, and planning for HAE attacks. Common triggers include dental and medical procedures, periods of stress, menstruation, pregnancy, infections, and certain medications, such as estrogens and angiotensin-converting enzyme inhibitors. (See 'General care' above.)

Planning for angioedema attacks – Immediately upon diagnosis, clinicians should help construct a plan for emergency care, including how and where the patient will access treatment and education about the need to seek care in a medical facility for laryngeal attacks. In countries where on-demand therapies (ie, C1 inhibitor concentrate [C1-INH], icatibant, or ecallantide) are available, all patients should have access to these medications irrespective of whether they receive long-term prophylaxis. Patients should be offered a medical identification bracelet and a written plan for treatment of HAE attacks for use in emergency department care (form 1 and form 2). (See 'Planning for acute treatment' above and 'Access to "on-demand" therapies' above.)

Indications for long-term prophylaxis – Long-term prophylaxis is indicated for patients who do not achieve adequate control of their disease with on-demand therapy alone. The decision to start long-term prophylactic treatment should be individualized, and the approach may differ somewhat around the world. (See 'Approach to long-term prophylaxis' above.)

Available medications – The options for long-term prophylaxis are regularly injected (intravenously [IV] or subcutaneously [SC]) plasma-derived C1-INH (pdC1-INH), lanadelumab (a monoclonal antibody to plasma kallikrein), berotralstat (an orally available plasma kallikrein inhibitor), attenuated androgens, and antifibrinolytics. Each agent has advantages and disadvantages (table 1). (See 'Overview of available therapies' above.)

The choice of which long-term prophylactic agent to use is influenced both by patient characteristics (age, sex, pregnancy/lactation) as well as regulatory requirements in different countries (table 2). There are few studies directly comparing the different therapies with each other. (See 'Choice of agent in specific groups' above.)

For patients requiring prophylaxis for severe disease (ie, laryngeal attacks or frequent attacks of other types) we recommend pdC1-INH, lanadelumab, or berotralstat over other agents (Grade 1B). (See 'Specific agents' above.)

-For patients receiving pdC1-INH, we suggest SC in preference to IV administration (Grade 2C). (See 'Compared with intravenous' above.)

-During pregnancy and lactation, we suggest pdC1-INH in preference to other agents (Grade 2C). If pdC1-INH is not available, we suggest tranexamic acid (Grade 2C). (See 'Pregnant and lactating people' above.)

If pdC1-INH, lanadelumab, or berotralstat are not available, tranexamic acid or androgens are options. Androgens are more effective but are contraindicated in children younger than Tanner stage 5 (table 4), pregnant or lactating people, and patients with certain comorbidities. Tranexamic acid is less effective but has fewer side effects. (See 'Attenuated androgens' above and 'Antifibrinolytics' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Marco Cicardi, MD, who contributed to earlier versions of this topic review.

  1. 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; 67:147.
  2. Kemper C, Pangburn MK, Fishelson Z. Complement nomenclature 2014. Mol Immunol 2014; 61:56.
  3. 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; 69:602.
  4. Bowen T, Cicardi M, Farkas H, et al. 2010 International consensus algorithm for the diagnosis, therapy and management of hereditary angioedema. Allergy Asthma Clin Immunol 2010; 6:24.
  5. 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; 73:1575.
  6. Betschel S, Badiou J, Binkley K, et al. The International/Canadian Hereditary Angioedema Guideline. Allergy Asthma Clin Immunol 2019; 15:72.
  7. 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.
  8. Bork K, Hardt J, Witzke G. Fatal laryngeal attacks and mortality in hereditary angioedema due to C1-INH deficiency. J Allergy Clin Immunol 2012; 130:692.
  9. Caballero T, Farkas H, Bouillet L, et al. 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; 129:308.
  10. Bouillet L, Longhurst H, Boccon-Gibod I, et al. Disease expression in women with hereditary angioedema. Am J Obstet Gynecol 2008; 199:484.e1.
  11. 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; 203:131.e1.
  12. Craig T, Aygören-Pürsün E, Bork K, et al. WAO Guideline for the Management of Hereditary Angioedema. World Allergy Organ J 2012; 5:182.
  13. Yoo SM, Khan DA. Implantable venous access device associated complications in patients with hereditary angioedema. J Allergy Clin Immunol Pract 2013; 1:524.
  14. 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; 390:1595.
  15. Zuraw BL, Bernstein JA, Lang DM, et al. A focused parameter update: hereditary angioedema, acquired C1 inhibitor deficiency, and angiotensin-converting enzyme inhibitor-associated angioedema. J Allergy Clin Immunol 2013; 131:1491.
  16. Berntorp E, Shapiro AD. Modern haemophilia care. Lancet 2012; 379:1447.
  17. Farkas H, Csuka D, Zotter Z, et al. Prophylactic therapy in children with hereditary angioedema. J Allergy Clin Immunol 2013; 131:579.
  18. Frank MM. 8. Hereditary angioedema. J Allergy Clin Immunol 2008; 121:S398.
  19. Gompels MM, Lock RJ, Abinun M, et al. C1 inhibitor deficiency: consensus document. Clin Exp Immunol 2005; 139:379.
  20. Waytes AT, Rosen FS, Frank MM. Treatment of hereditary angioedema with a vapor-heated C1 inhibitor concentrate. N Engl J Med 1996; 334:1630.
  21. Zuraw BL, Busse PJ, White M, et al. Nanofiltered C1 inhibitor concentrate for treatment of hereditary angioedema. N Engl J Med 2010; 363:513.
  22. 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; 117:904.
  23. Kreuz W, Rusicke E, Martinez-Saguer I, et al. Home therapy with intravenous human C1-inhibitor in children and adolescents with hereditary angioedema. Transfusion 2012; 52:100.
  24. Zuraw BL, Kalfus I. Safety and efficacy of prophylactic nanofiltered C1-inhibitor in hereditary angioedema. Am J Med 2012; 125:938.e1.
  25. Craig TJ, Bewtra AK, Hurewitz D, et al. Treatment response after repeated administration of C1 esterase inhibitor for successive acute hereditary angioedema attacks. Allergy Asthma Proc 2012; 33:354.
  26. Cicardi M, Zingale L. How do we treat patients with hereditary angioedema. Transfus Apher Sci 2003; 29:221.
  27. Riedl MA, Bygum A, Lumry W, et al. Safety and Usage of C1-Inhibitor in Hereditary Angioedema: Berinert Registry Data. J Allergy Clin Immunol Pract 2016; 4:963.
  28. Zingale LC, Pappalardo E, Zanichelli A, Cicardi M. C1 inhibitor concentrate: efficacy and adverse reaction. Int Immunopharmacol 2002; 318:1385.
  29. Gandhi PK, Gentry WM, Bottorff MB. Thrombotic events associated with C1 esterase inhibitor products in patients with hereditary angioedema: investigation from the United States Food and Drug Administration adverse event reporting system database. Pharmacotherapy 2012; 32:902.
  30. Busse P, Bygum A, Edelman J, et al. Safety of C1-esterase inhibitor in acute and prophylactic therapy of hereditary angioedema: findings from the ongoing international Berinert patient registry. J Allergy Clin Immunol Pract 2015; 3:213.
  31. Farkas H, Kőhalmi KV, Veszeli N, et al. Risk of thromboembolism in patients with hereditary angioedema treated with plasma-derived C1-inhibitor. Allergy Asthma Proc 2016; 37:164.
  32. Schürmann D, Herzog E, Raquet E, et al. C1-esterase inhibitor treatment: preclinical safety aspects on the potential prothrombotic risk. Thromb Haemost 2014; 112:960.
  33. Cicardi M, Mannucci PM, Castelli R, et al. Reduction in transmission of hepatitis C after the introduction of a heat-treatment step in the production of C1-inhibitor concentrate. Transfusion 1995; 35:209.
  34. De Filippi F, Castelli R, Cicardi M, et al. Transmission of hepatitis G virus in patients with angioedema treated with steam-heated plasma concentrates of C1 inhibitor. Transfusion 1998; 38:307.
  35. Farkas H, Jakab L, Temesszentandrási G, et al. Hereditary angioedema: a decade of human C1-inhibitor concentrate therapy. J Allergy Clin Immunol 2007; 120:941.
  36. 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:S51.
  37. Cicardi M, Zingale LC, Zanichelli A, et al. The use of plasma-derived C1 inhibitor in the treatment of hereditary angioedema. Expert Opin Pharmacother 2007; 8:3173.
  38. Gröner A, Nowak T, Schäfer W. Pathogen safety of human C1 esterase inhibitor concentrate. Transfusion 2012; 52:2104.
  39. 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; 70:1319.
  40. Craig T, Zuraw B, Longhurst H, et al. Long-Term Outcomes with Subcutaneous C1-Inhibitor Replacement Therapy for Prevention of Hereditary Angioedema Attacks. J Allergy Clin Immunol Pract 2019; 7:1793.
  41. US FDA approval letter: https://www.fda.gov/downloads/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/LicensedProductsBLAs/FractionatedPlasmaProducts/UCM564383.pdf (Accessed on June 27, 2017).
  42. Lumry WR, Martinez-Saguer I, Yang WH, et al. Fixed-Dose Subcutaneous C1-Inhibitor Liquid for Prophylactic Treatment of C1-INH-HAE: SAHARA Randomized Study. J Allergy Clin Immunol Pract 2019; 7:1610.
  43. Longhurst H, Cicardi M, Craig T, et al. Prevention of Hereditary Angioedema Attacks with a Subcutaneous C1 Inhibitor. N Engl J Med 2017; 376:1131.
  44. 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; 7:2035.
  45. https://www.ema.europa.eu/en/medicines/human/EPAR/takhzyro (Accessed on November 12, 2019).
  46. US FDA approval is available at https://www.fda.gov/Drugs/DrugSafety/ucm618261.htm (Accessed on August 28, 2018).
  47. Banerji A, Busse P, Shennak M, et al. Inhibiting Plasma Kallikrein for Hereditary Angioedema Prophylaxis. N Engl J Med 2017; 376:717.
  48. 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; 320:2108.
  49. Prescribing information is available at https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761090s000lbl.pdf (Accessed on August 28, 2018).
  50. Hwang JR, Hwang G, Johri A, Craig T. Oral plasma kallikrein inhibitor BCX7353 for treatment of hereditary angioedema. Immunotherapy 2019; 11:1439.
  51. Unites States FDA approval letter: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2020/214094Orig1s000ltr.pdf (Accessed on December 09, 2020).
  52. Product insert: https://biocryst.com/wp-content/uploads/2020/12/ORLADEYO_PI_V1_2020.pdf (Accessed on December 09, 2020).
  53. Efficacy and safety of BCX7353 to prevent angioedema attacks in subjects with hereditary angioedema [APeX-1]; NCT02870972. https://clinicaltrials.gov/ct2/show/NCT02870972 (Accessed on April 04, 2017).
  54. Aygören-Pürsün E, Bygum A, Grivcheva-Panovska V, et al. Oral Plasma Kallikrein Inhibitor for Prophylaxis in Hereditary Angioedema. N Engl J Med 2018; 379:352.
  55. 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; 148:164.
  56. Ohsawa I, Honda D, Suzuki Y, et al. Oral berotralstat for the prophylaxis of hereditary angioedema attacks in patients in Japan: A phase 3 randomized trial. Allergy 2021; 76:1789.
  57. Johnson AM, Alper CA, Rosen FS, Craig JM. C1 inhibitor: evidence for decreased hepatic synthesis in hereditary angioneurotic edema. Science 1971; 173:553.
  58. Pappalardo E, Zingale LC, Cicardi M. Increased expression of C1-inhibitor mRNA in patients with hereditary angioedema treated with Danazol. Immunol Lett 2003; 86:271.
  59. 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; 121:429.
  60. Ott HW, Mattle V, Hadziomerovic D, et al. Treatment of hereditary angioneurotic oedema (HANE) with tibolone. Clin Endocrinol (Oxf) 2007; 66:180.
  61. FOX M, MINOT AS, LIDDLE GW. Oxandrolone: a potent anabolic steroid of novel chemical configuration. J Clin Endocrinol Metab 1962; 22:921.
  62. Church JA. Oxandrolone treatment of childhood hereditary angioedema. Ann Allergy Asthma Immunol 2004; 92:377.
  63. Sheffer AL, Fearon DT, Austen KF. Methyltestosterone therapy in hereditary angioedema. Ann Intern Med 1977; 86:306.
  64. 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; 295:1444.
  65. Cicardi M, Castelli R, Zingale LC, Agostoni A. Side effects of long-term prophylaxis with attenuated androgens in hereditary angioedema: comparison of treated and untreated patients. J Allergy Clin Immunol 1997; 99:194.
  66. Cicardi M, Bergamaschini L, Tucci A, et al. Morphologic evaluation of the liver in hereditary angioedema patients on long-term treatment with androgen derivatives. J Allergy Clin Immunol 1983; 72:294.
  67. Sheffer AL, Fearon DT, Austen KF. Hereditary angioedema: a decade of management with stanozolol. J Allergy Clin Immunol 1987; 80:855.
  68. Sheffer AL, Fearon DT, Austen KF. Clinical and biochemical effects of impeded androgen (oxymetholone) therapy of hereditary angioedema. J Allergy Clin Immunol 1979; 64:275.
  69. 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; 100:153.
  70. Füst G, Farkas H, Csuka D, et al. Long-term efficacy of danazol treatment in hereditary angioedema. Eur J Clin Invest 2011; 41:256.
  71. Riedl MA. Critical appraisal of androgen use in hereditary angioedema: a systematic review. Ann Allergy Asthma Immunol 2015; 114:281.
  72. Zuraw BL, Davis DK, Castaldo AJ, Christiansen SC. Tolerability and Effectiveness of 17-α-Alkylated Androgen Therapy for Hereditary Angioedema: A Re-examination. J Allergy Clin Immunol Pract 2016; 4:948.
  73. Széplaki G, Varga L, Valentin S, et al. Adverse effects of danazol prophylaxis on the lipid profiles of patients with hereditary angioedema. J Allergy Clin Immunol 2005; 115:864.
  74. Sloane DE, Lee CW, Sheffer AL. Hereditary angioedema: Safety of long-term stanozolol therapy. J Allergy Clin Immunol 2007; 120:654.
  75. Zotter Z, Veszeli N, Csuka D, et al. Frequency of the virilising effects of attenuated androgens reported by women with hereditary angioedema. Orphanet J Rare Dis 2014; 9:205.
  76. Sheffer AL, Fearon DT, Austen KF. Clinical and biochemical effects of stanozolol therapy for hereditary angioedema. J Allergy Clin Immunol 1981; 68:181.
  77. Bork K, Schneiders V. Danazol-induced hepatocellular adenoma in patients with hereditary angio-oedema. J Hepatol 2002; 36:707.
  78. Crampon D, Barnoud R, Durand M, et al. Danazol therapy: an unusual aetiology of hepatocellular carcinoma. J Hepatol 1998; 29:1035.
  79. 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.
  80. Kőhalmi KV, Veszeli N, Luczay A, et al. [Effect of danazol treatment on growth in pediatric patients with hereditary angioedema due to C1-inhibitor deficiency]. Orv Hetil 2017; 158:1269.
  81. Zuraw BL. Clinical practice. Hereditary angioedema. N Engl J Med 2008; 359:1027.
  82. Bork K, Pitton M, Harten P, Koch P. Hepatocellular adenomas in patients taking danazol for hereditary angio-oedema. Lancet 1999; 353:1066.
  83. O'Neil KM. Complement deficiency. Clin Rev Allergy Immunol 2000; 19:83.
  84. Longstaff C. Studies on the mechanisms of action of aprotinin and tranexamic acid as plasmin inhibitors and antifibrinolytic agents. Blood Coagul Fibrinolysis 1994; 5:537.
  85. Frank MM, Sergent JS, Kane MA, Alling DW. Epsilon aminocaproic acid therapy of hereditary angioneurotic edema. A double-blind study. N Engl J Med 1972; 286:808.
  86. Ohsawa I, Honda D, Nagamachi S, et al. Clinical manifestations, diagnosis, and treatment of hereditary angioedema: survey data from 94 physicians in Japan. Ann Allergy Asthma Immunol 2015; 114:492.
  87. Wintenberger C, Boccon-Gibod I, Launay D, et al. Tranexamic acid as maintenance treatment for non-histaminergic angioedema: analysis of efficacy and safety in 37 patients. Clin Exp Immunol 2014; 178:112.
  88. Gwynn CM. Therapy in hereditary angioneurotic oedema. Arch Dis Child 1974; 49:636.
  89. Agostoni A, Marasini B, Cicardi M, et al. Hepatic function and fibrinolysis in patients with hereditary angioedema undergoing long-term treatment with tranexamic acid. Allergy 1978; 33:216.
  90. Farkas H, Varga L, Széplaki G, et al. Management of hereditary angioedema in pediatric patients. Pediatrics 2007; 120:e713.
  91. Berntorp E, Follrud C, Lethagen S. No increased risk of venous thrombosis in women taking tranexamic acid. Thromb Haemost 2001; 86:714.
  92. Taparia M, Cordingley FT, Leahy MF. Pulmonary embolism associated with tranexamic acid in severe acquired haemophilia. Eur J Haematol 2002; 68:307.
  93. Snir M, Axer-Siegel R, Buckman G, Yassur Y. Central venous stasis retinopathy following the use of tranexamic acid. Retina 1990; 10:181.
  94. Davoine C, Bouckaert C, Fillet M, Pochet L. Factor XII/XIIa inhibitors: Their discovery, development, and potential indications. Eur J Med Chem 2020; 208:112753.
  95. Ferrone JD, Bhattacharjee G, Revenko AS, et al. IONIS-PKKRx a Novel Antisense Inhibitor of Prekallikrein and Bradykinin Production. Nucleic Acid Ther 2019; 29:82.
  96. Cohn DM, Viney NJ, Fijen LM, et al. Antisense Inhibition of Prekallikrein to Control Hereditary Angioedema. N Engl J Med 2020; 383:1242.
  97. Fijen LM, Riedl MA, Bordone L, et al. Inhibition of Prekallikrein for Hereditary Angioedema. N Engl J Med 2022; 386:1026.
  98. Liu J, Qin J, Borodovsky A, et al. An investigational RNAi therapeutic targeting Factor XII (ALN-F12) for the treatment of hereditary angioedema. RNA 2019; 25:255.
  99. Lesage ASJ, Knolle J, Marceua F, et al. PHA-022121, the first-in-class orally active bradykinin receptor B2 antagonist for on-demand and prophylactic treatment of HAE [Abstract]. Allergy Asthma Clin Immunol 2019; 15:O23.
Topic 8100 Version 57.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟