Skip to main content
Erschienen in: Allergy, Asthma & Clinical Immunology 1/2020

Open Access 01.12.2020 | Research

Co-occurrence between C1 esterase inhibitor deficiency and autoimmune disease: a systematic literature review

verfasst von: Donald Levy, Timothy Craig, Paul K. Keith, Girishanthy Krishnarajah, Rachel Beckerman, Subhransu Prusty

Erschienen in: Allergy, Asthma & Clinical Immunology | Ausgabe 1/2020

Abstract

Background

Hereditary angioedema (HAE) is caused by a SERPING1 gene defect resulting in decreased (Type I) or dysfunctional (Type II) C1 esterase inhibitor (C1-INH). The prevalence of autoimmune diseases (ADs) in patients with HAE appears to be higher than the general population. A systematic literature review was conducted to examine the co-occurrence between HAE and ADs.

Methods

PubMed/EMBASE were searched for English-language reviews, case reports, observational studies, retrospective studies, and randomized controlled trials up to 04/15/2018 (04/15/2015-04/15/2018 for EMBASE) that mentioned patients with HAE Type I or II and comorbid ADs. Non-human or in vitro studies and publications of C1-INH deficiency secondary to lymphoproliferative disorders or angiotensin-converting-enzyme inhibitors were excluded.

Results

Of the 2880 records screened, 76 met the eligibility criteria and 155 individual occurrences of co-occurring HAE and AD were mentioned. The most common ADs were systemic lupus erythematosus (30 mentions), thyroid disease (21 mentions), and glomerulonephritis (16 mentions). When ADs were grouped by MedDRA v21.0 High Level Terms, the most common were: Lupus Erythematosus and Associated Conditions, n = 52; Endocrine Autoimmune Disorders, n = 21; Gastrointestinal Inflammatory Conditions, n = 16; Glomerulonephritis and Nephrotic Syndrome, n = 16; Rheumatoid Arthritis and Associated Conditions, n = 11; Eye, Salivary Gland and Connective Tissue Disorders, n = 10; and Immune and Associated Conditions Not Elsewhere Classified, n = 5.

Conclusions

Based on literature reports, systemic lupus erythematosus is the most common AD co-occurring with HAE Type I and II. Cause and effect for co-occurring HAE and AD has not been clinically established but could be related to lack of sufficient C1-INH function.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AD
Autoimmune disease
C1-INH
C1 esterase inhibitor
GN
Glomerulonephritis
HAE
Hereditary angioedema
MASP
Mannose-binding lectin-associated serine protease
RA
Rheumatoid arthritis
SERPING1
Serine protease inhibitor gene 1 present on the long arm of chromosome 11
SLE
Systemic lupus erythematosus

Background

The human C1 esterase inhibitor (C1-INH) binds to proteases involved in the initiation of complement pathways, the kallikrein-kinin system (often referred to as the “contact system”), fibrinolysis, and the coagulation cascade [1]. As a regulatory protein, C1-INH downregulates the production of the vasodilator bradykinin in the contact system [1]. In hereditary angioedema (HAE), genetic defects in the C1-INH gene (Serine Protease Inhibitor Gene 1 present on the Long Arm of Chromosome 11 [11q]: SERPING1) can result in a deficient or dysfunctional protein (HAE Type I and II, respectively [HAE-C1INH]) [2, 3]. Subsequently, extravasation of plasma into cutaneous or mucosal tissues can result in angioedema [4, 5]. Patients with HAE may experience angioedema of the face, extremities, intestines, and genitals, and may also experience life-threatening laryngeal edema. Recommended treatments for acute HAE attacks include plasma-derived C1-INH concentrate, recombinant C1-INH (conestat alfa), the bradykinin B2 receptor antagonist icatibant, and the recombinant kallikrein inhibitor ecallantide [69]. Therapies for prophylaxis include intravenous and subcutaneous C1-INH (plasma-derived preferred), a monoclonal antibody against kallikrein (lanadelumab), androgens, and tranexamic acid [610].
One of the main functions of C1-INH is regulation of the complement pathway by preventing excessive activation of C4 and C2 via inhibition of the complement proteases C1s in the classical pathway, and mannose-binding lectin-associated serine protease 1/2 (MASP1 and MASP2) in the lectin pathway [1, 11]. C1-INH also appears to regulate the alternative complement pathway via binding of C3b [12]. The deficient function of C1-INH in patients with HAE results in autoactivation of C1s, leading to chronic activation and consumption of C4 and other early components of the complement system, with a corresponding decrease in levels of circulating plasma C4 [13, 14]. C2 levels often decline during acute attacks [13].
Although the complement system is best known for its contribution to the defense from microbial pathogens, it also contributes to protection against the development of autoimmune disease (AD) through multiple mechanisms. These protective mechanisms include promotion of antigen/antibody immune complex clearance, clearance of apoptotic cells that could become a source of autoantigens, and contribution to tolerance to self-antigens [1517]. Genetic deficiencies in early components of complement (C1, C4, and C2) increase the risk of development of the ADs systemic lupus erythematosus (SLE) and glomerulonephritis (GN), presumably because of a decrease in these protective complement mechanisms [17, 18]. Hereditary C1-INH deficiency also appears to be associated with SLE and GN [1922], although there are conflicting data regarding the association between HAE-C1INH and the risk of developing AD in general [14, 2326]. The prevalence of ADs in patients with HAE is 12% compared with 4.5% (2.7% for males and 6.4% for females) in the general population [26, 27].
The objective of the current systematic literature review was to examine the number and type of co-occurring ADs reported in patients with HAE-C1INH based on relevant published mentions.

Methods

A protocol for the systematic literature review was developed and registered in PROSPERO (CRD42018096855).

Data sources and search strategy

Databases searched were PubMed/MEDLINE and EMBASE. PubMed/MEDLINE searches were limited to English publications up to the date April 15, 2018. EMBASE searches were limited to English abstracts from April 15, 2015 through April 15, 2018. Search strings based on key words or MeSH terms were used. The key word search string was “C1 AND inhibitor AND angioedema” and the MeSH term search string was “Complement C1 Inhibitor Protein”[MeSH]) AND “Angioedema”[MeSH]”. Search terms specific to ADs were intentionally not included in the search string since such a large number of ADs exist and it would not be possible to capture all potential ADs. When necessary to address the secondary objectives of the review, non-systematic PubMed searches were conducted to gather general information regarding genetics, etiology, and pathogenesis of HAE and ADs.

Eligibility criteria

Review articles, case reports, observational studies, retrospective studies, and randomized controlled trials written in English in manuscript or congress abstract form were eligible for inclusion. Only published records that mentioned an AD in association with HAE Type I and II were included. Acquired angioedema (i.e., secondary to lymphoproliferative disorders) is also known to be associated with ADs but was excluded because the focus of this literature review was HAE-C1INH. Additional exclusions were HAE with normal C1-INH (HAE Type III), acquired angioedema associated with angiotensin converting enzyme inhibitors, non-human studies, and in vitro studies.

Record selection and data collection processes

Titles and abstracts of records identified in the PubMed/MEDLINE and EMBASE searches were reviewed (in duplicate) by two reviewers to identify records of potential importance and, where discrepancies existed as to selected records, resolution was facilitated by a third reviewer. Full-length copies of the preliminarily identified manuscripts were obtained and reviewed by two reviewers within the context of the inclusion and exclusion criteria and reasons for any study exclusions were documented. Any discrepancies at the conclusion of the initial review process were resolved with the assistance of a third reviewer.
Records meeting the inclusion criteria and none of the exclusion criteria were reviewed by one reviewer and relevant information and data were extracted. Data were compiled into tables in an Excel format. A second reviewer conducted a quality assurance check with a duplicate review of approximately 10% of the records. When a record mentioned more than one patient with comorbid HAE-C1INH and AD, data from each patient was extracted individually and counted as a single mention. ADs extracted from the records were classified by MedDRA v21.0 High Level Terms (HLT).
The systematic literature review was intended to be exploratory and hypothesis generating. Furthermore, a broad scope of literature beyond clinical trials was assessed and risk of bias was not assessed.

Results

In all, 2880 titles and abstracts were screened and 245 were selected for full-text review. After full-text review, a total of 76 records were eligible for data extraction (Fig. 1). A total of 56/76 records were published as full-text manuscripts and 20/76 records were congress abstracts only. The majority of the 76 records (n = 48; 67%) were case reports or case series.
From the 76 records, 155 individual occurrences of HAE-C1INH and AD comorbidity were mentioned. Of these 155 co-occurrences, the most commonly identified ADs were SLE, thyroid disease, GN, rheumatoid arthritis (RA), Crohn’s disease, Sjogren’s syndrome, and celiac disease (Table 1). SLE was by far the most common co-occurring AD, accounting for 30 of the 155 reported occurrences. When the AD occurrences were grouped by MedDRA terms, groups with classified conditions of 5 or more occurrences were Lupus Erythematosus and Associated Conditions (n = 52), Endocrine Autoimmune Disorders (n = 21), Glomerulonephritis and Nephrotic Syndromes (n = 16), Gastrointestinal Inflammatory Conditions (n = 16), Rheumatoid Arthritis and Associated Conditions (n = 11), Eye, Salivary Gland and Connective Tissue Disorders (n = 10), and Immune and Associated Conditions Not Elsewhere Classified (n = 5; Table 2).
Table 1
Number of individual published mentions of HAE-C1INH and autoimmune disease comorbidity
Autoimmune disease
Number of HAE comorbid mentions
 Lupus or lupus-like diseases
52
  Systemic lupus erythematosus
30
  Lupus (unspecified)
7
  Discoid lupus
8
  Lupus-like disease
4
  Cutaneous lupus
2
  Drug-induced lupus
1
 Thyroid disease
21
  Hypothyroidism
10
  Thyroiditis
5
  Anti-thyroid antibodies
3
  Hashimotos
2
  Unspecified
1
 Glomerulonephritis
16
 Rheumatoid arthritis
11
 Crohn’s disease
8
 Various autoantibodies
8
 Sjogren’s syndrome
7
 Celiac disease
7
 Psoriasis
3
 Antiphospholipid syndrome
3
 Autoimmune hemolytic anemia/anemia
3
 General/unspecified autoimmune disease
2
 Psoriatic arthritis
2
 Systemic sclerosis
2
 Mixed connective tissue disease
2
 Raynauds syndrome
2
 Urticarial vasculitis
1
 Sicca syndrome
1
 Lipodystrophy
1
 Alopecia
1
 Multiple-sclerosis-like syndrome
1
 Ulcerative colitis
1
C1-INH C1 esterase inhibitor, HAE hereditary angioedema
Table 2
Number of individual published mentions of HAE-C1INH and AD comorbidity grouped by MedDRA v21.0 high level terms
High level MedDRA term
Number of HAE comorbid AD mentions
 Lupus erythematosus and associated conditions
52
  Systemic lupus erythematosus
30
  Lupus
7
  Discoid lupus
8
  Lupus-like disease
4
  Cutaneous lupus
2
  Drug-induced lupus
1
 Endocrine autoimmune disorders
21
  Thyroid diseases
21
 Glomerulonephritis and nephrotic syndrome
16
  Glomerulonephritis
16
 Gastrointestinal inflammatory conditions
16
  Crohn’s disease
8
  Celiac disease
7
  Ulcerative colitis
1
 Rheumatoid arthritis and associated conditions
11
  Rheumatoid arthritis
11
 Eye, salivary gland and connective tissue disorders
10
  Sjogren’s syndrome
7
  Sicca syndrome
1
  Mixed connective tissue disease
2
 Immune and associated conditions not elsewhere classified
5
  Psoriasis
3
  Psoriatic arthritis
2
Groups with ≥ 5 mentions are shown
AD autoimmune disease, C1-INH C1 esterase inhibitor, HAE hereditary angioedema, MedDRA Medical Dictionary for Regulatory Activities

Discussion

The results of the systematic literature review reveal a demonstrable co-occurrence of ADs in patients with HAE-C1INH, confirmed by clinical and biomarker evidence. Based on data collected in our systematic literature review, the most common co-occurring AD is SLE or lupus-like disease, followed by thyroid disease, GN, gastrointestinal diseases, RA, and Sjogren’s disease.
Although the C1-INH protein inhibits the development of autoimmunity by inhibiting both the classical and lectin complement pathways, the cause and effect between C1-INH deficiency (HAE) and development of ADs has yet to be established, and other etiologies need to be explored further. The lectin pathway has an activation scheme similar to that of the classical complement pathway, but lectins substitute for antibodies, and lectin-associated proteases replace C1r and C1s. The lectins bind sugar residues on microbial surfaces. MASPs subsequently cleave C4 and C2. C1-INH blocks the active sites of these MASPs (Fig. 2).
The alternative complement pathway generates a C3 convertase independent of C4 and C2 (C3bBb). Furthermore, C3 can spontaneously hydrolyze into C3a and C3b [28, 29]. Therefore, active C3b in patients with HAE-C1INH can still be produced to perform normal C3b functions, as evidenced by the low levels of circulating immune complexes in many patients with HAE-C1INH [19].
Reduced C4 levels as observed in SLE may be due to consumption or genetic deficiency of C4 alleles and both causes may be present in a given patient. In SLE, the measurement of C3 and C4 is typically used to assist the diagnosis and is useful for monitoring disease activity. SLE is also the prototypic disease for which the clinical information is available relative to interpreting and following low C3 and C4 levels. Low levels in SLE typically improve with treatment indicating classical pathway activation. Normalization of these complement values is also considered a good prognostic sign [30].
Serum C4 is also typically low in patients with HAE because of chronic activation and depletion, whereas C2 and C3 may be low or normal; C2 levels decline during acute HAE attacks [24, 3133]. In a retrospective observational study, patients with homozygous C4A deficiency were significantly more likely to have autoantibodies, SLE, and celiac disease compared with healthy controls [34]. Notably, mutations in the genes encoding C2, C3, C4A, C4B have been linked to SLE [3537], and Sjogren’s syndrome has been reported in patients with genetic deficiencies in C1q, C4, and C2 [3840]. Furthermore, the odds of having a C4B deficiency are two-fold higher in patients with RA vs non-RA patients [41]. Since subcomponents of C4 and C2 comprise the C3b convertase, which in turn acts upon C2 (Fig. 2), deficiencies of C2, C3, and C4 could result in a lack of C3b. C3b coats immune complexes and binds to complement receptor CR1, which then are opsonized and cleared by phagocytes (Fig. 2) [29, 42]. C3b also aids in the clearance of apoptotic cells by facilitating interaction between the apoptotic cell and phagocytes via binding of complement receptor CR3 [29]. Thus, C1-INH deficiency impacts the classical and lectin pathways and may predispose patients to increases in potentially damaging immune complexes (Fig. 3) [11, 17]. In addition, the defective clearance of apoptotic cells provides a source of autoantigens that can result in production of autoantibodies (Fig. 3) [11, 17]. Autoantibodies and deposition of immune complexes into tissues have been clearly linked to the pathogenesis of several of the ADs identified in this systematic review as most commonly co-occurring with HAE-C1INH, including SLE, GN, thyroid disease, RA, and Sjogren’s syndrome [18, 4346]. Together these biological pathways indicate that the C1-INH protein may play a key role in the complement dysregulation by preventing excessive complement activation on a target, as well as in plasma (classical and lectin pathways). The decreased function of C1-INH with subsequent dysregulation of the classical and lectin complement pathways may lead to increased co-occurrence of AD in patients with HAE-C1INH. Certainly other factors other than complement dysregulation contribute to AD development. Genetic susceptibilities and triggers such as viral infection are also thought to be required to induce SLE, autoimmune thyroid disease, RA, and Sjogren’s syndrome [4749].
Patients with SLE and other autoimmune conditions could develop acquired angioedema which can present with the same symptoms as HAE Type I or II along with a low C4 and a low C1 inhibitor function [5052]. There would be no family history and the patients present with the angioedema as the first manifestation of their autoimmune condition. The only way to differentiate the problem is to do a C1q level, which may be low, or genetic testing. There is one case in the literature of a patient who had HAE Type I but developed features of acquired angioedema after she developed lymphoma [53]. Her C1q, which had been normal, fell and only recovered once her lymphoma was treated.
Prophylactic administration of C1-INH in HAE patients results in normalization or near-normalization of C4 and C1-INH antigen and functional levels, as evidenced in clinical trials and pharmacokinetic/pharmacodynamic modeling [5456]. In a phase III clinical trial, the use of subcutaneous C1-INH resulted in sustained steady-state levels of functional C1-INH [54, 55]. It is unknown if the restoration of C4 levels in HAE patients treated with C1-INH prophylaxis affects any of the complement components or if there could be any additional benefits due to the potential restoration of proper complement regulation.
This systematic review is limited to search terms contained in the article’s title, abstract, and key words that mentioned the co-occurring HAE-C1INH and ADs, as described in the methods section. The prevalence of co-occurring HAE-C1INH and ADs cannot be inferred from this systematic review since the results only provide a report of the number of published mentions.

Conclusions

Although there is a compelling association for the cause and effect of the co-occurrence of HAE-C1INH and AD due to persistently low C1-INH functional levels, more research is needed in HAE patients to confirm this theory. Further clinical observation and research is needed to see if subcutaneous C1-INH replacement therapy can possibly ameliorate or prevent AD in HAE patients.

Acknowledgements

Medical writing and editorial support were provided by Erin P. Scott, PhD, of Scott Medical Communications, LLC. This support was funded by CSL Behring, King of Prussia, PA.
Not applicable.
Not applicable.

Competing interests

DL is a consultant, investigator, and/or speaker for Biocryst, CSL Behring, and Shire/Takeda. PKK is a consultant, investigator, and/or speaker for CSL Behring and Shire/Takeda, and serves on the Board of Directors for CHAEN (Canadian Hereditary Angioedema Network). TC is a consultant, investigator, and/or speaker for Biocryst, CSL Behring, Grifols, and Shire, and serves on the Board of Directors for the AAAAI and Mid-Atlantic ALA and on the Board of the Medical Advisors for the HAE-A of America. GK was an employee of CSL Behring at the time of the study. RB is an employee of Maple Health Group LLC, which provides contracted services to CSL Behring. SP is an employee of CSL Behring.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Davis AE 3rd, Lu F, Mejia P. C1 inhibitor, a multi-functional serine protease inhibitor. Thromb Haemost. 2010;104(5):886–93.PubMed Davis AE 3rd, Lu F, Mejia P. C1 inhibitor, a multi-functional serine protease inhibitor. Thromb Haemost. 2010;104(5):886–93.PubMed
2.
Zurück zum Zitat Steiner UC, Keller M, Schmid P, Cichon S, Wuillemin WA. Mutational spectrum of the SERPING1 gene in Swiss patients with hereditary angioedema. Clin Exp Immunol. 2017;188(3):430–6.PubMedPubMedCentralCrossRef Steiner UC, Keller M, Schmid P, Cichon S, Wuillemin WA. Mutational spectrum of the SERPING1 gene in Swiss patients with hereditary angioedema. Clin Exp Immunol. 2017;188(3):430–6.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Johnsrud I, Kulseth MA, Rodningen OK, Landro L, Helsing P, Waage Nielsen E, et al. A nationwide study of Norwegian patients with hereditary angioedema with C1 inhibitor deficiency identified six novel mutations in SERPING1. PLoS ONE. 2015;10(7):e0131637.PubMedPubMedCentralCrossRef Johnsrud I, Kulseth MA, Rodningen OK, Landro L, Helsing P, Waage Nielsen E, et al. A nationwide study of Norwegian patients with hereditary angioedema with C1 inhibitor deficiency identified six novel mutations in SERPING1. PLoS ONE. 2015;10(7):e0131637.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Agostoni A, Aygoren-Pursun E, Binkley KE, Blanch A, Bork K, Bouillet L, 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):S51–131.PubMedPubMedCentralCrossRef Agostoni A, Aygoren-Pursun E, Binkley KE, Blanch A, Bork K, Bouillet L, 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):S51–131.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Nussberger J, Cugno M, Cicardi M. Bradykinin-mediated angioedema. N Engl J Med. 2002;347(8):621–2.PubMedCrossRef Nussberger J, Cugno M, Cicardi M. Bradykinin-mediated angioedema. N Engl J Med. 2002;347(8):621–2.PubMedCrossRef
6.
Zurück zum Zitat Betschel S, Badiou J, Binkley K, Hebert J, Kanani A, Keith P, et al. Canadian hereditary angioedema guideline. Allergy Asthma Clin Immunol. 2014;10(1):50.PubMedPubMedCentralCrossRef Betschel S, Badiou J, Binkley K, Hebert J, Kanani A, Keith P, et al. Canadian hereditary angioedema guideline. Allergy Asthma Clin Immunol. 2014;10(1):50.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Craig T, Aygoren-Pursun E, Bork K, Bowen T, Boysen H, Farkas H, et al. WAO guideline for the management of hereditary angioedema. World Allergy Organ J. 2012;5(12):182–99.PubMedPubMedCentralCrossRef Craig T, Aygoren-Pursun E, Bork K, Bowen T, Boysen H, Farkas H, et al. WAO guideline for the management of hereditary angioedema. World Allergy Organ J. 2012;5(12):182–99.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Longhurst HJ, Tarzi MD, Ashworth F, Bethune C, Cale C, Dempster J, et al. C1 inhibitor deficiency: 2014 United Kingdom consensus document. Clin Exp Immunol. 2015;180(3):475–83.PubMedPubMedCentralCrossRef Longhurst HJ, Tarzi MD, Ashworth F, Bethune C, Cale C, Dempster J, et al. C1 inhibitor deficiency: 2014 United Kingdom consensus document. Clin Exp Immunol. 2015;180(3):475–83.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Maurer M, Magerl M, Ansotegui I, Aygoren-Pursun E, Betschel S, Bork K, et al. The international WAO/EAACI guideline for the management of hereditary angioedema-The 2017 revision and update. Allergy. 2018;73(8):1575–96.PubMedCrossRef Maurer M, Magerl M, Ansotegui I, Aygoren-Pursun E, Betschel S, Bork K, et al. The international WAO/EAACI guideline for the management of hereditary angioedema-The 2017 revision and update. Allergy. 2018;73(8):1575–96.PubMedCrossRef
10.
Zurück zum Zitat Takhzyro™ (lanadelumab-flyo). Full prescribing information. Lexington, MA: Shire; 2018. Takhzyro™ (lanadelumab-flyo). Full prescribing information. Lexington, MA: Shire; 2018.
11.
Zurück zum Zitat Lintner KE, Wu YL, Yang Y, Spencer CH, Hauptmann G, Hebert LA, et al. Early components of the complement classical activation pathway in human systemic autoimmune diseases. Front Immunol. 2016;7:36.PubMedPubMedCentralCrossRef Lintner KE, Wu YL, Yang Y, Spencer CH, Hauptmann G, Hebert LA, et al. Early components of the complement classical activation pathway in human systemic autoimmune diseases. Front Immunol. 2016;7:36.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Jiang H, Wagner E, Zhang H, Frank MM. Complement 1 inhibitor is a regulator of the alternative complement pathway. J Exp Med. 2001;194(11):1609–16.PubMedPubMedCentralCrossRef Jiang H, Wagner E, Zhang H, Frank MM. Complement 1 inhibitor is a regulator of the alternative complement pathway. J Exp Med. 2001;194(11):1609–16.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Kaplan AP. Enzymatic pathways in the pathogenesis of hereditary angioedema: the role of C1 inhibitor therapy. J Allergy Clin Immunol. 2010;126(5):918–25.PubMedCrossRef Kaplan AP. Enzymatic pathways in the pathogenesis of hereditary angioedema: the role of C1 inhibitor therapy. J Allergy Clin Immunol. 2010;126(5):918–25.PubMedCrossRef
14.
Zurück zum Zitat Triggianese P, Guarino MD, Ballanti E, Chimenti MS, Perricone R. Hereditary angioedema and autoimmunity. Isr Med Assoc J. 2014;16(10):622–4.PubMed Triggianese P, Guarino MD, Ballanti E, Chimenti MS, Perricone R. Hereditary angioedema and autoimmunity. Isr Med Assoc J. 2014;16(10):622–4.PubMed
15.
Zurück zum Zitat Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a key system for immune surveillance and homeostasis. Nat Immunol. 2010;11(9):785–97.PubMedPubMedCentralCrossRef Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a key system for immune surveillance and homeostasis. Nat Immunol. 2010;11(9):785–97.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Fischer MB, Ma M, Goerg S, Zhou X, Xia J, Finco O, et al. Regulation of the B cell response to T-dependent antigens by classical pathway complement. J Immunol. 1996;157(2):549–56.PubMed Fischer MB, Ma M, Goerg S, Zhou X, Xia J, Finco O, et al. Regulation of the B cell response to T-dependent antigens by classical pathway complement. J Immunol. 1996;157(2):549–56.PubMed
17.
Zurück zum Zitat Truedsson L, Bengtsson AA, Sturfelt G. Complement deficiencies and systemic lupus erythematosus. Autoimmunity. 2007;40(8):560–6.PubMedCrossRef Truedsson L, Bengtsson AA, Sturfelt G. Complement deficiencies and systemic lupus erythematosus. Autoimmunity. 2007;40(8):560–6.PubMedCrossRef
18.
Zurück zum Zitat Tsokos GC, Lo MS, Costa Reis P, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol. 2016;12(12):716–30.CrossRefPubMed Tsokos GC, Lo MS, Costa Reis P, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol. 2016;12(12):716–30.CrossRefPubMed
19.
Zurück zum Zitat D’Amelio R, Perricone R, De Carolis C, Pontesilli O, Matricardi PM, Fontana L. Immune complexes in hereditary angioneurotic edema (HANE). J Allergy Clin Immunol. 1986;78(3 Pt 1):486–7.PubMedCrossRef D’Amelio R, Perricone R, De Carolis C, Pontesilli O, Matricardi PM, Fontana L. Immune complexes in hereditary angioneurotic edema (HANE). J Allergy Clin Immunol. 1986;78(3 Pt 1):486–7.PubMedCrossRef
20.
Zurück zum Zitat Donaldson VH, Hess EV, McAdams AJ. Lupus-erythematosus-like disease in three unrelated women with hereditary angioneurotic edema. Ann Intern Med. 1977;86(3):312–3.PubMedCrossRef Donaldson VH, Hess EV, McAdams AJ. Lupus-erythematosus-like disease in three unrelated women with hereditary angioneurotic edema. Ann Intern Med. 1977;86(3):312–3.PubMedCrossRef
21.
Zurück zum Zitat Gallais Sérézal I, Bouillet L, Dhôte R, Gayet S, Jeandel PY, Blanchard-Delaunay C, et al. Hereditary angioedema and lupus: a French retrospective study and literature review. Autoimmun Rev. 2015;14(6):564–8.PubMedCrossRef Gallais Sérézal I, Bouillet L, Dhôte R, Gayet S, Jeandel PY, Blanchard-Delaunay C, et al. Hereditary angioedema and lupus: a French retrospective study and literature review. Autoimmun Rev. 2015;14(6):564–8.PubMedCrossRef
22.
Zurück zum Zitat Hory B, Haultier JJ. Glomerulonephritis and hereditary angioedema: report of 2 cases. Clin Nephrol. 1989;31(5):259–63.PubMed Hory B, Haultier JJ. Glomerulonephritis and hereditary angioedema: report of 2 cases. Clin Nephrol. 1989;31(5):259–63.PubMed
23.
Zurück zum Zitat Agostoni A, Cicardi M. Hereditary and acquired C1-inhibitor deficiency: biological and clinical characteristics in 235 patients. Medicine. 1992;71(4):206–15.PubMedCrossRef Agostoni A, Cicardi M. Hereditary and acquired C1-inhibitor deficiency: biological and clinical characteristics in 235 patients. Medicine. 1992;71(4):206–15.PubMedCrossRef
24.
Zurück zum Zitat Farkas H, Csuka D, Gacs J, Czaller I, Zotter Z, Fust G, et al. Lack of increased prevalence of immunoregulatory disorders in hereditary angioedema due to C1-inhibitor deficiency. Clin Immunol. 2011;141(1):58–66.PubMedCrossRef Farkas H, Csuka D, Gacs J, Czaller I, Zotter Z, Fust G, et al. Lack of increased prevalence of immunoregulatory disorders in hereditary angioedema due to C1-inhibitor deficiency. Clin Immunol. 2011;141(1):58–66.PubMedCrossRef
25.
Zurück zum Zitat Muhlemann MF, Macrae KD, Smith AM, Beck P, Hine I, Hegde U, et al. Hereditary angioedema and thyroid autoimmunity. J Clin Pathol. 1987;40(5):518–23.PubMedPubMedCentralCrossRef Muhlemann MF, Macrae KD, Smith AM, Beck P, Hine I, Hegde U, et al. Hereditary angioedema and thyroid autoimmunity. J Clin Pathol. 1987;40(5):518–23.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Brickman CM, Tsokos GC, Balow JE, Lawley TJ, Santaella M, Hammer CH, et al. Immunoregulatory disorders associated with hereditary angioedema. I. Clinical manifestations of autoimmune disease. J Allergy Clin Immunol. 1986;77(5):749–57.PubMedCrossRef Brickman CM, Tsokos GC, Balow JE, Lawley TJ, Santaella M, Hammer CH, et al. Immunoregulatory disorders associated with hereditary angioedema. I. Clinical manifestations of autoimmune disease. J Allergy Clin Immunol. 1986;77(5):749–57.PubMedCrossRef
27.
Zurück zum Zitat Hayter SM, Cook MC. Updated assessment of the prevalence, spectrum and case definition of autoimmune disease. Autoimmun Rev. 2012;11(10):754–65.PubMedCrossRef Hayter SM, Cook MC. Updated assessment of the prevalence, spectrum and case definition of autoimmune disease. Autoimmun Rev. 2012;11(10):754–65.PubMedCrossRef
28.
Zurück zum Zitat Ricklin D. Manipulating the mediator: modulation of the alternative complement pathway C3 convertase in health, disease and therapy. Immunobiology. 2012;217(11):1057–66.PubMedPubMedCentralCrossRef Ricklin D. Manipulating the mediator: modulation of the alternative complement pathway C3 convertase in health, disease and therapy. Immunobiology. 2012;217(11):1057–66.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Merle NS, Noe R, Halbwachs-Mecarelli L, Fremeaux-Bacchi V, Roumenina LT. Complement system part II: role in immunity. Front Immunol. 2015;6:257.PubMedPubMedCentral Merle NS, Noe R, Halbwachs-Mecarelli L, Fremeaux-Bacchi V, Roumenina LT. Complement system part II: role in immunity. Front Immunol. 2015;6:257.PubMedPubMedCentral
31.
Zurück zum Zitat Donaldson VH, Bissler JJ, Welch TR, Burton MF, Davis AE 3rd. Antibody to C1-inhibitor in a patient receiving C1-inhibitor infusions for treatment of hereditary angioneurotic edema with systemic lupus erythematosus reacts with a normal allotype of residue 458 of C1-inhibitor. J Lab Clin Med. 1996;128(4):438–43.PubMedCrossRef Donaldson VH, Bissler JJ, Welch TR, Burton MF, Davis AE 3rd. Antibody to C1-inhibitor in a patient receiving C1-inhibitor infusions for treatment of hereditary angioneurotic edema with systemic lupus erythematosus reacts with a normal allotype of residue 458 of C1-inhibitor. J Lab Clin Med. 1996;128(4):438–43.PubMedCrossRef
32.
Zurück zum Zitat Leru P, Zamfirescu M, Baicus C. Autoimmunity associated with a clear family history of hereditary angioedema. Allergy. 2009;64:254–5.CrossRef Leru P, Zamfirescu M, Baicus C. Autoimmunity associated with a clear family history of hereditary angioedema. Allergy. 2009;64:254–5.CrossRef
33.
Zurück zum Zitat Khan S, Tarzi MD, Dore PC, Sewell WA, Longhurst HJ. Secondary systemic lupus erythematosus: an analysis of 4 cases of uncontrolled hereditary angioedema. Clin Immunol. 2007;123(1):14–7.PubMedCrossRef Khan S, Tarzi MD, Dore PC, Sewell WA, Longhurst HJ. Secondary systemic lupus erythematosus: an analysis of 4 cases of uncontrolled hereditary angioedema. Clin Immunol. 2007;123(1):14–7.PubMedCrossRef
34.
Zurück zum Zitat Liesmaa I, Paakkanen R, Jarvinen A, Valtonen V, Lokki ML. Clinical features of patients with homozygous complement C4A or C4B deficiency. PLoS ONE. 2018;13(6):e0199305.PubMedPubMedCentralCrossRef Liesmaa I, Paakkanen R, Jarvinen A, Valtonen V, Lokki ML. Clinical features of patients with homozygous complement C4A or C4B deficiency. PLoS ONE. 2018;13(6):e0199305.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Tsukamoto H, Horiuchi T, Kokuba H, Nagae S, Nishizaka H, Sawabe T, et al. Molecular analysis of a novel hereditary C3 deficiency with systemic lupus erythematosus. Biochem Biophys Res Commun. 2005;330(1):298–304.PubMedCrossRef Tsukamoto H, Horiuchi T, Kokuba H, Nagae S, Nishizaka H, Sawabe T, et al. Molecular analysis of a novel hereditary C3 deficiency with systemic lupus erythematosus. Biochem Biophys Res Commun. 2005;330(1):298–304.PubMedCrossRef
36.
Zurück zum Zitat Yang Y, Lhotta K, Chung EK, Eder P, Neumair F, Yu CY. Complete complement components C4A and C4B deficiencies in human kidney diseases and systemic lupus erythematosus. J Immunol. 2004;173(4):2803–14.PubMedCrossRef Yang Y, Lhotta K, Chung EK, Eder P, Neumair F, Yu CY. Complete complement components C4A and C4B deficiencies in human kidney diseases and systemic lupus erythematosus. J Immunol. 2004;173(4):2803–14.PubMedCrossRef
37.
Zurück zum Zitat Sullivan KE, Petri MA, Schmeckpeper BJ, McLean RH, Winkelstein JA. Prevalence of a mutation causing C2 deficiency in systemic lupus erythematosus. J Rheumatol. 1994;21(6):1128–33.PubMed Sullivan KE, Petri MA, Schmeckpeper BJ, McLean RH, Winkelstein JA. Prevalence of a mutation causing C2 deficiency in systemic lupus erythematosus. J Rheumatol. 1994;21(6):1128–33.PubMed
38.
Zurück zum Zitat Holtman JH, Neustadt DH, Klein J, Callen JP. Dapsone is an effective therapy for the skin lesions of subacute cutaneous lupus erythematosus and urticarial vasculitis in a patient with C2 deficiency. J Rheumatol. 1990;17(9):1222–5.PubMed Holtman JH, Neustadt DH, Klein J, Callen JP. Dapsone is an effective therapy for the skin lesions of subacute cutaneous lupus erythematosus and urticarial vasculitis in a patient with C2 deficiency. J Rheumatol. 1990;17(9):1222–5.PubMed
39.
Zurück zum Zitat Hersey P, Lawrence S, Prendergast D, Bindon C, Benson W, Valk P. Association of Sjogren’s syndrome with C4 deficiency, defective reticuloendothelial function and circulating immune complexes. Clin Exp Immunol. 1983;52(3):551–60.PubMedPubMedCentral Hersey P, Lawrence S, Prendergast D, Bindon C, Benson W, Valk P. Association of Sjogren’s syndrome with C4 deficiency, defective reticuloendothelial function and circulating immune complexes. Clin Exp Immunol. 1983;52(3):551–60.PubMedPubMedCentral
40.
Zurück zum Zitat Hoppenreijs EP, van Dijken PJ, Kabel PJ, Th Draaisma JM. Hereditary C1q deficiency and secondary Sjogren’s syndrome. Ann Rheum Dis. 2004;63(11):1524–5.PubMedPubMedCentralCrossRef Hoppenreijs EP, van Dijken PJ, Kabel PJ, Th Draaisma JM. Hereditary C1q deficiency and secondary Sjogren’s syndrome. Ann Rheum Dis. 2004;63(11):1524–5.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Rigby WF, Wu YL, Zan M, Zhou B, Rosengren S, Carlson C, et al. Increased frequency of complement C4B deficiency in rheumatoid arthritis. Arthritis Rheum. 2012;64(5):1338–44.PubMedPubMedCentralCrossRef Rigby WF, Wu YL, Zan M, Zhou B, Rosengren S, Carlson C, et al. Increased frequency of complement C4B deficiency in rheumatoid arthritis. Arthritis Rheum. 2012;64(5):1338–44.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Schifferli JA, Taylor RP. Physiological and pathological aspects of circulating immune complexes. Kidney Int. 1989;35(4):993–1003.PubMedCrossRef Schifferli JA, Taylor RP. Physiological and pathological aspects of circulating immune complexes. Kidney Int. 1989;35(4):993–1003.PubMedCrossRef
43.
Zurück zum Zitat Garcia-Carrasco M, Pinto CM, Poblano JCS, Morales IE, Cervera R, Anaya JM. Systemic lupus erthematosus. In: Anaya JM, Shoenfeld Y, Rojas-Villarraga A, editors. Autoimmunity: from bench to bedside. Bogota: El Rosario University Press; 2013. Garcia-Carrasco M, Pinto CM, Poblano JCS, Morales IE, Cervera R, Anaya JM. Systemic lupus erthematosus. In: Anaya JM, Shoenfeld Y, Rojas-Villarraga A, editors. Autoimmunity: from bench to bedside. Bogota: El Rosario University Press; 2013.
44.
Zurück zum Zitat Mincer DL, Jialal I. Hashimoto thyroiditis. In: StatPearls. Treasure Island, FL: StatPearls Publishing LLC; 2018. Mincer DL, Jialal I. Hashimoto thyroiditis. In: StatPearls. Treasure Island, FL: StatPearls Publishing LLC; 2018.
46.
Zurück zum Zitat Derksen V, Huizinga TWJ, van der Woude D. The role of autoantibodies in the pathophysiology of rheumatoid arthritis. Semin Immunopathol. 2017;39(4):437–46.PubMedPubMedCentralCrossRef Derksen V, Huizinga TWJ, van der Woude D. The role of autoantibodies in the pathophysiology of rheumatoid arthritis. Semin Immunopathol. 2017;39(4):437–46.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Hansen A, Lipsky PE, Dorner T. Immunopathogenesis of primary Sjogren’s syndrome: implications for disease management and therapy. Curr Opin Rheumatol. 2005;17(5):558–65.PubMedCrossRef Hansen A, Lipsky PE, Dorner T. Immunopathogenesis of primary Sjogren’s syndrome: implications for disease management and therapy. Curr Opin Rheumatol. 2005;17(5):558–65.PubMedCrossRef
48.
Zurück zum Zitat James JA, Harley JB, Scofield RH. Role of viruses in systemic lupus erythematosus and Sjogren syndrome. Curr Opin Rheumatol. 2001;13(5):370–6.PubMedCrossRef James JA, Harley JB, Scofield RH. Role of viruses in systemic lupus erythematosus and Sjogren syndrome. Curr Opin Rheumatol. 2001;13(5):370–6.PubMedCrossRef
49.
Zurück zum Zitat Effraimidis G, Wiersinga WM. Mechanisms in endocrinology: autoimmune thyroid disease: old and new players. Eur J Endocrinol. 2014;170(6):R241–52.PubMedCrossRef Effraimidis G, Wiersinga WM. Mechanisms in endocrinology: autoimmune thyroid disease: old and new players. Eur J Endocrinol. 2014;170(6):R241–52.PubMedCrossRef
50.
51.
Zurück zum Zitat Markovic SN, Inwards DJ, Frigas EA, Phyliky RP. Acquired C1 esterase inhibitor deficiency. Ann Intern Med. 2000;132(2):144–50.PubMedCrossRef Markovic SN, Inwards DJ, Frigas EA, Phyliky RP. Acquired C1 esterase inhibitor deficiency. Ann Intern Med. 2000;132(2):144–50.PubMedCrossRef
52.
Zurück zum Zitat Tekin ZE, Yener GO, Yuksel S. Acquired angioedema in juvenile systemic lupus erythematosus: case-based review. Rheumatol Int. 2018;38(8):1577–84.PubMedCrossRef Tekin ZE, Yener GO, Yuksel S. Acquired angioedema in juvenile systemic lupus erythematosus: case-based review. Rheumatol Int. 2018;38(8):1577–84.PubMedCrossRef
53.
Zurück zum Zitat Tran JP, McCracken JL, Morsy A, Gonzalez EB. Systemic lupus erythematous presenting as acquired angioedema: a case report and review of the literature. J Autoimmune Syst Dis. 2017;1(1):1000002. Tran JP, McCracken JL, Morsy A, Gonzalez EB. Systemic lupus erythematous presenting as acquired angioedema: a case report and review of the literature. J Autoimmune Syst Dis. 2017;1(1):1000002.
54.
Zurück zum Zitat Guilarte M, Luengo O, Nogueiras C, Labrador-Horrillo M, Munoz E, Lopez A, et al. Acquired angioedema associated with hereditary angioedema due to C1 inhibitor deficiency. J Investig Allergol Clin Immunol. 2008;18(2):126–30.PubMed Guilarte M, Luengo O, Nogueiras C, Labrador-Horrillo M, Munoz E, Lopez A, et al. Acquired angioedema associated with hereditary angioedema due to C1 inhibitor deficiency. J Investig Allergol Clin Immunol. 2008;18(2):126–30.PubMed
55.
Zurück zum Zitat Longhurst H, Cicardi M, Craig T, Bork K, Grattan C, Baker J, et al. Prevention of Hereditary Angioedema Attacks with a Subcutaneous C1 Inhibitor. N Engl J Med. 2017;376(12):1131–40.PubMedCrossRef Longhurst H, Cicardi M, Craig T, Bork K, Grattan C, Baker J, et al. Prevention of Hereditary Angioedema Attacks with a Subcutaneous C1 Inhibitor. N Engl J Med. 2017;376(12):1131–40.PubMedCrossRef
56.
Zurück zum Zitat Craig T, Zuraw B, Longhurst H, Cicardi M, Bork K, Grattan C, et al. Long-term outcomes with subcutaneous C1-inhibitor replacement therapy for prevention of hereditary angioedema attacks. J Allergy Clin Immunol Pract. 2019;7(6):1793–802.PubMedCrossRef Craig T, Zuraw B, Longhurst H, Cicardi M, Bork K, Grattan C, et al. Long-term outcomes with subcutaneous C1-inhibitor replacement therapy for prevention of hereditary angioedema attacks. J Allergy Clin Immunol Pract. 2019;7(6):1793–802.PubMedCrossRef
57.
Zurück zum Zitat Pawaskar D, Tortorici MA, Zuraw B, Craig T, Cicardi M, Longhurst H, et al. Population pharmacokinetics of subcutaneous C1-inhibitor for prevention of attacks in patients with hereditary angioedema. Clin Exp Allergy. 2018;48(10):1325–32.CrossRefPubMed Pawaskar D, Tortorici MA, Zuraw B, Craig T, Cicardi M, Longhurst H, et al. Population pharmacokinetics of subcutaneous C1-inhibitor for prevention of attacks in patients with hereditary angioedema. Clin Exp Allergy. 2018;48(10):1325–32.CrossRefPubMed
Metadaten
Titel
Co-occurrence between C1 esterase inhibitor deficiency and autoimmune disease: a systematic literature review
verfasst von
Donald Levy
Timothy Craig
Paul K. Keith
Girishanthy Krishnarajah
Rachel Beckerman
Subhransu Prusty
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Allergy, Asthma & Clinical Immunology / Ausgabe 1/2020
Elektronische ISSN: 1710-1492
DOI
https://doi.org/10.1186/s13223-020-00437-x

Weitere Artikel der Ausgabe 1/2020

Allergy, Asthma & Clinical Immunology 1/2020 Zur Ausgabe

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Nur selten Nachblutungen nach Abszesstonsillektomie

03.05.2024 Tonsillektomie Nachrichten

In einer Metaanalyse von 18 Studien war die Rate von Nachblutungen nach einer Abszesstonsillektomie mit weniger als 7% recht niedrig. Nur rund 2% der Behandelten mussten nachoperiert werden. Die Therapie scheint damit recht sicher zu sein.

Rezidivierender Peritonsillarabszess nach Oralsex

02.05.2024 Peritonsillarabszess Kasuistik

Die erotischen Dimensionen von Peritonsillarabszessen scheinen eng begrenzt zu sein. Das heißt aber nicht, solche Abszesse und Erotik hätten nichts miteinander gemein, wie ein Fallbericht verdeutlicht.

Update HNO

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.