Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 311
Filter
1.
J Invest Dermatol ; 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39320300

ABSTRACT

Laminin ß4 was recently identified as a structural component of the dermal-epidermal junction and autoantigen of anti-p200 pemphigoid. In this study, we provided further evidence of the pathogenic effect of anti-laminin ß4 IgG and identified potential binding partners of laminin ß4. We showed that laminin ß4 immune complexes led to activation of normal leukocytes and dose-dependent ROS release. Using cryosections of normal skin, we demonstrated that anti-laminin ß4 patient serum IgG but not anti-laminin γ1 IgG, which are also detectable in patients with anti-p200 pemphigoid, cause dermal-epidermal separation in the presence of leukocytes. Proximity ligation assay and indirect immunofluorescence staining suggested that laminin ß4 localizes closely to laminin α3 and γ2 in primary keratinocytes. Subsequent coimmunoprecipitation experiments using epidermal extracts confirmed the interaction of laminin ß4 with the α3 and γ2 chains and indicated additional affinity to laminin γ1. The laminin ß4-α3/ß4-γ1 protein complexes were also detected using mass spectrometry. In conclusion, this study showed that anti-laminin ß4 IgG can exert tissue damage in the skin, supporting their pathogenic role in anti-p200 pemphigoid. Our data further provide strong evidence for an interaction of laminin ß4 with laminin α3, whereas its association to the laminin γ1 and γ2 chains is ambiguous.

2.
Immunity ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39226901

ABSTRACT

Pro-inflammatory autoantigen-specific CD4+ T helper (auto-Th) cells are central orchestrators of autoimmune diseases (AIDs). We aimed to characterize these cells in human AIDs with defined autoantigens by combining human leukocyte antigen (HLA)-tetramer-based and activation-based multidimensional ex vivo analyses. In aquaporin4-antibody-positive neuromyelitis optica spectrum disorder (AQP4-NMOSD) patients, auto-Th cells expressed CD154, but proliferative capacity and pro-inflammatory cytokines were strongly reduced. Instead, exhaustion-associated co-inhibitory receptors were expressed together with FOXP3, the canonical regulatory T cell (Treg) transcription factor. Auto-Th cells responded in vitro to checkpoint inhibition and provided potent B cell help. Cells with the same exhaustion-like (ThEx) phenotype were identified in soluble liver antigen (SLA)-antibody-autoimmune hepatitis and BP180-antibody-positive bullous pemphigoid, AIDs of the liver and skin, respectively. While originally described in cancer and chronic infection, our data point to T cell exhaustion as a common mechanism of adaptation to chronic (self-)stimulation across AID types and link exhausted CD4+ T cells to humoral autoimmune responses, with implications for therapeutic targeting.

3.
Exp Dermatol ; 33(9): e15172, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39219105

ABSTRACT

The pemphigoid disease epidermolysis bullosa acquisita (EBA) is an autoimmune blistering skin disease characterized by autoantibodies against type VII collagen (COL7), immune cell infiltrates at the dermal-epidermal junction and subepidermal blistering. Proteases, particularly granzyme B (GzmB), have been established as therapeutic targets for the treatment of EBA and other pemphigoid diseases. We investigated the impact of the novel GzmB inhibitor SNT-6935 on anti-COL7 IgG-induced subepidermal blistering in a well-established EBA ex vivo model. Our findings demonstrate that pharmacological targeting of GzmB with its selective inhibitor SNT-6935 significantly reduced autoantibody-induced dermal-epidermal separation in human skin cryosections. Interestingly, treatment of skin cryosections with recombinant human GzmB alone did not cause dermal-epidermal separation, suggesting that additional mechanisms alongside GzmB are required for tissue damage in EBA. In conclusion, our study highlights the significant contribution of GzmB to the pathogenesis of EBA and supports the notion of GzmB as a therapeutic target in EBA and other pemphigoid diseases.


Subject(s)
Autoantibodies , Collagen Type VII , Epidermis , Epidermolysis Bullosa Acquisita , Granzymes , Epidermolysis Bullosa Acquisita/drug therapy , Epidermolysis Bullosa Acquisita/immunology , Humans , Granzymes/metabolism , Granzymes/antagonists & inhibitors , Collagen Type VII/immunology , Epidermis/pathology , Dermis/pathology , Skin/pathology
4.
Nat Commun ; 15(1): 8044, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39271654

ABSTRACT

Cell-cell junctions, and specifically desmosomes, are crucial for robust intercellular adhesion. Desmosomal function is compromised in the autoimmune blistering skin disease pemphigus vulgaris. We combine whole-genome knockout screening and a promotor screen of the desmosomal gene desmoglein 3 in human keratinocytes to identify novel regulators of intercellular adhesion. Kruppel-like-factor 5 (KLF5) directly binds to the desmoglein 3 regulatory region and promotes adhesion. Reduced levels of KLF5 in patient tissue indicate a role in pemphigus vulgaris. Autoantibody fractions from patients impair intercellular adhesion and reduce KLF5 levels in in vitro and in vivo disease models. These effects were dependent on increased activity of histone deacetylase 3, leading to transcriptional repression of KLF5. Inhibiting histone deacetylase 3 increases KLF5 levels and protects against the deleterious effects of autoantibodies in murine and human pemphigus vulgaris models. Together, KLF5 and histone deacetylase 3 are regulators of desmoglein 3 gene expression and intercellular adhesion and represent potential therapeutic targets in pemphigus vulgaris.


Subject(s)
Cell Adhesion , Desmoglein 3 , Keratinocytes , Kruppel-Like Transcription Factors , Pemphigus , Humans , Pemphigus/metabolism , Pemphigus/pathology , Pemphigus/immunology , Desmoglein 3/metabolism , Desmoglein 3/genetics , Animals , Keratinocytes/metabolism , Mice , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Autoantibodies/immunology , Desmosomes/metabolism , Disease Models, Animal , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Gene Expression Regulation , Promoter Regions, Genetic/genetics , Male
5.
Ocul Immunol Inflamm ; : 1-14, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39241171

ABSTRACT

Ocular predominant mucous membrane pemphigoid (oMMP) is a severe subtype of MMP that can lead to scarring and blindness. While conjunctival biopsy for direct immunofluorescence (DIF) is considered the gold standard for diagnosis, limited sensitivity results in a false-negative rate upwards of 40%. Likewise, it remains unclear to what extent a negative biopsy, whether false-negative or true-negative, results in a different prognosis, with patients previously termed "pseudopemphigoid" demonstrating comparable disease progression. Serologic testing allows for a less invasive means to demonstrate circulating autoantibodies against known autoantigens in pemphigoid diseases. Patients with MMP, particularly oMMP, however, typically demonstrate low titers of circulating autoantibodies, limiting the diagnostic utility of these tests. The autoantigen integrin ß4 has been previously reported to be a specific marker of pure ocular MMP, while in the majority of patients with oMMP, the identified target antigens are BP180 (type XVII collagen) and laminin 332. Recent studies have, however, demonstrated inconsistent reactivity and specificity for integrin ß4 as an ocular-specific marker in MMP. Herein, we review the role of serologic testing in the diagnosis and prognosis of oMMP, as well as the current understanding of autoantigens in oMMP.Abbreviations: BMZ - basement membrane zone, DIF - direct immunofluorescence, IIF - indirect immunofluorescence, MMP - mucous membrane pemphigoid, oMMP - ocular predominant mucous membrane pemphigoid.

6.
Exp Dermatol ; 33(7): e15125, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38946225

ABSTRACT

The 16th non-collagenous domain (NC16A) of BP180 is the main antigenic target of autoantibodies in bullous pemphigoid (BP) and mucous membrane pemphigoid (MMP). Commercially available assays detect serum autoantibodies against NC16A in the majority of BP (80%-90%) and in approximately 50% of MMP patients. However, a standardized test system for detecting antibodies against other regions of BP180 is still lacking. Moreover, anti-BP180 autoantibodies have been found in neurological conditions such as multiple sclerosis and Parkinson disease. This study aimed at identifying primary epitopes recognized by BP autoantibodies on the BP180 ectodomain. Serum samples of 51 BP and 30 MMP patients both without anti-NC16A reactivity were included along with 44 multiple sclerosis and 75 Parkinson disease sera. Four overlapping His-tagged proteins covering the entire BP180 ectodomain (BP180(ec)1-4) were cloned, expressed, purified and tested for reactivity by immunoblot. IgG antibodies to BP180(ec)3 were detected in 98% of BP, 77% of MMP and 2% of normal human sera. Only weak reactivity was detected for neurological diseases against BP180(ec)1, BP180(ec)2 and BP180(ec)4, in 3%, 11% and 7% of tested multiple sclerosis sera, respectively. 8% of Parkinson disease sera reacted with BP180(ec)2 and 9% with BP180(ec)4. In conclusion, this study successfully identified epitopes recognized by BP autoantibodies outside the NC16A domain in pemphigoid diseases. These findings contribute to a better understanding of the immune response in BP and MMP with potential implications for a future diagnostic assay for NC16A-negative pemphigoid patients.


Subject(s)
Autoantibodies , Autoantigens , Collagen Type XVII , Multiple Sclerosis , Non-Fibrillar Collagens , Parkinson Disease , Pemphigoid, Benign Mucous Membrane , Pemphigoid, Bullous , Humans , Parkinson Disease/immunology , Parkinson Disease/blood , Non-Fibrillar Collagens/immunology , Pemphigoid, Bullous/immunology , Pemphigoid, Bullous/blood , Autoantigens/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/blood , Autoantibodies/blood , Autoantibodies/immunology , Pemphigoid, Benign Mucous Membrane/immunology , Pemphigoid, Benign Mucous Membrane/blood , Immunoglobulin G/blood , Immunoglobulin G/immunology , Epitopes/immunology , Protein Domains , Female , Male , Aged
10.
Front Immunol ; 15: 1363032, 2024.
Article in English | MEDLINE | ID: mdl-38903493

ABSTRACT

Autoimmune blistering disorders (AIBDs) are a heterogeneous group of approximately a dozen entities comprising pemphigus and pemphigoid disorders and dermatitis herpetiformis. The exact diagnosis of AIBDs is critical for both prognosis and treatment and is based on the clinical appearance combined with the detection of tissue-bound and circulating autoantibodies. While blisters and erosions on the skin and/or inspectable mucosal surfaces are typical, lesions may be highly variable with erythematous, urticarial, prurigo-like, or eczematous manifestations. While direct immunofluorescence microscopy (IFM) of a perilesional biopsy is still the diagnostic gold standard, the molecular identification of the major target antigens opened novel therapeutic avenues. At present, most AIBDs can be diagnosed by the detection of autoantigen-specific serum antibodies by enzyme-linked immunosorbent assay (ELISA) or indirect IFM when the clinical picture is known. This is achieved by easily available and highly specific and sensitive assays employing recombinant immunodominant fragments of the major target antigens, i.e., desmoglein 1 (for pemphigus foliaceus), desmoglein 3 (for pemphigus vulgaris), envoplakin (for paraneoplastic pemphigus), BP180/type XVII collagen (for bullous pemphigoid, pemphigoid gestationis, and mucous membrane pemphigoid), laminin 332 (for mucous membrane pemphigoid), laminin ß4 (for anti-p200 pemphigoid), type VII collagen (for epidermolysis bullosa acquisita and mucous membrane pemphigoid), and transglutaminase 3 (for dermatitis herpetiformis). Indirect IFM on tissue substrates and in-house ELISA and immunoblot tests are required to detect autoantibodies in some AIBD patients including those with linear IgA disease. Here, a straightforward modern approach to diagnosing AIBDs is presented including diagnostic criteria according to national and international guidelines supplemented by long-term in-house expertise.


Subject(s)
Autoantibodies , Humans , Autoantibodies/immunology , Autoantibodies/blood , Autoimmune Diseases/diagnosis , Autoimmune Diseases/immunology , Autoantigens/immunology , Skin Diseases, Vesiculobullous/diagnosis , Skin Diseases, Vesiculobullous/immunology , Enzyme-Linked Immunosorbent Assay
11.
J Invest Dermatol ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38692406

ABSTRACT

Intravenous Ig (IVIg) is used to treat mucous membrane pemphigoid, although its therapeutic effectivity is not sufficiently supported by randomized controlled clinical trials, and its mode of action is only insufficiently understood. We have examined the effect of IVIg in a mouse model of anti-laminin 332 mucous membrane pemphigoid and found that IVIg ameliorates both cutaneous and mucosal inflammatory lesions. Our investigation into the modes of action of IVIg in mucous membrane pemphigoid indicated effective anti-inflammatory mechanisms beyond the enhanced degradation of IgG mediated through inhibition of the FcRn. Our results suggest that IVIg curbs the activation of neutrophils at several levels. This includes a direct, immediate inhibitory effect on neutrophil activation by immune complexes but not C5a, which blunts the release of ROS and leukotriene B4 from neutrophils. IVIg also suppresses the formation of neutrophil extracellular traps in response to calcium ion ionophore. In vivo treatment with IVIg altered the transcriptome of blood leukocytes and bone marrow neutrophils toward less proinflammatory phenotypes. Collectively, our results support the effectivity of IVIg in the treatment of mucous membrane pemphigoid and indicate that effects on neutrophils at multiple levels may significantly contribute to its therapeutic effects.

12.
Br J Dermatol ; 191(4): 539-547, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-38798074

ABSTRACT

BACKGROUND: According to current guidelines, systemic or topical corticosteroids are both recommended as first-line treatments for bullous pemphigoid (BP). There is evidence to suggest that topical application may be associated with a lower risk of mortality. However, there is a lack of comprehensive large-scale data comparing mortality rates, as well as the risk of major adverse cardiac events (MACE), infections and relapse, between systemic and topical corticosteroid treatments. OBJECTIVES: To evaluate the risk of death, MACE, infections and relapse in patients with BP treated with systemic or topical corticosteroids. METHODS: A population-based retrospective cohort study was performed using the TriNetX US Collaborative Network. As a measure against bias, propensity score matching for age, sex, 10 diseases and 6 medications was done, and 3 sensitivity analyses were conducted. RESULTS: All-time risk of death was increased in US patients with BP exposed to any dose of systemic corticosteroids (n = 2917) vs. patients treated with topical clobetasol propionate [n = 2932; hazard ratio (HR) 1.43, 95% confidence interval (CI) 1.28-1.58 (P < 0.001)]. This was consistent in time-stratified analysis (1- and 3-year mortality rates) and in analysis contrasting prednisone (equivalent) doses of 1-10 mg (low) or 30-100 mg (medium-high) systemic corticosteroid to topical treatment. The increased risk of death in US patients with BP exposed to any dose of systemic corticosteroids vs. topical treatment was accompanied by increased risks for MACE (HR 1.33, 95% CI 1.08-1.64; P = 0.008) and infections (HR 1.33, 95% CI 1.15-1.54; P < 0.001). The risk of continued disease or relapse was decreased in patients treated with systemic vs. topical corticosteroids (HR 0.85, 95% CI 0.77-0.94; P = 0.002). Results regarding mortality and continued disease or relapse persisted in three sensitivity analyses. Potential limitations included the retrospective data collection, bias for treatment selection and miscoding. CONCLUSIONS: Pending validation in prospective studies, where feasible - and despite the heightened risk of relapse - topical corticosteroid treatment may be advantageous over systemic corticosteroid treatment owing to its significantly lower risk of death.


Bullous pemphigoid (BP) is an autoimmune skin disease that commonly affects older people. The recommended treatment is medication called corticosteroids, either taken as a tablet or injection ('systemic') or applied to the skin ('topical'). Some evidence suggests that topical corticosteroids might be safer in terms of the risk of dying. We looked at whether there was a difference between the risk of dying, having heart problems, getting sick or the condition coming back in people with BP who use the different medications. To do this, we used people's past health information and compared people of similar ages and genders, 10 diseases and 6 medications. We found that people with BP who used systemic corticosteroids had a higher chance of dying. This also meant that people taking this form of the medication had a greater chance of having heart problems and getting infections. However, the risk of BP coming back was lower for people who used systemic corticosteroids. Our findings suggest that although there is a higher risk of BP coming back, there may be fewer risks from topical corticosteroids than systemic corticosteroids.


Subject(s)
Clobetasol , Pemphigoid, Bullous , Recurrence , Humans , Pemphigoid, Bullous/mortality , Pemphigoid, Bullous/drug therapy , Male , Female , Retrospective Studies , Aged , Clobetasol/administration & dosage , Clobetasol/adverse effects , Administration, Topical , Aged, 80 and over , Middle Aged , Glucocorticoids/administration & dosage , Glucocorticoids/adverse effects , Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/adverse effects , Heart Diseases/mortality , Administration, Cutaneous
13.
Int J Dermatol ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702860

ABSTRACT

INTRODUCTION: Uncertainty surrounds the optimal routine laboratory monitoring in acne patients treated with isotretinoin. OBJECTIVE: Our aim was to evaluate the risk of mild and severe laboratory abnormalities in patients with acne starting isotretinoin versus oral antibiotic treatment. METHODS: A global population-based retrospective cohort study assigned two groups of patients with acne-prescribed isotretinoin (n = 79,012) and oral antibiotics (n = 79,012). Comprehensive propensity-score matching was conducted. RESULTS: Compared to acne patients treated with oral antibiotics, those under isotretinoin demonstrated an increased risk of grade ≥3 hypertriglyceridemia (hazard ratio [HR], 7.85; 95% confidence interval [CI], 5.58-11.05; P < 0.001) and grade ≥3 elevated aspartate transaminase (AST) levels (HR, 1.45; 95% CI, 1.13-1.85; P = 0.003) within the initial 3 months of treatment. The absolute risk of these abnormalities among isotretinoin initiators was 0.4% and 0.2%, respectively. The risk difference of these findings was clinically marginal: 3 and 1 additional cases per 1,000 patients starting isotretinoin, respectively. There was no significant risk of grade ≥3 impairment in cholesterol, alanine transaminase, gamma-glutamyl transferase, or creatinine levels under isotretinoin. Most laboratory abnormalities were documented 1-3 months after drug initiation in time-stratified analysis. CONCLUSION: Isotretinoin is associated with a clinically marginal increased risk of severe hypertriglyceridemia and hypertransaminasemia. Routine blood testing should be performed 1-3 months after commencing therapy.

14.
J Dermatol Sci ; 114(2): 64-70, 2024 May.
Article in English | MEDLINE | ID: mdl-38582700

ABSTRACT

BACKGROUND: Bullous pemphigoid (BP), the most common subepidermal autoimmune blistering disease, is classically defined by the presence of IgG autoantibodies directed against the hemidesmosomal proteins BP180 (type XVII collagen) and BP230 and the predominance of skin lesions. Several studies have addressed the role of anti-BP180 IgE in patients and experimental models, while data on anti-BP230 IgE are scarce. OBJECTIVE: To assess anti-BP230 IgE level by ELISA in BP sera and to correlate it with disease severity and clinical characteristics. METHODS: BP sera underwent anti-BP230 IgE ELISA and Western blotting against human BP230 fragments. RESULTS: We demonstrate that 36/154 (23%) of BP sera were positive for anti-BP230 IgE. Anti-BP230 IgE levels had no correlation with clinical phenotype or disease activity per se. Interestingly, anti-BP230 IgE was significantly associated with disease activity within individuals during the course of the disease. Additionally, anti-BP230 IgE and total IgE levels showed a significant correlation. Notably, anti-BP230 IgG correlated interindividually with disease activity. By Western blotting, the C-terminal domain of BP230 fragments (C2; amino acids 2024-2349 and C3; amino acids 2326-2649), provided the best serological assay for anti-BP230 IgE detection. CONCLUSION: As a complementary tool, IgE immunoblotting is recommended to obtain an optimal serological diagnosis, particularly in patients with severe disease without IgG reactivity by BP180- or BP230-specific ELISA. Although the detection of serum anti-BP230 IgE is not of major diagnostic significance, it may be relevant for therapeutic decisions, e.g., for anti-IgE-directed treatment, which has been successfully used in case series of BP.


Subject(s)
Autoantibodies , Autoantigens , Dystonin , Enzyme-Linked Immunosorbent Assay , Immunoglobulin E , Immunoglobulin G , Non-Fibrillar Collagens , Pemphigoid, Bullous , Severity of Illness Index , Humans , Pemphigoid, Bullous/immunology , Pemphigoid, Bullous/blood , Pemphigoid, Bullous/diagnosis , Immunoglobulin E/blood , Immunoglobulin E/immunology , Autoantibodies/blood , Autoantibodies/immunology , Male , Female , Aged , Autoantigens/immunology , Dystonin/immunology , Aged, 80 and over , Non-Fibrillar Collagens/immunology , Middle Aged , Immunoglobulin G/blood , Immunoglobulin G/immunology , Collagen Type XVII , Adult , Blotting, Western
15.
J Invest Dermatol ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38642796

ABSTRACT

Pemphigus is a severe blistering disease caused by autoantibodies primarily against the desmosomal cadherins desmoglein (DSG)1 and DSG3, which impair desmosome integrity. Especially for the acute phase, additional treatment options allowing to reduce corticosteroids would fulfill an unmet medical need. In this study, we provide evidence that EGFR inhibition by erlotinib ameliorates pemphigus vulgaris IgG-induced acantholysis in intact human epidermis. Pemphigus vulgaris IgG caused phosphorylation of EGFR (Y845) and Rous sarcoma-related kinase in human epidermis. In line with this, a phosphotyrosine kinome analysis revealed a robust response associated with EGFR and Rous sarcoma-related kinase family kinase signaling in response to pemphigus vulgaris IgG but not to pemphigus foliaceus autoantibodies. Erlotinib inhibited pemphigus vulgaris IgG-induced epidermal blistering and EGFR phosphorylation, loss of desmosomes, as well as ultrastructural alterations of desmosome size, plaque symmetry, and keratin filament insertion and restored the desmosome midline considered as hallmark of mature desmosomes. Erlotinib enhanced both single-molecule DSG3-binding frequency and strength and delayed DSG3 fluorescence recovery, supporting that EGFR inhibition increases DSG3 availability and cytoskeletal anchorage. Our data indicate that EGFR is a promising target for pemphigus therapy owing to its link to several signaling pathways known to be involved in pemphigus pathogenesis.

17.
Dermatologie (Heidelb) ; 75(8): 641-646, 2024 Aug.
Article in German | MEDLINE | ID: mdl-38656540

ABSTRACT

Paraneoplastic pemphigus is a rare, life-threatening autoimmune disease that is clinically characterized by mostly extensive and refractory mucosal erosions and polymorphous skin lesions. We report here on a 16-year-old girl with isolated oral erosions, in whom mucosal pemphigoid was initially suspected and after treatment with prednisolone and dapsone marked improvement was achieved. However, a few months later the patient developed massive respiratory insufficiency as a result of bronchiolitis obliterans, so that a lung transplant was planned. As part of the preparatory diagnostic workup, unicentric, abdominally localized Castleman's disease was diagnosed, which ultimately led to the diagnosis of paraneoplastic pemphigus as evidenced by envoplakin autoantibodies. Tumor resection and subsequent lung transplantation achieved good results with sustained mucocutaneous remission.


Subject(s)
Castleman Disease , Paraneoplastic Syndromes , Pemphigus , Humans , Female , Castleman Disease/complications , Castleman Disease/immunology , Castleman Disease/diagnosis , Castleman Disease/drug therapy , Castleman Disease/pathology , Adolescent , Pemphigus/diagnosis , Pemphigus/immunology , Pemphigus/complications , Pemphigus/pathology , Pemphigus/drug therapy , Paraneoplastic Syndromes/diagnosis , Paraneoplastic Syndromes/immunology , Paraneoplastic Syndromes/pathology , Lung Transplantation , Bronchiolitis Obliterans/immunology , Bronchiolitis Obliterans/diagnosis , Bronchiolitis Obliterans/etiology , Bronchiolitis Obliterans/drug therapy
20.
Mol Ther ; 32(4): 1061-1079, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38382529

ABSTRACT

Complement-mediated diseases can be treated using systemic inhibitors. However, complement components are abundant in circulation, affecting systemic inhibitors' exposure and efficacy. Furthermore, because of complement's essential role in immunity, systemic treatments raise infection risk in patients. To address these challenges, we developed antibody fusion proteins combining the alternative-pathway complement inhibitor factor H (fH1-5) with an anti-C3d monoclonal antibody (C3d-mAb-2fH). Because C3d is deposited at sites of complement activity, this molecule localizes to tissue complement while minimizing circulating complement engagement. These fusion proteins bind to deposited complement in diseased human skin sections and localize to activated complement in a primate skin injury model. We further explored the pharmacology of C3d-mAb-2fH proteins in rodent models with robust tissue complement activation. Doses of C3d-mAb-2fH >1 mg/kg achieved >75% tissue complement inhibition in mouse and rat injury models while avoiding circulating complement blockade. Glomerular-specific complement inhibition reduced proteinuria and preserved podocyte foot-process architecture in rat membranous nephropathy, indicating disease-modifying efficacy. These data indicate that targeting local tissue complement results in durable and efficacious complement blockade in skin and kidney while avoiding systemic inhibition, suggesting broad applicability of this approach in treating a range of complement-mediated diseases.


Subject(s)
Complement Factor H , Kidney Diseases , Humans , Mice , Rats , Animals , Complement Factor H/genetics , Complement C3d/metabolism , Kidney Diseases/etiology , Antibodies , Complement Activation
SELECTION OF CITATIONS
SEARCH DETAIL