Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 332
Filter
1.
N Engl J Med ; 390(18): 1690-1698, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38718359

ABSTRACT

In patients with immune thrombotic thrombocytopenic purpura (iTTP), autoantibodies against the metalloprotease ADAMTS13 lead to catastrophic microvascular thrombosis. However, the potential benefits of recombinant human ADAMTS13 (rADAMTS13) in patients with iTTP remain unknown. Here, we report the clinical use of rADAMTS13, which resulted in the rapid suppression of disease activity and complete recovery in a critically ill patient whose condition had proved to be refractory to all available treatments. We also show that rADAMTS13 causes immune complex formation, which saturates the autoantibody and may promote its clearance. Our data support the role of rADAMTS13 as a novel adjunctive therapy in patients with iTTP.


Subject(s)
ADAMTS13 Protein , Purpura, Thrombotic Thrombocytopenic , Female , Humans , ADAMTS13 Protein/immunology , ADAMTS13 Protein/therapeutic use , Antigen-Antibody Complex/blood , Antigen-Antibody Complex/immunology , Autoantibodies/blood , Autoantibodies/immunology , Purpura, Thrombotic Thrombocytopenic/diagnosis , Purpura, Thrombotic Thrombocytopenic/drug therapy , Purpura, Thrombotic Thrombocytopenic/immunology , Purpura, Thrombotic Thrombocytopenic/therapy , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Adult , Black or African American , Plasma Exchange , Treatment Outcome
2.
Anal Chem ; 96(16): 6209-6217, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38607319

ABSTRACT

Vaccine-induced immune thrombotic thrombocytopenia (VITT) is a rare but dangerous side effect of adenoviral-vectored COVID-19 vaccines. VITT had been linked to production of autoantibodies recognizing platelet factor 4 (PF4). Here, we characterize anti-PF4 antibodies obtained from a VITT patient's blood. Intact mass measurements indicate that a significant fraction of these antibodies represent a limited number of clones. MS analysis of large antibody fragments (the light chain and the Fc/2 and Fd fragments of the heavy chain) confirms the monoclonal nature of this component of the anti-PF4 antibodies repertoire and reveals the presence of a mature complex biantennary N-glycan within the Fd segment. Peptide mapping using two complementary proteases and LC-MS/MS was used to determine the amino acid sequence of the entire light chain and over 98% of the heavy chain (excluding a short N-terminal segment). The sequence analysis allows the monoclonal antibody to be assigned to the IgG2 subclass and verifies that the light chain belongs to the λ-type. Incorporation of enzymatic de-N-glycosylation into the peptide mapping routine allows the N-glycan in the Fab region of the antibody to be localized to the framework 3 region of the VH domain. This novel N-glycosylation site is the result of a single mutation within the germline sequence. Peptide mapping also provides information on lower-abundance (polyclonal) components of the anti-PF4 antibody ensemble, revealing the presence of all four subclasses (IgG1-IgG4) and both types of the light chain (λ and κ). This case study demonstrates the power of combining the intact, middle-down, and bottom-up MS approaches for meaningful characterization of ultralow quantities of pathogenic antibodies extracted directly from patients' blood.


Subject(s)
Platelet Factor 4 , Humans , Platelet Factor 4/immunology , Platelet Factor 4/chemistry , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , COVID-19 Vaccines/chemistry , Autoantibodies/immunology , Autoantibodies/blood , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry , Amino Acid Sequence , Purpura, Thrombotic Thrombocytopenic/chemically induced , Purpura, Thrombotic Thrombocytopenic/immunology
3.
J Thromb Haemost ; 22(5): 1358-1365, 2024 May.
Article in English | MEDLINE | ID: mdl-38360215

ABSTRACT

BACKGROUND: Immune-mediated thrombotic thrombocytopenic purpura is caused by autoantibodies against ADAMTS-13, a plasma enzyme that cleaves von Willebrand factor. However, the mechanism resulting in severe deficiency of plasma ADAMTS-13 activity remains controversial. OBJECTIVES: To determine the mechanism of autoantibody-mediated severe deficiency of plasma ADAMTS13 activity in immune-mediated thrombotic thrombocytopenic purpura. METHODS: Fluorescence resonance energy transfer-VWF73 was used to determine plasma ADAMTS-13 activity. Enzyme-linked immunosorbent assay (ELISA) was used to determine anti-ADAMTS-13 immunoglobulin G. ELISA and capillary electrophoresis-based Western blotting were employed to assess plasma ADAMTS-13 antigen. RESULTS: We showed that plasma ADAMTS-13 antigen levels varied substantially in the samples collected on admission despite all showing plasma ADAMTS-13 activity of <10 IU/dL (or <10% of normal level) using either ELISA or Western blotting. More severe deficiency of plasma ADAMTS-13 antigen (<10%) was detected in admission samples by ELISA than by capillary Western blotting. There was a significant but moderate correlation between plasma ADAMTS-13 activity and ADAMTS-13 antigen by either assay method, suggesting that severe deficiency of plasma ADAMTS-13 activity is not entirely associated with low levels of ADAMTS-13 antigen. CONCLUSION: We conclude that severe deficiency of plasma ADAMTS-13 activity primarily resulted from antibody-mediated inhibition, but the accelerated clearance of plasma ADAMTS-13 antigen via immune complexes may also contribute significantly to severe deficiency of plasma ADAMTS-13 activity in a subset of patients with acute immune-mediated thrombotic thrombocytopenic purpura.


Subject(s)
ADAM Proteins , ADAMTS13 Protein , Autoantibodies , Enzyme-Linked Immunosorbent Assay , Purpura, Thrombotic Thrombocytopenic , ADAMTS13 Protein/blood , ADAMTS13 Protein/immunology , Humans , Purpura, Thrombotic Thrombocytopenic/blood , Purpura, Thrombotic Thrombocytopenic/immunology , Purpura, Thrombotic Thrombocytopenic/diagnosis , Purpura, Thrombotic Thrombocytopenic/enzymology , Autoantibodies/blood , Male , ADAM Proteins/blood , ADAM Proteins/immunology , ADAM Proteins/deficiency , Adult , Female , Middle Aged , Immunoglobulin G/blood , Fluorescence Resonance Energy Transfer , Blotting, Western , von Willebrand Factor/metabolism , von Willebrand Factor/analysis , Aged
4.
Br J Haematol ; 204(5): 1994-1998, 2024 May.
Article in English | MEDLINE | ID: mdl-38168722

ABSTRACT

Pregnancy is a potential trigger of acute thrombotic thrombocytopenic purpura (TTP). The management of pregnancy-associated immune-mediated TTP (iTTP) can be challenging, especially when it is refractory to standard treatment. Caplacizumab, a nanobody to von Willebrand factor (VWF) blocking its A1 domain, is a valuable new therapeutic option. Its use is, however, not approved during pregnancy and breastfeeding. We describe the successful off-label administration of caplacizumab during pregnancy and delivery in a patient with refractory iTTP. The favourable outcome without significant thrombotic or haemorrhagic complications indicates that caplacizumab may be an effective and safe treatment option in refractory iTTP during pregnancy.


Subject(s)
Pregnancy Complications, Hematologic , Purpura, Thrombotic Thrombocytopenic , Single-Domain Antibodies , Humans , Pregnancy , Female , Purpura, Thrombotic Thrombocytopenic/drug therapy , Purpura, Thrombotic Thrombocytopenic/immunology , Single-Domain Antibodies/therapeutic use , Adult , Pregnancy Complications, Hematologic/drug therapy , von Willebrand Factor/antagonists & inhibitors
6.
Front Immunol ; 13: 837629, 2022.
Article in English | MEDLINE | ID: mdl-35273612

ABSTRACT

Both qualitative and quantitative platelet abnormalities are common in patients with coronavirus disease 2019 (COVID-19) and they correlate with clinical severity and mortality. Activated platelets contribute to the prothrombotic state in COVID-19 patients. Several groups have shown immune-mediated activation of platelets in critically ill COVID-19 patients. Vaccine-induced immune thrombotic thrombocytopenia is an autoimmune condition characterized by thrombocytopenia and life-threatening thrombotic events in the arterial and venous circulation. Although the initial trigger has yet to be determined, activation of platelets by immune complexes through Fc gamma RIIA results in platelet consumption and thrombosis. A better understanding of platelet activation in COVID-19 as well as in vaccine-induced thrombotic complications will have therapeutic implications. In this review, we focused on the role of immune-mediated platelet activation in thrombotic complications during COVID-19 infection and vaccine-induced immune thrombotic thrombocytopenia.


Subject(s)
Blood Platelets/physiology , COVID-19/immunology , Purpura, Thrombotic Thrombocytopenic/immunology , SARS-CoV-2/physiology , Animals , Blood Coagulation , Humans , Platelet Activation , Vaccination/adverse effects
9.
Front Immunol ; 12: 757192, 2021.
Article in English | MEDLINE | ID: mdl-34858410

ABSTRACT

Immune thrombotic thrombocytopenic purpura (iTTP) is an autoimmune disorder of which the etiology is not fully understood. Autoantibodies targeting ADAMTS13 in iTTP patients have extensively been studied, the immunological mechanisms leading to the breach of tolerance remain to be uncovered. This review addresses the current knowledge on genetic factors associated with the development of iTTP and the interplay between the patient's immune system and environmental factors in the induction of autoimmunity against ADAMTS13. HLA-DRB1*11 has been identified as a risk factor for iTTP in the Caucasian population. Interestingly, HLA-DRB1*08:03 was recently identified as a risk factor in the Japanese population. Combined in vitro and in silico MHC class II peptide presentation approaches suggest that an ADAMTS13-derived peptide may bind to both HLA-DRB1*11 and HLA-DRB1*08:03 through different anchor-residues. It is apparent that iTTP is associated with the presence of infectious microorganisms, viruses being the most widely associated with development of iTTP. Infections may potentially lead to loss of tolerance resulting in the shift from immune homeostasis to autoimmunity. In the model we propose in this review, infections disrupt the epithelial barriers in the gut or lung, promoting exposure of antigen presenting cells in the mucosa-associated lymphoid tissue to the microorganisms. This may result in breach of tolerance through the presentation of microorganism-derived peptides that are homologous to ADAMTS13 on risk alleles for iTTP.


Subject(s)
Purpura, Thrombotic Thrombocytopenic/immunology , ADAMTS13 Protein/immunology , Antigen-Presenting Cells/immunology , Autoantibodies/immunology , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Forecasting , Gastrointestinal Microbiome , Genes, MHC Class II , Genetic Predisposition to Disease , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , Humans , Immunity, Mucosal/immunology , Infections/complications , Molecular Mimicry , Polymorphism, Single Nucleotide , Purpura, Thrombotic Thrombocytopenic/etiology , Purpura, Thrombotic Thrombocytopenic/genetics , Self Tolerance , Toll-Like Receptor 9/genetics , Vaccines/adverse effects
11.
Am J Hematol ; 96(12): 1587-1594, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34460124

ABSTRACT

Cardiovascular disease is a leading cause of death in survivors of immune-mediated thrombotic thrombocytopenic purpura (iTTP), but the epidemiology of major adverse cardiovascular events (MACE) in iTTP survivors is unknown. We evaluated the prevalence and risk factors for MACE, defined as the composite of non-fatal or fatal myocardial infarction (MI), stroke, and cardiac revascularization, during clinical remission in two large iTTP cohorts (Johns Hopkins University and Ohio State University). Of 181 patients followed for ≥ 3 months after recovery from acute iTTP, 28.6% had a MACE event over a median follow up of 7.6 years. Stroke was the most common type of MACE (18.2%), followed by non-fatal MI (6.6%), cardiac revascularization (4.9%) and fatal MI (0.6%). Compared to the general United States population, iTTP survivors were younger at first stroke in remission (males [56.5 years vs. 68.6 years, p = 0.031], females [49.7 years vs. 72.9 years, p < 0.001]) or MI in remission (males [56.5 years vs. 65.6 years, p < 0.001] and females [53.1 years vs. 72.0 years, p < 0.001]). Age (HR 1.03 [95% CI 1.002-1.054]), race (Black/Other vs. White) (HR 2.32 [95% CI 1.12-4.82]), and diabetes mellitus (HR 2.37 [95% CI 1.09-0.03]) were associated with MACE in a Cox regression model also adjusted for sex, hypertension, obesity, hyperlipidemia, chronic kidney disease, atrial fibrillation, autoimmune disease, and relapsing iTTP. Remission ADAMTS13 activity was not significantly associated with MACE. In conclusion, iTTP survivors experience high rates of MACE and may benefit from aggressively screening for and managing cardiovascular risk factors.


Subject(s)
Cardiovascular Diseases/etiology , Purpura, Thrombotic Thrombocytopenic/complications , Adult , Aged , Cardiovascular Diseases/immunology , Cohort Studies , Female , Heart Disease Risk Factors , Humans , Male , Middle Aged , Myocardial Infarction/etiology , Myocardial Infarction/immunology , Prevalence , Purpura, Thrombotic Thrombocytopenic/immunology , Stroke/etiology , Stroke/immunology
12.
Transfus Apher Sci ; 60(4): 103174, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34088601

ABSTRACT

Vaccine-induced immune thrombotic thrombocytopenia (VITT) is associated with high titers of immunoglobulin G class antibodies directed against the cationic platelet chemokine platelet factor 4 (PF4). These antibodies activate platelets via FcγIIa receptors. VITT closely resembles heparin-induced thrombocytopenia. Inflammation and tissue trauma substantially increase the risk for forming pathogenic PF4 antibodies. We therefore propose the use of therapeutic plasma exchange as rescue therapy in VITT to deplete antibodies plus factors promoting inflammation such as excess cytokines in the circulation as well as extracellular vesicles derived from activated platelets.


Subject(s)
COVID-19 Vaccines/adverse effects , Plasma Exchange , Platelet Factor 4/immunology , Purpura, Thrombotic Thrombocytopenic/therapy , Salvage Therapy , Albumins , Antibody Specificity , Anticoagulants , Autoantibodies/immunology , COVID-19 Vaccines/pharmacology , ChAdOx1 nCoV-19 , Citrates , Contraindications, Procedure , Cytokines/blood , Extracellular Vesicles , Humans , Immunoglobulin G/immunology , Immunosuppression Therapy , Inflammation , Plasma Exchange/adverse effects , Plasma Exchange/methods , Platelet Activation , Platelet Transfusion/adverse effects , Purpura, Thrombotic Thrombocytopenic/etiology , Purpura, Thrombotic Thrombocytopenic/immunology , Registries , Thrombocytopenia/etiology , Thrombosis/etiology
13.
Blood ; 138(4): 299-303, 2021 07 29.
Article in English | MEDLINE | ID: mdl-33988688

ABSTRACT

Vaccination using the adenoviral vector COVID-19 vaccine ChAdOx1 nCoV-19 (AstraZeneca) has been associated with rare vaccine-induced immune thrombotic thrombocytopenia (VITT). Affected patients test strongly positive in platelet factor 4 (PF4)/polyanion enzyme immunoassays (EIAs), and serum-induced platelet activation is maximal in the presence of PF4. We determined the frequency of anti-PF4/polyanion antibodies in healthy vaccinees and assessed whether PF4/polyanion EIA+ sera exhibit platelet-activating properties after vaccination with ChAdOx1 nCoV-19 (n = 138) or BNT162b2 (BioNTech/Pfizer; n = 143). In total, 19 of 281 participants tested positive for anti-PF4/polyanion antibodies postvaccination (All: 6.8% [95% confidence interval (CI), 4.4-10.3]; BNT162b2: 5.6% [95% CI, 2.9-10.7]; ChAdOx1 nCoV-19: 8.0% [95% CI, 4.5% to 13.7%]). Optical densities were mostly low (between 0.5 and 1.0 units; reference range, <0.50), and none of the PF4/polyanion EIA+ samples induced platelet activation in the presence of PF4. We conclude that positive PF4/polyanion EIAs can occur after severe acute respiratory syndrome coronavirus 2 vaccination with both messenger RNA- and adenoviral vector-based vaccines, but many of these antibodies likely have minor (if any) clinical relevance. Accordingly, low-titer positive PF4/polyanion EIA results should be interpreted with caution when screening asymptomatic individuals after vaccination against COVID-19. Pathogenic platelet-activating antibodies that cause VITT do not occur commonly following vaccination.


Subject(s)
Autoantibodies/immunology , COVID-19 Vaccines/adverse effects , COVID-19/prevention & control , Platelet Factor 4/immunology , Polyelectrolytes , Purpura, Thrombotic Thrombocytopenic/etiology , Vaccination/adverse effects , Adult , Asymptomatic Diseases , Autoantibodies/blood , BNT162 Vaccine , ChAdOx1 nCoV-19 , Female , Health Personnel , Humans , Immunoenzyme Techniques , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , Platelet Activation , Purpura, Thrombotic Thrombocytopenic/immunology , Seroconversion , Thrombophilia/etiology
15.
Blood ; 137(19): 2694-2698, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33544829

ABSTRACT

Immune-mediated thrombotic thrombocytopenic purpura (iTTP) is an autoimmune disorder caused by the development of autoantibodies targeting different domains of ADAMTS13. Profiling studies have shown that residues R568, F592, R660, Y661, and Y665 within exosite-3 of the spacer domain provide an immunodominant region of ADAMTS13 for pathogenic autoantibodies that develop in patients with iTTP. Modification of these 5 core residues with the goal of reducing autoantibody binding revealed a significant tradeoff between autoantibody resistance and proteolytic activity. Here, we employed structural bioinformatics to identify a larger epitope landscape on the ADAMTS13 spacer domain. Models of spacer-antibody complexes predicted that residues R568, L591, F592, K608, M609, R636, L637, R639, R660, Y661, Y665, and L668 contribute to an expanded epitope within the spacer domain. Based on bioinformatics-guided predictions, we designed a panel of N-glycan insertions in this expanded epitope to reduce the binding of spacer domain autoantibodies. One N-glycan variant (NGLY3-ADAMTS13, containing a K608N substitution) showed strongly reduced reactivity with TTP patient sera (28%) as compared with WT-ADAMTS13 (100%). Insertion of an N-glycan at amino acid position 608 did not interfere with processing of von Willebrand factor, positioning the resulting NGLY3-ADAMTS13 variant as a potential novel therapeutic option for treatment of iTTP.


Subject(s)
ADAMTS13 Protein/immunology , Antigen-Antibody Complex/chemistry , Antigen-Antibody Reactions , Autoantibodies/immunology , Autoantigens/immunology , Polysaccharides/immunology , Purpura, Thrombotic Thrombocytopenic/immunology , ADAMTS13 Protein/chemistry , ADAMTS13 Protein/metabolism , Amino Acid Substitution , Amino Acids , Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , Autoantibodies/metabolism , Autoantigens/chemistry , Autoantigens/metabolism , Epitopes/immunology , Epitopes/metabolism , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , von Willebrand Factor/metabolism
16.
Crit Care Med ; 49(5): e512-e520, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33591004

ABSTRACT

OBJECTIVES: Prevention and therapy of immunothrombosis remain crucial challenges in the management of coronavirus disease 2019, since the underlying mechanisms are incompletely understood. We hypothesized that endothelial damage may lead to substantially increased concentrations of von Willebrand factor with subsequent relative deficiency of a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13). DESIGN: Prospective controlled cross-over trial. SETTING: Blood samples of patients with confirmed coronavirus disease 2019 and healthy controls were obtained in three German hospitals and analyzed in a German hemostaseologic laboratory. PATIENTS: Seventy-five patients with confirmed coronavirus disease 2019 of mild to critical severity and 30 healthy controls. MEASUREMENTS AND MAIN RESULTS: von Willebrand factor antigen, ADAMTS13, and von Willebrand factor multimer formation were analyzed. von Willebrand factor antigen was 4.1 times higher in COVID-19 patients compared with healthy controls (p < 0.0001), whereas ADAMTS13 activities were not significantly different (p = 0.18). The ADAMTS13/von Willebrand factor antigen ratio was significantly lower in COVID-19 than in the control group (24.4 ± 20.5 vs 82.0 ± 30.7; p < 0.0001). Fourteen patients (18.7%) undercut a critical ratio of 10 as described in thrombotic thrombocytopenic purpura. Gel analysis of multimers resembled a thrombotic thrombocytopenic purpura pattern with loss of the largest multimers in 75% and a smeary triplet pattern in 39% of the patients. The ADAMTS13/von Willebrand factor antigen ratio decreased continuously from mild to critical disease (analysis of variance p = 0.026). Furthermore, it differed significantly between surviving patients and those who died from COVID-19 (p = 0.001) yielding an area under the curve of 0.232 in receiver operating characteristic curve curve analysis. CONCLUSION: COVID-19 is associated with a substantial increase in von Willebrand factor levels, which can exceed the ADAMTS13 processing capacity resulting in the formation of large von Willebrand factor multimers indistinguishable from thrombotic thrombocytopenic purpura. The ADAMTS13/von Willebrand factor antigen ratio is an independent predictor of severity of disease and mortality. These findings provide a rationale to consider plasma exchange as a therapeutic option in COVID-19 and to include von Willebrand factor and ADAMTS13 in the diagnostic workup.


Subject(s)
ADAMTS13 Protein/deficiency , COVID-19/blood , COVID-19/immunology , Purpura, Thrombotic Thrombocytopenic/immunology , SARS-CoV-2/immunology , von Willebrand Factor/metabolism , Aged , Aged, 80 and over , Cross-Over Studies , Female , Germany/epidemiology , Humans , Male , Middle Aged , Plasma Exchange , Prospective Studies , Purpura, Thrombotic Thrombocytopenic/therapy
18.
Transfusion ; 61(1): 322-328, 2021 01.
Article in English | MEDLINE | ID: mdl-33119913

ABSTRACT

BACKGROUND: Checkpoint inhibitors enhance T-lymphocyte-mediated antitumor responses, resulting in increased survival for patients with neoplastic disease. However, a subset of patients receiving checkpoint inhibitor therapy may experience adverse complications that include the development of autoimmune conditions, such as thrombotic thrombocytopenic purpura (TTP). Given the potential etiologic differences of checkpoint inhibitor-related autoimmunity, TTP that develops in the presence of checkpoint inhibitors may be refractory to current treatment methods and therefore may require additional treatment and prognostic consideration. CASE REPORT: Herein, we describe the unique clinical course of a patient who was treated with the combined checkpoint inhibitors nivolumab and ipilimumab for Stage IV malignant melanoma, who subsequently developed TTP. Unlike many patients with TTP, this patient failed to develop a sustained response to therapeutic plasma exchange. Additional use of steroids, anti-CD20, and plasma cell-targeting therapy (bortezomib) also failed to substantially reverse thrombocytopenia in a sustainable fashion. During this time, her melanoma progressed, and she ultimately succumbed. CONCLUSION: This case illustrates not only that TTP may be a potential complication of checkpoint inhibitor therapy, but also that TTP developing in this setting may result in an unpredictable response to commonly employed TTP treatment modalities. Ultimately, checkpoint inhibitor-related TTP may require distinct management approaches and prognostic considerations.


Subject(s)
Immune Checkpoint Inhibitors/adverse effects , Melanoma/drug therapy , Purpura, Thrombotic Thrombocytopenic/chemically induced , Autoimmunity/immunology , Disease Progression , Fatal Outcome , Female , Humans , Immunotherapy/adverse effects , Melanoma/classification , Middle Aged , Neoplasm Staging , Plasma Exchange/methods , Purpura, Thrombotic Thrombocytopenic/immunology , Purpura, Thrombotic Thrombocytopenic/therapy , Skin Neoplasms/pathology
20.
Intern Med ; 59(10): 1315-1321, 2020.
Article in English | MEDLINE | ID: mdl-32418955

ABSTRACT

We herein report a patient with mixed connective tissue disease (MCTD) who had been stable for years but suddenly developed thrombotic thrombocytopenic purpura (TTP). The patient showed a clinical pentad of signs of TTP, low activity of ADAMTS13, and positivity of anti-ADAMTS13 antibodies. She did not respond to plasma exchange or steroid therapy and died five days after admission. An autopsy revealed microthrombi in the brain, heart, kidney, adrenal glands, esophageal submucosa, and bone marrow as well as diffuse alveolar hemorrhaging. Physicians should bear in mind that TTP can occur in MCTD patients regardless of disease activity.


Subject(s)
Mixed Connective Tissue Disease/complications , Purpura, Thrombotic Thrombocytopenic/complications , ADAMTS13 Protein/immunology , Adrenal Cortex Hormones/therapeutic use , Autoantibodies/blood , Autopsy , Fatal Outcome , Female , Humans , Male , Plasma Exchange , Purpura, Thrombotic Thrombocytopenic/diagnosis , Purpura, Thrombotic Thrombocytopenic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...