Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
FASEB J ; 34(7): 9337-9357, 2020 07.
Article in English | MEDLINE | ID: mdl-32463151

ABSTRACT

Thrombin converts fibrinogen to fibrin and activates blood and vascular cells in thrombo-inflammatory diseases. Platelets are amplifiers of thrombin formation when activated by leukocyte- and vascular cell-derived thrombin. CD36 on platelets acts as sensitizer for molecules with damage-associated molecular patterns, thereby increasing platelet reactivity. Here, we investigated the role of CD36 in thrombin-generation on human platelets, including selected patients with advanced chronic kidney disease (CKD). Platelets deficient in CD36 or blocked by anti-CD36 antibody FA6.152 showed impaired thrombin generation triggered by thrombin in calibrated automated thrombography. Using platelets with congenital function defects, blocking antibodies, pharmacological inhibitors, and factor-depleted plasma, CD36-sensitive thrombin generation was dependent on FXI, fibrin, and platelet signaling via GPIbα and SFKs. CD36-deficiency or blocking suppressed thrombin-induced platelet αIIbß3 activation, granule exocytosis, binding of adhesion proteins and FV, FVIII, FIX, FX, but not anionic phospholipid exposure determined by flow cytometry. CD36 ligated specifically soluble fibrin, which recruited distinct coagulation factors via thiols. Selected patients with CKD showed elevated soluble fibrin plasma levels and enhanced thrombin-induced thrombin generation, which was normalized by CD36 blocking. Thus, CD36 is an important amplifier of platelet-dependent thrombin generation when exposure of anionic phospholipids is limited. This pathway might contribute to hypercoagulability in CKD.


Subject(s)
Blood Platelets/metabolism , CD36 Antigens/metabolism , Factor XI/metabolism , Fibrin/metabolism , Renal Insufficiency, Chronic/metabolism , Thrombin/metabolism , Blood Coagulation Factors , Humans , Platelet Activation , Renal Insufficiency, Chronic/pathology
2.
Blood ; 130(6): 808-816, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28507083

ABSTRACT

Several studies showed that neutralizing anti-factor VIII (anti-fVIII) antibodies (inhibitors) in patients with acquired hemophilia A (AHA) and congenital hemophilia A (HA) are primarily directed to the A2 and C2 domains. In this study, the frequency and epitope specificity of anti-C1 antibodies were analyzed in acquired and congenital hemophilia inhibitor patients (n = 178). The domain specificity of antibodies was studied by homolog-scanning mutagenesis (HSM) with single human domain human/porcine fVIII proteins and antibody binding to human A2, C1, and C2 domains presented as human serum albumin (HSA) fusion proteins. The analysis with HSA-fVIII domain proteins confirmed the results of the HSM approach but resulted in higher detection levels. The higher detection levels with HSA-fVIII domain proteins are a result of antibody cross-reactivity with human and porcine fVIII leading to false-negative HSM results. Overall, A2-, C1-, and C2-specific antibodies were detected in 23%, 78%, and 68% of patients with AHA (n = 115) and in 52%, 57%, and 81% of HA inhibitor patients (n = 63). Competitive binding of the human monoclonal antibody (mAb) LE2E9 revealed overlapping epitopes with murine C1-specific group A mAbs including 2A9. Mutational analyses identified distinct crucial binding residues for LE2E9 (E2066) and 2A9 (F2068) that are also recognized by anti-C1 antibodies present in patients with hemophilia. A strong contribution of LE2E9- and 2A9-like antibodies was particularly observed in patients with AHA. Overall, our study demonstrates that the C1 domain, in addition to the A2 and C2 domains, contributes significantly to the humoral anti-fVIII immune response in acquired and congenital hemophilia inhibitor patients.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Factor VIII/immunology , Hemophilia A/immunology , Immunoglobulin G/immunology , Animals , Epitope Mapping , Factor VIII/chemistry , Humans , Mice , Protein Domains , Swine
3.
Haematologica ; 104(6): 1256-1267, 2019 06.
Article in English | MEDLINE | ID: mdl-30545925

ABSTRACT

In combination with microspotting, whole-blood microfluidics can provide high-throughput information on multiple platelet functions in thrombus formation. Based on assessment of the inter- and intra-subject variability in parameters of microspot-based thrombus formation, we aimed to determine the platelet factors contributing to this variation. Blood samples from 94 genotyped healthy subjects were analyzed for conventional platelet phenotyping: i.e. hematologic parameters, platelet glycoprotein (GP) expression levels and activation markers (24 parameters). Furthermore, platelets were activated by ADP, CRP-XL or TRAP. Parallel samples were investigated for whole-blood thrombus formation (6 microspots, providing 48 parameters of adhesion, aggregation and activation). Microspots triggered platelet activation through GP Ib-V-IX, GPVI, CLEC-2 and integrins. For most thrombus parameters, inter-subject variation was 2-4 times higher than the intra-subject variation. Principal component analyses indicated coherence between the majority of parameters for the GPVI-dependent microspots, partly linked to hematologic parameters, and glycoprotein expression levels. Prediction models identified parameters per microspot that were linked to variation in agonist-induced αIIbß3 activation and secretion. Common sequence variation of GP6 and FCER1G, associated with GPVI-induced αIIbß3 activation and secretion, affected parameters of GPVI-and CLEC-2-dependent thrombus formation. Subsequent analysis of blood samples from patients with Glanzmann thrombasthenia or storage pool disease revealed thrombus signatures of aggregation-dependent parameters that were subject-dependent, but not linked to GPVI activity. Taken together, this high-throughput elucidation of thrombus formation revealed patterns of inter-subject differences in platelet function, which were partly related to GPVI-induced activation and common genetic variance linked to GPVI, but also included a distinct platelet aggregation component.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Thrombosis/etiology , Thrombosis/metabolism , Biomarkers , Flow Cytometry , High-Throughput Screening Assays , Humans , Immunophenotyping , Platelet Aggregation , Platelet Count , Platelet Function Tests , Platelet Membrane Glycoproteins/metabolism , Thrombosis/diagnosis
4.
Blood Cells Mol Dis ; 67: 96-101, 2017 09.
Article in English | MEDLINE | ID: mdl-27865683

ABSTRACT

BACKGROUND: To better understand self-reported health-related quality-of-life (HrQoL) in children and adults with chronic hemostatic conditions compared with healthy controls. METHODS/PATIENTS/RESULTS: Group 1 consisted of 74 children/adolescents aged 8-18years with hereditary bleeding disorders (H-BD), 12 siblings and 34 peers. Group 2 consisted of 82 adult patients with hereditary/acquired bleeding disorders (H/A-BD), and group 3 of 198 patients with deep venous thrombosis (DVT) on anticoagulant therapy. Adult patients were compared to 1011 healthy blood donors. HrQoL was assessed with a 'revised KINDer Lebensqualitaetsfragebogen' (KINDL-R)-questionnaire adapted to adolescents and adults. No differences were found in multivariate analyses of self-reported HrQoL in children with H-BD. In contrast, apart from family and school-/work-related wellbeing in female patients with DVT the adult patients showed significantly lower HrQoL sub-dimensions compared to heathy control subjects. Furthermore, adults with H/A-BD disorders reported better friend-related HrQoL compared to patients with DVT, mainly due to a decreased HrQoL subscale in women on anticoagulation. CONCLUSION: In children with H-BD, HrQoL was comparable to siblings and peers. In adults with H/A-BD HrQoL was comparable to patients with DVT while healthy blood donors showed better HrQoL. The friend-related HrQoL subscale was significantly reduced in female compared to male patients.


Subject(s)
Blood Coagulation Disorders/epidemiology , Hemorrhage/epidemiology , Quality of Life , Venous Thrombosis/epidemiology , Adolescent , Adult , Age Factors , Aged , Blood Coagulation Disorders/psychology , Child , Family , Female , Hemorrhage/psychology , Humans , Male , Middle Aged , Sex Factors , Surveys and Questionnaires , Venous Thrombosis/psychology , Young Adult
5.
Blood ; 125(7): 1091-7, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25525118

ABSTRACT

Acquired hemophilia A (AHA) is caused by autoantibodies against factor VIII (FVIII). Immunosuppressive treatment (IST) results in remission of disease in 60% to 80% of patients over a period of days to months. IST is associated with frequent adverse events, including infections as a leading cause of death. Predictors of time to remission could help guide IST intensity but have not been established. We analyzed prognostic factors in 102 prospectively enrolled patients treated with a uniform IST protocol. Partial remission (PR; defined as no active bleeding, FVIII restored >50 IU/dL, hemostatic treatment stopped >24 hours) was achieved by 83% of patients after a median of 31 days (range 7-362). Patients with baseline FVIII <1 IU/dL achieved PR less often and later (77%, 43 days) than patients with ≥1 IU/dL (89%, 24 days). After adjustment for other baseline characteristics, low FVIII remained associated with a lower rate of PR (hazard ratio 0.52, 95% confidence interval 0.33-0.81, P < .01). In contrast, PR achieved on steroids alone within ≤21 days was more common in patients with FVIII ≥1 IU/dL and inhibitor concentration <20 BU/mL (odds ratio 11.2, P < .0001). Low FVIII was also associated with a lower rate of complete remission and decreased survival. In conclusion, presenting FVIII and inhibitor concentration are potentially useful to tailor IST in AHA.


Subject(s)
Hemophilia A , Immunosuppressive Agents/therapeutic use , Adult , Aged , Aged, 80 and over , Autoantibodies/adverse effects , Factor VIII/analysis , Factor VIII/immunology , Female , Hemophilia A/diagnosis , Hemophilia A/mortality , Hemophilia A/therapy , Humans , Male , Middle Aged , Prognosis , Remission Induction , Survival Analysis , Treatment Outcome
7.
Hamostaseologie ; 41(3): 190-196, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33860513

ABSTRACT

BACKGROUND: Currently available coronavirus disease 2019 (COVID-19) vaccines are approved for intramuscular injection and efficacy may not be ensured when given subcutaneously. For years, subcutaneous vaccination was recommended in patients with hemophilia to avoid intramuscular bleeds. Therefore, recommendations for the application of COVID-19 vaccines are needed. METHODS: The Delphi methodology was used to develop consensus recommendations. An initial list of recommendations was prepared by a steering committee and evaluated by 39 hemophilia experts. Consensus was defined as ≥75% agreement and strong consensus as ≥95% agreement, and agreement as a score ≥7 on a scale of 1 to 9. After four rounds, a final list of statements was compiled. RECOMMENDATIONS: Consensus was achieved that COVID-19 vaccines licensed only for intramuscular injection should be administered intramuscularly in hemophilia patients. Prophylactic factor replacement, given on the day of vaccination with a maximum interval between prophylaxis and vaccination of 24 hours (factor VIII and conventional factor IX concentrates) or 48 hours (half-life extended factor IX), should be provided in patients with moderate or severe hemophilia. Strong consensus was achieved that patients with mild hemophilia and residual factor activity greater than 10% with mild bleeding phenotype or patients on emicizumab usually do not need factor replacement before vaccination. Swelling, erythema, and hyperthermia after vaccination are not always signs of bleeding but should prompt consultation of a hemophilia care center. In case of injection-site hematoma, patients should receive replacement therapy until symptoms disappear. CONCLUSIONS: Consensus was achieved on recommendations for intramuscular COVID-19 vaccination after replacement therapy for hemophilia patients depending on disease severity.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , Hemophilia A/pathology , Hemophilia B/pathology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , COVID-19/virology , COVID-19 Vaccines/adverse effects , Factor IX/therapeutic use , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemophilia B/drug therapy , Humans , Injections, Intramuscular , SARS-CoV-2/isolation & purification , Severity of Illness Index
8.
Ann Hematol ; 88(4): 365-70, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19083235

ABSTRACT

Acquired haemophilia is an autoimmune disorder characterised by autoantibody formation against coagulation factor VIII. Immunosuppressive treatments including steroids, cytotoxic drugs, rituximab or combinations thereof have been used to eradicate autoantibodies. Very few prospective studies exist evaluating the use of these treatments. Here, we performed a survey among 73 physicians from 57 haemophilia treatment centres in order to describe current practice patterns and critical issues for future research in acquired haemophilia. The results demonstrate a high diversity of first- and second-line treatments. Factors influencing treatment decision were underlying disorder, severity of bleeding and inhibitor titre. Frequently used first-line treatments were steroids plus cyclophosphamide (44%) and steroids alone (11%). Second-line treatment was most often rituximab (30%), with or without steroids and/or cyclophosphamide. Most participants indicated to change from first- to second-line treatment after 4 weeks in case of failure to obtain partial remission (31%), continued bleeding (40%) or continued severe bleeding requiring bypass treatment (59%). Immunoadsorption was preferred for first- and second-line treatment by 10% and 9% of participants, respectively. These results highlight critical issues in the field. Open questions and directions for future research are discussed.


Subject(s)
Hemophilia A/drug therapy , Immunosuppressive Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Austria , Autoantibodies/blood , Cyclophosphamide/therapeutic use , Data Collection , Germany , Hemophilia A/etiology , Humans , Immunosorbent Techniques , Practice Patterns, Physicians' , Remission Induction , Rituximab , Steroids/therapeutic use , Switzerland , Treatment Failure
9.
Acta Neurochir (Wien) ; 151(8): 873-900; discussion 900, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19557305

ABSTRACT

BACKGROUND: Abnormalities of the hemostasis can lead to hemorrhage, and on the other hand to thrombosis. Intracranial neoplasms, complex surgical procedures, and head injury have a specific impact on coagulation and fibrinolysis. Moreover, the number of neurosurgical patients on medication (which interferes with platelet function and/or the coagulation systems) has increased over the past years. METHOD: The objective of this review is to recall common hemostatic disorders in neurosurgical patients on the basis of the "new concept of hemostasis". Therefore the pertinent literature was searched to provide a structured and up to date manuscript about hemostasis in Neurosurgery. FINDINGS: According to recent scientific publications abnormalities of the coagulation system are discussed. Pathophysiological background and the rational for specific (cost)-effective perioperative hemostatic therapy is provided. CONCLUSIONS: Perturbations of hemostasis can be multifactorial and maybe encountered in the daily practice of neurosurgery. Early diagnosis and specific treatment is the prerequisite for successful treatment and good patients outcome.


Subject(s)
Blood Coagulation Disorders/diagnosis , Blood Coagulation Disorders/physiopathology , Blood Coagulation/physiology , Hemorrhage/physiopathology , Hemostasis/physiology , Thrombosis/physiopathology , Algorithms , Blood Coagulation Disorders/therapy , Clinical Protocols , Coagulants/pharmacology , Coagulants/therapeutic use , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Hemorrhage/etiology , Hemorrhage/therapy , Hemostasis, Surgical/methods , Humans , Neurosurgical Procedures/adverse effects , Neurosurgical Procedures/methods , Thrombosis/etiology , Thrombosis/prevention & control
10.
Thromb Haemost ; 93(5): 867-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15886801

ABSTRACT

From 1998 to 2003, 133 Caucasian women aged 17-40 years (median 29 years) suffering from unexplained recurrent miscarriage (uRM) were consecutively enrolled. In patients and 133 age-matched healthy controls prothrombotic risk factors (factor V (FV) G1691A, factor II (FII) G20210A, MTHFR T677T, 4G/5G plasminogen activator inhibitor (PAI)-1, lipoprotein (Lp) (a), protein C (PC), protein S (PS), antithrombin (AT), antiphospholipid/anticardiolipin (APA/ACA) antibodies) as well as associated environmental conditions (smoking and obesity) were investigated. 70 (52.6%) of the patients had at least one prothrombotic risk factor compared with 26 control women (19.5%; p<0.0001). Body mass index (BMI; p=0.78) and smoking habits (p=0.44) did not differ significantly between the groups investigated. Upon univariate analysis the heterozygous FV mutation, Lp(a) > 30 mg/dL, increased APA/ACA and BMI > 25 kg/m(2) in combination with a prothrombotic risk factor were found to be significantly associated with uRM. In multivariate analysis, increased Lp(a) (odds ratio (OR): 4.7/95% confidence interval (CI): 2.0-10.7), the FV mutation (OR:3.8/CI:1.4-10.7), and increased APA/ACA (OR: 4.5/CI: 1.1-17.7) had independent associations with uRM.


Subject(s)
Abortion, Habitual/blood , Lipoprotein(a)/chemistry , Thrombosis/blood , Abortion, Habitual/diagnosis , Adolescent , Adult , Antibodies, Anticardiolipin/biosynthesis , Antibodies, Antiphospholipid/blood , Anticoagulants/pharmacology , Antithrombins/biosynthesis , Body Mass Index , Case-Control Studies , Factor V/biosynthesis , Female , Follow-Up Studies , Heterozygote , Humans , Lipoprotein(a)/biosynthesis , Logistic Models , Methylenetetrahydrofolate Dehydrogenase (NAD+)/biosynthesis , Multivariate Analysis , Mutation , Odds Ratio , Plasminogen Activator Inhibitor 1/biosynthesis , Protein C/biosynthesis , Protein S/biosynthesis , Prothrombin/biosynthesis , Risk Factors , Thrombosis/diagnosis
11.
Thromb Haemost ; 111(2): 249-57, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24196373

ABSTRACT

Mutations in the antithrombin (AT) gene can impair the capacity of AT to bind heparin (AT deficiency type IIHBS), its target proteases such as thrombin (type IIRS), or both (type IIPE). Type II AT deficiencies are almost exclusively caused by missense mutations, whereas type I AT deficiency can originate from missense or null mutations. In a retrospective cohort study, we investigated the impact of the type of mutation and type of AT deficiency on the manifestation of thromboembolic events in 377 patients with hereditary AT deficiencies (133 from our own cohort, 244 reported in the literature). Carriers of missense mutations showed a lower risk of venous thromboembolism (VTE) than those of null mutations (adjusted hazard ratio [HR] 0.39, 95% confidence interval [CI] 0.27-0.58, p<0.001), and the risk of VTE was significantly decreased among patients with type IIHBS AT deficiency compared to patients with other types of AT deficiency (HR 0.23, 95%CI 0.13-0.41, p<0.001). The risk of pulmonary embolism complicating deep-vein thrombosis was lower in all type II AT deficiencies compared to type I AT deficiency (relative risk 0.69, 95%CI 0.56-0.84). By contrast, the risk of arterial thromboembolism tended to be higher in carriers of missense mutations than in those with null mutations (HR 6.08-fold, 95%CI 0.74-49.81, p=0.093) and was 5.9-fold increased (95%CI 1.22-28.62, p=0.028) in type IIHBS versus other types of AT deficiency. Our data indicate that the type of inherited AT defect modulates not only the risk of thromboembolism but also the localisation and encourage further studies to unravel this phenomenon.


Subject(s)
Antithrombin III Deficiency/blood , Antithrombin III Deficiency/genetics , Antithrombin III/genetics , Arterial Occlusive Diseases/blood , Arterial Occlusive Diseases/genetics , Blood Coagulation/genetics , Mutation, Missense , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Thromboembolism/blood , Thromboembolism/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Heredity , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Odds Ratio , Phenotype , Proportional Hazards Models , Retrospective Studies , Risk Factors , Venous Thromboembolism/blood , Venous Thromboembolism/genetics , Young Adult
13.
Thromb Haemost ; 105(4): 635-46, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21264449

ABSTRACT

Antithrombin (AT) is the most important physiological inhibitor of coagulation proteases. It is activated by glycosaminoglycans such as heparin. Hereditary antithrombin deficiency is a rare disease that is mainly associated with venous thromboembolism. So far, more than 200 different mutations in the antithrombin gene (SERPINC1) have been described. The aim of our study was to characterise the molecular background in a large cohort of patients with AT deficiency. Mutation analysis was performed by direct sequencing of SERPINC1 in 272 AT-deficient patients. Large deletions were identified by multiplex PCR coupled with liquid chromatography or multiplex ligation-dependent probe amplification (MLPA) analysis. To predict the effect of SERPINC1 sequence variations on the pathogenesis of AT deficiency, in silico assessments, multiple sequence alignment, and molecular graphic imaging were performed. The mutation profile consisted of 59% missense, 10% nonsense, 8% splice site mutations, 15% small deletions/insertions/duplications, and 8% large deletions. Altogether 87 different mutations, including 42 novel mutations (22 missense and 20 null mutations), were identified. Of the novel missense mutations, nine are suspected to impair the conformational changes that are needed for AT activation, two to affect the central reactive loop or the heparin binding site, and six to impair the structural integrity of the molecule. Despite the heterogeneous background of AT deficiency, 10 AT variants occurred in multiple index patients. Characterisation of the SERPINC1 mutation profile in large cohorts of patients may help to further elucidate the pathogenesis of AT deficiency and to establish genotype-phenotype associations.


Subject(s)
Antithrombin III Deficiency/genetics , Antithrombin III/genetics , Mutation/genetics , Animals , Antithrombin III Deficiency/blood , Antithrombin III Deficiency/epidemiology , Binding Sites/genetics , Cohort Studies , Computer Simulation , DNA Mutational Analysis , Family , Heparin/metabolism , Humans , Protein Binding/genetics , Protein Conformation , Protein Stability , Sequence Alignment
14.
J Clin Apher ; 22(4): 233-40, 2007.
Article in English | MEDLINE | ID: mdl-17610289

ABSTRACT

BACKGROUND: Acquired hemophilia A in a setting of bleeding or required surgery frequently places patients into a state of critical illness with high mortality. In this context immunoadsorption (IA) can be used to eliminate coagulation inhibitors quickly to employ recombinant coagulation factors more effectively. However, since acquired hemophilia is a rare condition the therapy is little standardized. METHODS: We report on a retrospective analysis of nine cases of acquired hemophilia A treated with IA using disposable adsorber columns. Data collection was performed by retrospectively reviewing the patients' files regarding clinical course, mode of therapy, inhibitor titers, and coagulation status. RESULTS: Inhibitor titers were effectively reduced in all but one patient following the treatment with IA. In two out of seven patients surviving the acute bleeding an inhibitor relapse occurred. The overall remission rate was determined as 77.8% within a median follow-up of 613 days. In two of our nine patients fatal outcome resulted due to major bleeding complications. IA treatment showed good tolerability and no fatal complications were caused. CONCLUSION: The presented cases support our assumption that patients with acquired hemophilia A benefit from IA with disposable columns in a setting of acute bleeding. This modality of IA is able to eliminate inhibitors reliably and quickly. IA in general is substantially speeding up the progress of therapy preventing bleeding complications constantly threatening the patient and reducing the dosages of coagulation factor therapy. We encourage IA with disposable columns in all bleeding patients with acquired hemophilia to aggressively lower the inhibitors.


Subject(s)
Coagulants/therapeutic use , Hemophilia A/complications , Hemorrhage/therapy , Aged , Combined Modality Therapy , Female , Hemorrhage/drug therapy , Hemorrhage/etiology , Humans , Immunosorbent Techniques/instrumentation , Male , Middle Aged
15.
Br J Haematol ; 129(5): 644-52, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15916687

ABSTRACT

The therapeutic efficacy of plasma exchange (PE) in thrombotic thrombocytopenic purpura (TTP) is attributed to the restoration in ADAMTS-13 (a disintegrin and metalloproteinase with thrombospondin motif-13) activity by substitution of the enzyme and removal of ADAMTS-13-neutralizing autoantibodies. We explored this rationale by analysing ADAMTS-13 activity and corresponding inhibitor levels during PE-treatment in 27 episodes from 23 adults with TTP. All patients with an initial episode of TTP (n = 14) and nine of 11 patients with a relapse showed severe ADAMTS-13 deficiency. ADAMTS-13 inhibitors were detected in 81% of these patients. Twenty-one patients responded to PE-therapy and two patients died. For patients with severe ADAMTS-13 deficiency, 15 patients (71%) showed a PE-induced recovery in ADAMTS-13 activity and six patients (29%) had persistent severe ADAMTS-13 deficiency despite clinical response. Three patients with recurrent TTP demonstrated a permanent increase in inhibitor titre during therapy. Six patients (43%) with an initial episode of TTP displayed a transient increase in inhibitor titre during PE-therapy, which was associated with deterioration in clinical and haematological symptoms of TTP. Treatment with vincristine induced an immediate increase in platelet count and ADAMTS-13 activity in seven of eight patients. We conclude that ADAMTS-13 activity and inhibitor levels, as measured using current methodology, do not solely determine the clinical course of TTP.


Subject(s)
Antineoplastic Agents/therapeutic use , Metalloendopeptidases/blood , Plasma Exchange , Purpura, Thrombotic Thrombocytopenic/enzymology , Purpura, Thrombotic Thrombocytopenic/therapy , Vincristine/therapeutic use , ADAM Proteins , ADAMTS13 Protein , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Female , Humans , Male , Metalloendopeptidases/antagonists & inhibitors , Middle Aged , Platelet Count , Protease Inhibitors/metabolism , Purpura, Thrombotic Thrombocytopenic/immunology , Recurrence , Rituximab , Statistics, Nonparametric , Treatment Outcome , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL