Skip to main content
Erschienen in: memo - Magazine of European Medical Oncology 3/2018

Open Access 17.08.2018 | review

Thrombotic thrombocytopenic purpura

verfasst von: Prof. Dr. Paul Knöbl, MD

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 3/2018

Summary

Thrombotic thrombocytopenic purpura (TTP) is a clearly defined entity of the thrombotic microangiopathies (TMA), a heterogeneous group of disorders characterized by microangiopathic hemolytic anemia with red cell fragmentation, thrombocytopenia and organ dysfunction due to disturbed microcirculation. TTP is characterized by a severe deficiency of ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13), an enzyme responsible for physiological cleavage of von Willebrand factor (VWF). Organ dysfunction can be severe and life-threatening, and immediate start of appropriate therapy is necessary to avoid permanent damage or death. Until recently, therapeutic options were limited to symptomatic measures, which were not standardized or based on high scientific evidence. In recent years, not only considerable progress has been made in better diagnosis of TTP, but also new therapeutic strategies have been established. Initial treatment is still based on plasma exchange and symptomatic measures to protect organ function, but new concepts (immunosuppression, targeted anti-VWF or anti-complement therapy, replacement with recombinant enzymes) have recently demonstrated impressive advantages.

Introduction

Thrombotic thrombocytopenic purpura (TTP) is a well-defined entity of a heterogeneous group of disorders, the thrombotic microangiopathies (TMA). TMAs are characterized by microangiopathic hemolytic anemia with red cell fragmentation, thrombocytopenia and signs of organ dysfunction due to disturbed microcirculation. TTP is a rare disorder with an incidence of about 5 per million per year. It was first described in 1924 by Eli Moschcowitz, who reported a 16-year-old girl who died after acute onset hemolytic anemia, thrombocytopenia, petechiae, fever and severe neurological symptoms [1]. In 1982, Moake et al. recognized the abnormal composition of von Willebrand factor (VWF) multimers in the plasma of patients with TTP [2], and in 1998 Furlan et al. [3] and Tsai and Lian [4] identified ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) deficiency as the pathogenic cause of TTP. In recent years, not only considerable progress has been made in the diagnosis of TTP, but also new therapeutic strategies have been established [57], including new drugs targeting specific parts of the pathophysiologic processes leading to TTP.

Thrombotic thrombocytopenic purpura

The current pathophysiological concept understands TTP as a state of severe deficiency of ADAMTS13, which can be caused either by genetic abnormalities (congenital TTP) or by autoantibodies affecting function or clearance of ADAMTS13 (autoimmune TTP). Lack of ADAMTS13 leads to the persistence of UL-VWF MM (ultralarge VWF multimeres). In the presence of additional triggers causing shear stress and unfolding of VWF (pregnancy, infections, certain drugs, surgery, etc.) enhanced platelet aggregation with the UL-VWF MM occurs. These platelet aggregates affect the blood flow in the microcirculation and cause organ damage and clinical symptoms [8]. In TTP, the central nervous system is mainly affected [9], but other organs especially involved are the kidneys and the heart. VWF-rich platelet thrombi containing low/no fibrin can be found in the capillaries, both smaller and larger vessels, in histological samples of patients with acute TTP.
The determination of ADAMTS13 activity is essential in TTP, as only very low levels (below the detection limit of most assays) are associated with TTP. As soon as at least low ADAMTS13 activity is detectable (i. e. >10%), VWF can be cleaved and TTP will not occur. Current laboratory methods (FRETS-VWF73 or GST-VWF73) have a low detection limit, great accuracy and fast throughput, older methods (collagen-binding assay, VWF degradation assays) are essentially outdated. Measurement of ADAMTS13 antigen may reveal detectable levels, as ADAMTS13 protein or immune complexes are also detected. Other assays are used to detect antibodies binding to ADAMTS13 and to distinguish between congenital and acquired TTP. Functional inhibitors of ADAMTS13 are quantified by dilution series according to the Bethesda method used for FVIII inhibitors [10].

Congenital ADAMTS 13 deficiency (Upshaw–Schulman syndrome)

Numerous mutations and polymorphisms in the ADAMTS13 gene are known [11, 12], leading to a severe reduction of ADAMTS13 activity (below the detection limit of most assays, i. e. <1 U/mL). Congenital (familial) TTP (OMIM No. 274150; http://​www.​ncbi.​nlm.​nih.​gov/​omim) can manifest early in childhood, but also later in life (in women often during the first pregnancy) and tends to relapse. Bouts of TTP can be triggered by factors associated with high shear rates (infections, pregnancy, drugs, etc.). Individuals with higher endogenous ADAMTS13 activity are usually safe and never experience TTP [13].
Diagnosis is performed by demonstrating the lack of ADAMTS13 activity with an assay of appropriate sensitivity, ruling out anti-ADAMTS13 antibodies (by testing for ADAMTS13 inhibitors and ADAMTS13-binding antibodies) and by sequencing the ADAMTS13 gene. ADAMTS13 antigen (but representing dysfunctional protein) may be detectable in the plasma, depending on the type of mutation. An international registry on patients with Upshaw–Schulman syndrome (www.​clinicaltrials.​gov; NCT01257269) is currently collecting all available cases of this rare disorder [14].

Acquired ADAMTS13 deficiency (autoimmune TTP)

Autoantibodies targeting ADAMTS13 either inhibit the function or enhance clearance. Several factors are known as triggers of the autoimmune process, such as infections with viruses (Epstein–Barr virus, cytomegalovirus, HIV, etc.) or other pathogens, malignancy, certain drugs, other concomitant autoimmune diseases, pregnancy, but in many cases no underlying disorder is found. During the oligo-/polyclonal immune reaction several types of autoantibodies can be formed over time [15], and the detection of these antibodies is dependent on the assays used [16, 17].
Diagnosis is performed by demonstrating the absence of ADAMTS13 activity with an assay of appropriate sensitivity, and the detection of anti-ADAMTS13 antibodies (by testing for ADAMTS13 inhibitors and/or ADAMTS13-binding antibodies) confirms the immunologic nature, but sensitivity of the available assays is moderate and there may be false-positive and false-negative results. ADAMTS13 antigen (representing ADAMTS13 bound in immune complexes) may be detectable [18, 19].

Clinical symptoms

The key clinical symptoms of TTP are, as for all types of TMA, symptoms of Coombs negative hemolysis with red cell fragmentation (recognized by anemia, elevated LDH, free serum hemoglobin, reticulocyte and schistocyte counts, reduced haptoglobin levels, and hemoglobinurea), consumption thrombocytopenia and signs disturbed microcirculation. The symptoms of organ dysfunction are often nonspecific and very variable [5, 7, 10]. Brain hypoperfusion can cause a broad variety of unspecific neurological symptoms, ranging from headache, blurry speech, dizziness, or agitation to stroke, amaurosis, epileptic seizures or coma. Renal involvement leads to increased serum creatinine, oligo- or anuria, and hemolysis-induced hemoglobinuria. Hemolysis may also cause jaundice and signs of anemia; thrombocytopenia is only sometimes associated with purpura, and bleeding is rare. Cardiac involvement with increases in cardiac enzymes (troponin, creatine kinase of cardiac isotype), patterns in the electrocardiogram resembling myocardial hypoperfusion/infarction, or arrhythmia, and the development of myocardial failure, is a dangerous complication of TTP, which may lead to immediate death of the patient [20]. Other organ manifestations may include lung (gas exchange problems, lung infiltrates), pancreas (increased enzymes, diabetes) or gut [21].

Initial diagnosis

A patient presenting with a bout of TTP is one of the most challenging hematological emergencies [68]. Immediate appropriate diagnostic procedures (Table 1) are necessary to clearly identify TTP and to distinguish this from other forms of TMA. Careful examination of the patient’s history will reveal possible causes of TMA. Before any therapeutic approach is started, the patient should be screened for the eligibility for a clinical trial, and necessary procedures need to be performed before start of therapy (informed consent, sample acquisition, randomization, etc.). Moreover, samples of patient’s plasma, serum and blood cells should be obtained to be either stored for later analysis (biobanking) and immediate analysis of ADAMTS13-, VWF-, and complement-related parameters. Assessment of organ function should be performed with appropriate methods. In any case, until diagnosis has been confirmed, TTP should be suspected and immediate appropriate therapy with plasma exchange and steroids (Table 2) should be initiated without unnecessary delay to achieve the best outcome for affected patients [68].
Table 1
Diagnostic approach to acute thrombotic microangiopathy
Condition
Diagnostic tests
Hemolysis
Hemoglobin, red blood cells, indices
Reticulocyte and schistocyte counts
Lactate dehydrogenase, haptoglobin
Direct antiglobin test (DAT; Coombs test)
Thrombocytopenia
Platelet counts; Immature platelet fraction
Organ damage
Brain
Imaging: CT scana, perfusion MRIa
Electroencephalograma
S100 beta, neuron-specific enolase
Neurocognitive testinga
Kidneys
Serum creatinine, glomerular filtration rate
Urine output
Heart
Electrocardiogram
Troponin, NT-proBNP
Echocardiography
Lung
Oxygen saturation, gas exchange
Imaging: chest x‑ray, high-resolution lung CT scana
Coagulation
Plasmatic coagulation assays
Antiphospholipid antibodies
Pancreas
Blood glucose, serum lipase
Specific diagnosis
General
Biobanking, sampling for possible clinical trials
Blood group typing
Pregnancy testa
Tests for viral infections (HIV, hepatitis B and C)
Urine analysis
Thyroid function tests
Thrombotic thrombocytopenic purpura (TTP)
ADAMTS13 activity, antigen
Anti-ADAMTS13 antibodies and -inhibitor
VWF:Ag, VWF:activity, VWF:RCo, VWF:CBA, VWF multimeric pattern
ADAMTS13 gene analysis
Hemolytic uremic syndrome (HUS)
Tests for bacterial infection/toxins (E. coli, Shigella, etc)
Complement C3 activation products, C4, CH50, APH50, C5a, terminal complement complex, CFH antibody
Complement factors gene analyses
Medical history
Concomitant and previous diseases
Underlying conditions (cancer, infections, systemic diseases, transplantation, pregnancy, surgery)
Drugs, medication
Family history
CT computerized tomography; MRI magnetic resonance imaging, BNP brain natriuretic peptide; VWF von Willebrand factor; Ag antigen; RCo ristocetin cofactor activity; CBA collagen binding activity; CFH anti-complement factor H; HIV human immunodeficiency virus; ADAMTS13 a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13; CH50 total complement activity; APH50 complement alternate pathway 50
aWhen clinically indicated and appropriate
Table 2
Therapeutic options for thrombotic thrombocytopenic purpura
Therapeutic option
Indication
Dose
Mechanism of action
Established options
Plasma exchange
Initial therapy in all types of TMA
Treatment of choice in autoimmune TTP
60–80 ml/kg/day
Elimination of autoantibodies, immune complexes, UL-VWF MM, sludge
Replacement of ADAMTS13 and regularly composed VWF
Plasma infusion
Congenital ADAMTS13 deficiency (Upshaw–Schulman syndrome)
10–40 ml/kg every
2–3 weeks
Replacement of ADAMTS13
Corticosteroids
Autoantibody-induced TTP
1–2 mg/kg/day
Immunosuppression
Rituximab
Autoantibody-induced TTP
4 doses of 100–375 mg/m2/week
Immunosuppression
Immunomodulators (vincristine, MMF, cyclosporine, cyclophosphamide)
Autoantibody-induced TTP
(3rd line immunotherapy)
As indicated
Immunosuppression
Anti-platelet agents (ASS, clopidogrel, prasugrel, ticagrelor)
TTP with severe organ damage
Clopidogrel:
75–150 mg/day
Inhibition of platelet aggregation
Splenectomy
Refractory TTP
(after rituximab failure)
Unknown.
Elimination of memory cells?
Supportive therapy
Anemia: RBC transfusion
organ failure: ICU
(Details: see text)
Future options
Caplacizumaba
Acute autoimmune TTP
10 mg/day sc
Blocking VWF A1 domains, competition with platelet GP Ib/IX
Recombinant ADAMTS13b
Congenital deficiency of ADAMTS13 (Upshaw-Schulman syndrome)
20–40 U/kg every 2–4 weeks
Replacement of ADAMTS13
Recombinant ADAMTS13b
Autoimmune TTP?
Unknown
Replacement of ADAMTS13 to overcome inhibitors
N-acetylcysteineb
Acute and chronic TTP
300 mg/kg/day
Cleavage of UL-VWF MM
TMA thrombotic microangiopathy; TTP thrombotic thrombocytopenic purpura; UL-VWF MM unusually large von Willebrand factor multimers; RBC red blood cells; ICU intensive care unit; ADAMTS13 a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13; GP glycoprotein
aEarly access program available
bIn development

Therapeutic options for TTP

Plasma exchange therapy

Plasma exchange (PEX) was introduced in the treatment of TTP by Rock et al. [22] and has improved survival from about 10 to 80–90%. During this procedure, the 1.5-fold plasma volume is removed and replaced by donor’s plasma (either fresh frozen single donor plasma or pooled, virus-inactivated plasma are acceptable replacement fluids; cryosupernatant is also used in some countries). During PEX, autoantibodies, UL-VWF MM, immune complexes and sludge are removed, and ADAMTS13 and VWF-MM of normal composition are replaced. In patients with TTP, daily PEX should be continued until platelet counts and LDH are normal and signs of hemolysis and organ dysfunction have resolved. In refractory cases and severe organ dysfunction, treatment intensity can be increased either by increasing the exchanged plasma volume or by performing PEX twice daily [68].
Patients with congenital ADAMTS13 deficiency usually respond rapidly and platelet counts normalize within a few days. Patients with secondary types of TMA (i. e. transplant-associated, some types of drug- or infection-induced TMA) will not respond to PEX, and treatment should not be continued except as a symptomatic measure for patients in poor condition.

Plasma infusion

Therapy of congenital TTP is usually performed by plasma infusions to substitute for the missing enzyme. Even acute bouts with severe symptoms usually respond well to the infusion of plasma (20–40 ml per kg body weight). In patients with frequent relapses or smoldering disease regularly prophylactic plasma infusions (every 2–3 weeks) are necessary, as such patients often have low-grade, but detectable neurologic disturbance [2325]. In autoimmune TTP, however, response to plasma infusions is poor [22] and should be used only to bridge the time to the start of sufficient PEX therapy.

Immunosuppression

Immunosuppression with corticosteroids (i. e. prednisone, 1–2 mg/kg/day) is useful in cases of autoimmune TTP to suppress further antibody formation. In addition, even in other forms of TMA, steroids may be useful to reduce shear stress and improve endothelial function. Moreover, data from the Oklahoma Registry suggest that the number of PEX necessary to achieve remission and the incidence of complications of PEX were considerably reduced since the introduction of steroids in TTP therapy [26]. Steroids are usually maintained until hematological remission or until resolution of the autoimmune process, monitoring ADAMTS13 activity and anti-ADAMTS13 antibodies is helpful to guide therapy.
Several reports have been published on the effects of rituximab to eliminate anti-ADAMTS13 antibodies in patients with TTP [2729]. Efficacy to eradicate the autoantibodies is high; 95% had a complete clinical and laboratory response within 1–3 weeks, and the effect usually lasts for more than 2 years. A randomized clinical trial (STAR trial; NCT00799773), however, has been terminated due to a low enrollment rate. Considering the high risks of permanent organ damage in relapsing or refractory TMA, the reported side effects of rituximab have to be weighed against the chance to obtain a long-lasting response.
Refractory or frequently relapsing patients (e.g., failure of steroids) are often treated with splenectomy or fourth line immunosuppressive treatment with vincristine, mycophenolate mofetil (MMF), cyclophosphamide, bortezomib, or bendamustine (all of them off-label and based on case reports only [30, 31]).

Supportive care

Transfusion of red blood cells is necessary when hemolysis causes severe anemia, but the optimal transfusion threshold is not determined. Platelet transfusions are considered to aggravate platelet aggregation and disturbance of microcirculation, and current guidelines give a 1 A recommendation to avoid platelet transfusions unless there is life-threatening hemorrhage [7]. Intensive care treatment is often necessary in patients with severe organ damage. Neurologic deterioration or brain ischemia may require sedation and mechanical ventilation, antiepileptic therapy and an experienced intensive care team (stroke unit). Acute coronary syndrome requires hemodynamic monitoring, myocardial or circulatory support, or coronary interventions. Renal failure requires careful fluid and electrolyte management and often renal replacement. During all these sophisticated medical treatments, PEX and specific TMA treatment need to be continued, which may cause logistic challenges. Prophylaxis of venous thromboembolism is necessary and recommended even with lower platelet counts [7]. Low molecular weight heparins are usually used, monitoring of dose is necessary in patients with renal dysfunction.

New/approaching therapeutic options for TTP

Anti-VWF agents: ALX-0081 (caplacizumab; Cablivi®)

Caplacizumab is a bivalent nanobody, recombinantly composed from the functional fragments of the variable domains of the heavy-chain-only immunoglobulins from camelidae (llama). It is designed to bind specifically and with a high affinity to the A1 domains of VWF, the physiological ligands of platelet receptors GP Ib/IX. Thus, caplacizumab competes with platelet binding and therefore inhibits platelet aggregation and activation, but does not affect collagen binding or ADAMTS13 susceptibility of VWF. Caplacizumab can be administered subcutaneously and causes suppression of VWF function for up to 48 h after a single dose [32]. Caplacizumab has been studied two randomized controlled trials (TITAN [33, 34], EudraCT 2010-019375-30 and HERCULES [35], EudraCT 2015-001098-42) in patients with acute autoimmune TTP, and has been proven to reduce the time to platelet normalization, organ damage, exacerbations, health care resource utilization and mortality. The drug is available via an early access program; it will soon be approved by the authorities under the brand name Cablivi®.

Recombinant ADAMTS13

The current development of recombinant ADAMTS13 (rADAMTS13) as a therapeutic agent to replace for the missing enzyme will probably be a great advancement in the treatment of congenital TTP [36]. The possibility to avoid plasma therapy with all the disadvantages (time-consuming, thawing, large fluid volume, potential risk of pathogen transmission, side effects, immunogenicity, allergic reaction) and to establish prophylactic or therapeutic home-treatment (like in hemophiliacs) will be a clear improvement of the current situation. Clinical trials with rADAMTS13 in congenital TTP are in progress and approval can hopefully be expected within the next few years [37]. Due to the long half-life and the low necessary plasma concentration of ADAMTS13 prophylactic therapy in Upshaw–Shulman syndrome will probably be sufficient with 20–40 U/kg every 2–4 weeks, depending on the results of the current trials. Dosing for acute bouts needs to be determined, but single or dual shots of the same dose may be sufficient to terminate an episode. There is, however, the considerable risk of inducing anti-ADAMTS13 alloantibodies during treatment (like in hemophiliacs with FVIII inhibitors), which will cause major problems, as neither plasma infusion nor PEX or rADAMTS13 will be sufficient to treat acute bouts of TTP.

N-acetylcysteine

N-acetylcysteine (NAC) is an antioxidative substance that has already been clinically used as a mucolytic agent for many years [38]. In models using human plasma, purified VWF and ADAMTS13 knock-out mice it could be demonstrated that NAC leads to a reduction of UL-VWF MM by disrupting the disulfide bonds in the A1 domain of VWF [39]. This also leads to a reduced binding of platelets to VWF released from endothelial cells [39]. Considering the pathophysiology of TTP, cleavage of the UL-VWF MM by a substance with low side effects would be advantageous. One recent case report that showed promising results in a patient with refractory TTP treated with NAC has been published [40]. Thus, a clinical trial with NAC in addition to PEX for the treatment of TTP has been started and is currently recruiting (N-acetylcysteine in TTP. U.S. National Institutes of Health http://​clinicaltrials.​gov/​show/​NCT01808521).

Concluding remarks

Considerable progress has been made in diagnosis and therapy of TTP. New assays to measure ADAMTS13 activity and anti-ADAMTS13 antibodies can establish the diagnosis (distinguish between congenital and autoimmune TTP and the different other types of TMA) within a few hours and are useful to guide treatment. Consequent PEX therapy clearly has improved survival. Congenital TTP responds well to the replacement of ADAMTS13 by plasma infusion. Autoimmune types of TTP often respond to PEX, immunosuppression and rituximab.
The near future will probably bring a variety of other therapeutic possibilities: blocking the VWF-platelet interaction with anti-VWF A1 agents, replacement of ADAMTS13 by a recombinant ADAMTS13 concentrate, cleavage of VWF with NAC, or better immunosuppressive strategies. However, the low incidence of TTP clearly reduces the possibility to perform randomized controlled trials with a sufficient number of patients. Even in ongoing worldwide trials recruitment is poor, thus, making the development of a new drug expensive and time-consuming. This encourages the treating physicians to use off-label treatments when the therapeutic principle fits in the pathophysiological understanding of the disease. The severity of the disease and the high risk of developing permanent damage clearly justifies such an approach in some situations.

Conflict of interest

P. Knöbl declares that he served as member of steering boards and advisory boards and received consultancy, speaker and travel fees from Ablynx, Shire, and Alexion.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Moschcowitz E. Hyaline thrombosis of the terminal arterioles and capillaries: A hitherto undescribed disease. Proc NY Pathol Soc. 1924;24:21–4. Moschcowitz E. Hyaline thrombosis of the terminal arterioles and capillaries: A hitherto undescribed disease. Proc NY Pathol Soc. 1924;24:21–4.
2.
Zurück zum Zitat Moake JL, Rudy CK, Troll JH, et al. Unusually large plasma factor VIII: Von Willebrand factor multimers in chronic relapsing thrombotic thrombocytopenic purpura. N Engl J Med. 1982;307(23):1432–5.CrossRefPubMed Moake JL, Rudy CK, Troll JH, et al. Unusually large plasma factor VIII: Von Willebrand factor multimers in chronic relapsing thrombotic thrombocytopenic purpura. N Engl J Med. 1982;307(23):1432–5.CrossRefPubMed
3.
Zurück zum Zitat Furlan M, Robles R, Galbusera M, et al. Von Willebrand factor-cleaving protease in thrombotic thrombocytopenic purpura and the hemolytic-uremic syndrome. N Engl J Med. 1998;339(22):1578–84.CrossRefPubMed Furlan M, Robles R, Galbusera M, et al. Von Willebrand factor-cleaving protease in thrombotic thrombocytopenic purpura and the hemolytic-uremic syndrome. N Engl J Med. 1998;339(22):1578–84.CrossRefPubMed
4.
Zurück zum Zitat Tsai HM, Lian EC. Antibodies to von Willebrand factor-cleaving protease in acute thrombotic thrombocytopenic purpura. N Engl J Med. 1998;339(22):1585–94.CrossRefPubMedPubMedCentral Tsai HM, Lian EC. Antibodies to von Willebrand factor-cleaving protease in acute thrombotic thrombocytopenic purpura. N Engl J Med. 1998;339(22):1585–94.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Knöbl PN. Treatment of thrombotic microangiopathy with a focus on new treatment options. Haemostaseologie. 2013;33(2):149–59.CrossRef Knöbl PN. Treatment of thrombotic microangiopathy with a focus on new treatment options. Haemostaseologie. 2013;33(2):149–59.CrossRef
6.
Zurück zum Zitat Scully M, Hunt BJ, Benjamin S, et al. British Committee for Standards in Haematology. Guidelines on the diagnosis and management of thrombotic thrombocytopenic purpura and other thrombotic microangiopathies. Br J Haematol. 2012;158(3):323–35.CrossRefPubMed Scully M, Hunt BJ, Benjamin S, et al. British Committee for Standards in Haematology. Guidelines on the diagnosis and management of thrombotic thrombocytopenic purpura and other thrombotic microangiopathies. Br J Haematol. 2012;158(3):323–35.CrossRefPubMed
7.
Zurück zum Zitat Fontana S, Kremer Hovinga JA, Lämmle B, et al. Treatment of thrombotic thrombocytopenic purpura. Vox Sang. 2006;90(4):245–54.CrossRefPubMed Fontana S, Kremer Hovinga JA, Lämmle B, et al. Treatment of thrombotic thrombocytopenic purpura. Vox Sang. 2006;90(4):245–54.CrossRefPubMed
8.
Zurück zum Zitat Chapman K, Seldon M, Richards R. Thrombotic microangiopathies, thrombotic thrombocytopenic purpura, and ADAMTS-13. Semin Thromb Hemost. 2012;38(1):47–54.CrossRefPubMed Chapman K, Seldon M, Richards R. Thrombotic microangiopathies, thrombotic thrombocytopenic purpura, and ADAMTS-13. Semin Thromb Hemost. 2012;38(1):47–54.CrossRefPubMed
9.
Zurück zum Zitat Lämmle B, Kremer Hovinga JA, Alberio L. Thrombotic thrombocytopenic purpura. J Thromb Haemost. 2005;3(8):1663–75.CrossRefPubMed Lämmle B, Kremer Hovinga JA, Alberio L. Thrombotic thrombocytopenic purpura. J Thromb Haemost. 2005;3(8):1663–75.CrossRefPubMed
10.
Zurück zum Zitat Tripodi A, Peyvandi F, Chantarangkul V, et al. Second international collaborative study evaluating performance characteristics of methods measuring the von Willebrand factor cleaving protease (ADAMTS-13). J Thromb Haemost. 2008;6(9):1534–41.CrossRefPubMed Tripodi A, Peyvandi F, Chantarangkul V, et al. Second international collaborative study evaluating performance characteristics of methods measuring the von Willebrand factor cleaving protease (ADAMTS-13). J Thromb Haemost. 2008;6(9):1534–41.CrossRefPubMed
11.
Zurück zum Zitat Kokame K, Kokubo Y, Miyata T. Polymorphisms and mutations of ADAMTS13 in the Japanese population and estimation of the number of patients with Upshaw-Schulman syndrome. J Thromb Haemost. 2011;9(8):1654–6.CrossRefPubMed Kokame K, Kokubo Y, Miyata T. Polymorphisms and mutations of ADAMTS13 in the Japanese population and estimation of the number of patients with Upshaw-Schulman syndrome. J Thromb Haemost. 2011;9(8):1654–6.CrossRefPubMed
12.
Zurück zum Zitat Miyata T, Kokame K, Matsumoto M, Fujimura Y. ADAMTS13 activity and genetic mutations in Japan. Haemostaseologie. 2013;33(2):131–7.CrossRef Miyata T, Kokame K, Matsumoto M, Fujimura Y. ADAMTS13 activity and genetic mutations in Japan. Haemostaseologie. 2013;33(2):131–7.CrossRef
13.
Zurück zum Zitat Fujimura Y, Matsumoto M, Isonishi A, Yagi H, Kokame K, Soejima K, Murata M, Miyata T. Natural history of Upshaw-Schulman syndrome based on ADAMTS13 gene analysis in Japan. J Thromb Haemost. 2011;9:283–301.CrossRefPubMed Fujimura Y, Matsumoto M, Isonishi A, Yagi H, Kokame K, Soejima K, Murata M, Miyata T. Natural history of Upshaw-Schulman syndrome based on ADAMTS13 gene analysis in Japan. J Thromb Haemost. 2011;9:283–301.CrossRefPubMed
14.
Zurück zum Zitat Mansouri Taleghani M, von Krogh AS, Fujimura Y, George JN, Hrachovinová I, Knöbl PN, et al. Hereditary thrombotic thrombocytopenic purpura and the hereditary TTP registry. Hamostaseologie. 2013;33(2):138–43.CrossRefPubMed Mansouri Taleghani M, von Krogh AS, Fujimura Y, George JN, Hrachovinová I, Knöbl PN, et al. Hereditary thrombotic thrombocytopenic purpura and the hereditary TTP registry. Hamostaseologie. 2013;33(2):138–43.CrossRefPubMed
15.
Zurück zum Zitat Scheiflinger F, Knöbl P, Trattner B, et al. Nonneutralizing IgM and IgG antibodies to von Willebrand factor-cleaving protease (ADAMTS-13) in a patient with thrombotic thrombocytopenic purpura. Blood. 2003;102:3241–3.CrossRefPubMed Scheiflinger F, Knöbl P, Trattner B, et al. Nonneutralizing IgM and IgG antibodies to von Willebrand factor-cleaving protease (ADAMTS-13) in a patient with thrombotic thrombocytopenic purpura. Blood. 2003;102:3241–3.CrossRefPubMed
16.
Zurück zum Zitat Tsai HM. Autoimmune thrombotic microangiopathy: Advances in pathogenesis, diagnosis, and management. Semin Thromb Hemost. 2012;38(5):469–82.CrossRefPubMed Tsai HM. Autoimmune thrombotic microangiopathy: Advances in pathogenesis, diagnosis, and management. Semin Thromb Hemost. 2012;38(5):469–82.CrossRefPubMed
17.
Zurück zum Zitat Schaller M, Studt JD, Voorberg J, Kremer Hovinga JA. Acquired thrombotic thrombocytopenic purpura. Development of an autoimmune response. Haemostaseologie. 2013;33(2):121–30.CrossRef Schaller M, Studt JD, Voorberg J, Kremer Hovinga JA. Acquired thrombotic thrombocytopenic purpura. Development of an autoimmune response. Haemostaseologie. 2013;33(2):121–30.CrossRef
18.
Zurück zum Zitat Ferrari S, Knöbl P, Kolovratova V, et al. Inverse correlation of free and immune complex-sequestered anti-ADAMTS13 antibodies in a patient with acquired thrombotic thrombocytopenic purpura. J Thromb Haemost. 2012;10(1):156–8.CrossRefPubMed Ferrari S, Knöbl P, Kolovratova V, et al. Inverse correlation of free and immune complex-sequestered anti-ADAMTS13 antibodies in a patient with acquired thrombotic thrombocytopenic purpura. J Thromb Haemost. 2012;10(1):156–8.CrossRefPubMed
20.
Zurück zum Zitat Mariotte E, Azoulay E, Galicier L, et al. Epidemiology and pathophysiology of adulthood-onset thrombotic microangiopathy with severe ADAMTS13 deficiency (thrombotic thrombocytopenic purpura): A cross-sectional analysis of the French national registry for thrombotic microangiopathy. Lancet Haematol. 2016;3:e237–e45.CrossRefPubMed Mariotte E, Azoulay E, Galicier L, et al. Epidemiology and pathophysiology of adulthood-onset thrombotic microangiopathy with severe ADAMTS13 deficiency (thrombotic thrombocytopenic purpura): A cross-sectional analysis of the French national registry for thrombotic microangiopathy. Lancet Haematol. 2016;3:e237–e45.CrossRefPubMed
21.
Zurück zum Zitat Knöbl P. Inherited and acquired thrombotic thrombocytopenic purpura (TTP) in adults. Semin Thromb Hemost. 2014;40:493–502.CrossRefPubMed Knöbl P. Inherited and acquired thrombotic thrombocytopenic purpura (TTP) in adults. Semin Thromb Hemost. 2014;40:493–502.CrossRefPubMed
22.
Zurück zum Zitat Rock GA, Shumak KH, Buskard NA, et al. Comparison of plasma exchange with plasma infusion in the treatment of thrombotic thrombocytopenic purpura. Canadian Apheresis Study Group. N Engl J Med. 1991;325(6):393–7.CrossRefPubMed Rock GA, Shumak KH, Buskard NA, et al. Comparison of plasma exchange with plasma infusion in the treatment of thrombotic thrombocytopenic purpura. Canadian Apheresis Study Group. N Engl J Med. 1991;325(6):393–7.CrossRefPubMed
23.
Zurück zum Zitat Cataland SR, Scully MA, Paskavitz J, et al. Evidence of persistent neurologic injury following thrombotic thrombocytopenic purpura. Am J Hematol. 2011;86(1):87–9.CrossRefPubMed Cataland SR, Scully MA, Paskavitz J, et al. Evidence of persistent neurologic injury following thrombotic thrombocytopenic purpura. Am J Hematol. 2011;86(1):87–9.CrossRefPubMed
24.
Zurück zum Zitat Falter T, Alber KJ, Scharrer I. Long term outcome and sequelae in patients after acute thrombotic thrombocytopenic purpura episodes. Haemostaseologie. 2013;33(2):113–20.CrossRef Falter T, Alber KJ, Scharrer I. Long term outcome and sequelae in patients after acute thrombotic thrombocytopenic purpura episodes. Haemostaseologie. 2013;33(2):113–20.CrossRef
25.
Zurück zum Zitat Deford CC, Reese JA, Schwartz LH, et al. Multiple major morbidities and increased mortality during long-term follow-up after recovery from thrombotic thrombocytopenic purpura. Blood. 2013;122(12):2023–9.CrossRefPubMedPubMedCentral Deford CC, Reese JA, Schwartz LH, et al. Multiple major morbidities and increased mortality during long-term follow-up after recovery from thrombotic thrombocytopenic purpura. Blood. 2013;122(12):2023–9.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Som S, Deford CC, Kaiser ML, et al. Decreasing frequency of plasma exchange complications in patients treated for thrombotic thrombocytopenic purpura-hemolytic uremic syndrome, 1996 to 2011. Transfusion. 2012;52(12):2525–32.CrossRefPubMedPubMedCentral Som S, Deford CC, Kaiser ML, et al. Decreasing frequency of plasma exchange complications in patients treated for thrombotic thrombocytopenic purpura-hemolytic uremic syndrome, 1996 to 2011. Transfusion. 2012;52(12):2525–32.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Knoebl P, Koder S, Schellongowski P, et al. Monitoring of ADAMTS13 in patients with thrombotic thrombocytopenic purpura: Prediction of response to therapy, risk of relapse, and long-term outcome. Blood. 2008;112:2291. Knoebl P, Koder S, Schellongowski P, et al. Monitoring of ADAMTS13 in patients with thrombotic thrombocytopenic purpura: Prediction of response to therapy, risk of relapse, and long-term outcome. Blood. 2008;112:2291.
28.
Zurück zum Zitat Froissart A, Buffet M, Veyradier A, et al. Efficacy and safety of first-line rituximab in severe, acquired thrombotic thrombocytopenic purpura with a suboptimal response to plasma exchange. Experience of the French Thrombotic Microangiopathies Reference Center. Crit Care Med. 2012;40(1):104–11.CrossRefPubMed Froissart A, Buffet M, Veyradier A, et al. Efficacy and safety of first-line rituximab in severe, acquired thrombotic thrombocytopenic purpura with a suboptimal response to plasma exchange. Experience of the French Thrombotic Microangiopathies Reference Center. Crit Care Med. 2012;40(1):104–11.CrossRefPubMed
29.
Zurück zum Zitat Scully M, McDonald V, Cavenagh J, et al. A phase 2 study of the safety and efficacy of rituximab with plasma exchange in acute acquired thrombotic thrombocytopenic purpura. Blood. 2011;118(7):1746–53.CrossRefPubMed Scully M, McDonald V, Cavenagh J, et al. A phase 2 study of the safety and efficacy of rituximab with plasma exchange in acute acquired thrombotic thrombocytopenic purpura. Blood. 2011;118(7):1746–53.CrossRefPubMed
30.
Zurück zum Zitat Bobbio-Pallavicini E, Porta C, Centurioni R, et al. Vincristine sulfate for the treatment of thrombotic thrombocytopenic purpura refractory to plasma-exchange. The Italian Cooperative Group for TTP. Eur J Haematol. 1994;52(4):222–6.CrossRefPubMed Bobbio-Pallavicini E, Porta C, Centurioni R, et al. Vincristine sulfate for the treatment of thrombotic thrombocytopenic purpura refractory to plasma-exchange. The Italian Cooperative Group for TTP. Eur J Haematol. 1994;52(4):222–6.CrossRefPubMed
31.
Zurück zum Zitat Beloncle F, Buffet M, Coindre JP, et al. Splenectomy and/or cyclophosphamide as salvage therapies in thrombotic thrombocytopenic purpura: The French TMA Reference Center experience. Transfusion. 2012;52(11):2436–44.CrossRefPubMed Beloncle F, Buffet M, Coindre JP, et al. Splenectomy and/or cyclophosphamide as salvage therapies in thrombotic thrombocytopenic purpura: The French TMA Reference Center experience. Transfusion. 2012;52(11):2436–44.CrossRefPubMed
33.
Zurück zum Zitat Peyvandi F, Scully M, Kremer Hovinga JA, et al. Caplacizumab for acquired thrombotic thrombocytopenic purpura. N Engl J Med. 2016;374:511–22.CrossRefPubMed Peyvandi F, Scully M, Kremer Hovinga JA, et al. Caplacizumab for acquired thrombotic thrombocytopenic purpura. N Engl J Med. 2016;374:511–22.CrossRefPubMed
34.
Zurück zum Zitat Peyvandi F, Scully M, Kremer Hovinga JA, Knöbl P, Cataland S, De Beuf K, Callewaert F, De Winter H, Zeldin RK. Caplacizumab reduces the frequency of major thromboembolic events, exacerbations and death in patients with acquired thrombotic thrombocytopenic purpura. J Thromb Haemost. 2017;15(7):1448–52.CrossRefPubMed Peyvandi F, Scully M, Kremer Hovinga JA, Knöbl P, Cataland S, De Beuf K, Callewaert F, De Winter H, Zeldin RK. Caplacizumab reduces the frequency of major thromboembolic events, exacerbations and death in patients with acquired thrombotic thrombocytopenic purpura. J Thromb Haemost. 2017;15(7):1448–52.CrossRefPubMed
35.
Zurück zum Zitat Scully M, Cataland SR, Peyvandi F, Coppo P, Knöbl P, Kremer Hovinga JA, et al. Results of the randomized, double-blind, placebo-controlled, phase 3 Hercules study of Caplacizumab in patients with acquired thrombotic thrombocytopenic purpura. Blood. 2017;130:LBA-1.CrossRef Scully M, Cataland SR, Peyvandi F, Coppo P, Knöbl P, Kremer Hovinga JA, et al. Results of the randomized, double-blind, placebo-controlled, phase 3 Hercules study of Caplacizumab in patients with acquired thrombotic thrombocytopenic purpura. Blood. 2017;130:LBA-1.CrossRef
36.
Zurück zum Zitat Schiviz A, Wuersch K, Piskernik C, et al. A new mouse model mimicking thrombotic thrombocytopenic purpura: correction of symptoms by recombinant human ADAMTS13. Blood. 2012;119(25):6128–35.CrossRefPubMed Schiviz A, Wuersch K, Piskernik C, et al. A new mouse model mimicking thrombotic thrombocytopenic purpura: correction of symptoms by recombinant human ADAMTS13. Blood. 2012;119(25):6128–35.CrossRefPubMed
37.
Zurück zum Zitat Scully M, Knöbl P, Kentouche K, Rice L, Windyga J, Schneppenheim R, et al. Recombinant ADAMTS-13: First-in-human pharmacokinetics and safety in congenital thrombotic thrombocytopenic purpura. Blood. 2017;130(19):2055–63.CrossRefPubMedPubMedCentral Scully M, Knöbl P, Kentouche K, Rice L, Windyga J, Schneppenheim R, et al. Recombinant ADAMTS-13: First-in-human pharmacokinetics and safety in congenital thrombotic thrombocytopenic purpura. Blood. 2017;130(19):2055–63.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Kelly GS. Clinical applications of N‑acetylcysteine. Altern Med Rev. 1998;3(2):114–27.PubMed Kelly GS. Clinical applications of N‑acetylcysteine. Altern Med Rev. 1998;3(2):114–27.PubMed
39.
Zurück zum Zitat Chen J, Reheman A, Gushiken FC, et al. N‑acetylcysteine reduces the size and activity of von Willebrand factor in human plasma and mice. J Clin Invest. 2011;121(2):593–603.CrossRefPubMedPubMedCentral Chen J, Reheman A, Gushiken FC, et al. N‑acetylcysteine reduces the size and activity of von Willebrand factor in human plasma and mice. J Clin Invest. 2011;121(2):593–603.CrossRefPubMedPubMedCentral
Metadaten
Titel
Thrombotic thrombocytopenic purpura
verfasst von
Prof. Dr. Paul Knöbl, MD
Publikationsdatum
17.08.2018
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 3/2018
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-018-0429-6

Weitere Artikel der Ausgabe 3/2018

memo - Magazine of European Medical Oncology 3/2018 Zur Ausgabe