Inhibitor Index in the Clot Waveform Analysis-Based Mixing Test Differentiates among Hemophilia A without and with Inhibitors, and Lupus Anticoagulant

Author(s):  
Naruto Shimonishi ◽  
Kenichi Ogiwara ◽  
Yukio Oda ◽  
Toshiki Kawabe ◽  
Mari Emmi ◽  
...  

Abstract Background The mixing test is used to identify the pathway to follow-up testing and is also useful for the investigation of lupus anticoagulant (LA) positivity. “To completely correct” indicates clotting factor deficiency, while “to not correct” indicates the presence of a clotting factor inhibitor including LA. “Index of circulation anticoagulant” and/or “percent correction” is used to interpret the results of mixing studies, but it does not accurately differentiate factor inhibitors from LA. Aim To precisely differentiate hemophilia A (HA), HA with inhibitor (HA-inh), and LA using the clot waveform analysis (CWA)-based mixing test. Methods Plasma samples from HA, LA, and HA-inh including acquired HA were incubated with normal plasma in 9:1, 1:1, and 1:9 mix ratios. From activated partial thromboplastin time CWA at 0-minute (immediately) and 12-minute incubation, the ratios of CWA parameters at 12 minutes/0 minute (inhibitor index) were assessed. Results The inhibitor index values of CWA parameters obtained using the mixing test in a 1:1 ratio demonstrated a significant difference between HA-inh and LA but could not differentiate LA from HA-inh completely. Plasmas used for the mixing tests in 9:1 and 1:9 ratios were able to fully distinguish between HA-inh (>0.5 BU/mL) and LA. These indices significantly correlated with inhibitor titer below 40 BU/mL (r > 0.90), possibly estimating FVIII inhibitor titer from the inhibitor index. Plasmas in HA and LA could be distinguished by mixing in a 1:1 ratio at 0 minute (immediately). Conclusion The inhibitor index from CWA-based mixing tests with a 12-minute incubation could differentiate among HA, HA-inh, and LA quickly.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1187-1187 ◽  
Author(s):  
Shannon L. Meeks ◽  
Courtney Cox ◽  
John F. Healey ◽  
Ernest T Parker ◽  
Bagirath Gangadharan ◽  
...  

Abstract Abstract 1187 A major complication in the treatment of severe hemophilia A is the development of anti-fVIII antibodies in approximately 30% of patients. In these patients and in animal models injection of fVIII by an intravenous route, which traditionally is considered a tolerogenic route of protein delivery, not only fails to induce tolerance but induces a brisk T- and B- cell immune response. The role of fVIII structure and function in the immunogenicity of fVIII remains unclear. In this study we tested four interrelated hypotheses: (i) FVIII is immunogenic due to its role in promoting production of thrombin, leading to downstream events that provide an immunogenic milieu. (ii) FVIII is immunogenic because it is exposed to the immune system in the context of active inflammation (i.e. at the site of a clot). (iii) Structural determinants intrinsic to the fVIII molecule are immunogenic. (iv) FVIII is protected from the immune system until it is released from its large carrier protein von Willebrand factor (VWF). To address these hypotheses we constructed wild-type B domain deleted fVIII (wt fVIII) and 2 structurally intact inactive fVIII molecules, R372A/R1689A fVIII and an A2 domain point mutant, V634M fVIII. R372A/R1689A fVIII is inactive due to substitutions at thrombin and factor X proteolytic activation sites. It is not released from VWF, and thus may not be present at the site of a clot. V634M fVIII undergoes normal thrombin cleavage but has specific procoagulant activity that is less than 1% of wt fVIII. The immunogenicity of the fVIII molecules was compared in 3 protocols. In a low dose protocol, fVIII deficient mice were injected with 6 weekly tail vein injections of 0.2 μg followed by 2 injections of 0.5 μg wt fVIII or R372A/R1689A fVIII. In a varying dose protocol, the immunogenicity of wt fVIII, R372A/R1689A fVIII, and V634M fVIII was determined in fVIII deficient mice following 4 weekly tail vein injections of 0.5 μg, 1.0 μg, 1.5 μg, or 2.0 μg fVIII per dose followed by 1 boost at twice the dose. Finally, the immunogenicity of wt fVIII, R372A/R1689A fVIII, and V634M fVIII was compared in fVIII/VWF deficient mice following 6 weekly injections at 0.6 μg followed by 2 boost injections at 1.5 μg. In the low dose protocol 68% of fVIII deficient mice injected with wt fVIII had positive ELISA titers with a median titer of 400 compared with 40% of those injected with R372A/R1689A fVIII with a median titer of 0. Mice injected with wt fVIII had a median inhibitor titer of 10 BU/ml compared with a median titer of 0. Although R372A/R1689A fVIII was statistically less immunogenic with lower ELISA and inhibitor titers (p=0.027 and 0.018, respectively, Mann-Whitney test) this may not be clinically relevant as 40% of the mice mounted an immune response. In the varying dose protocol, there was no difference in median ELISA fVIII inhibitor titers at any dosing level. At the 2.0 μg dose all mice except for 1 in the V634M fVIII cohort mounted an immune response. The median ELISA titers at 2.0 μg were1760 for wt fVIII, 447 for R372A/R1689A fVIII, and 1480 for V634M fVIII. The median inhibitor titers at the 2.0 μg dose were 310 BU/ml for wt fVIII, 103 BU/ml for R372A/R1689A fVIII, and 288 BU/ml for V634M fVIII. There was no significant difference between wt fVIII, R372A/R1689A fVIII and V634M fVIII in either ELISA or inhibitor titers (p=0.2 and p=0.35, respectively, Kruskal-Wallis test). In the fVIII/VWF deficient mouse protocol, 85% of mice had positive ELISA titers in the wt fVIII cohort compared with 79% for R372A/R1689A fVIII and 85% for V634M fVIII. The median ELISA titers were similar for each group at 354 for wt fVIII, 179 for R372A/R1689A fVIII, and 363 for V634M fVIII. Inhibitor titers were similar for each group with a median inhibitor titer of 107 BU/ml for wt fVIII, 46 BU/ml for R372A/R1689A fVIII, and 198 BU/ml for V634M fVIII. There was no significant difference between wt fVIII, R372A/R1689A fVIII and V634M fVIII in either ELISA or inhibitor titers (p=0.46 and p=0.32, respectively, Kruskal-Wallis test). In conclusion, there was no significant difference in the immunogenicity of wt fVIII and V634M fVIII in fVIII deficient mice. R372A/R1689A fVIII was slightly less immunogenic in fVIII deficient mice in 1 of 2 protocols tested. In the absence of VWF, wt fVIII, R372A/R1689A fVIII, and V634M fVIII were equally immunogenic. This suggests that the immunogenicity of fVIII is intrinsic to fVIII structure and not its cofactor activity, while VWF may have a small protective effect. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3988-3988
Author(s):  
Khaldoun J. Alkayed ◽  
Kandice Kottke-Marchant

Abstract 3988 Poster Board III-924 Abstract: Introduction The International Society of Thrombosis and Hemostasis (ISTH) criteria for the diagnosis of the lupus anticoagulant (LAC) include: Screening test that demonstrates the prolongation of a phospholipid-dependent (PL-D) clotting time; mixing test that confirms the presence of an inhibitor; the confirmation that the inhibitor is PL-D and exclusion of other coagulopathies. Test results that do not fulfill all the criteria are considered indeterminate. These indeterminate results are common (Kottke-Marchant et al. J Thromb Haemost. 2007; 5 Supplement 2: P-M-455), still there is no published data regarding clinical significance. Patients/methods This study investigated the prevalence of thrombotic events in an initial cohort of unselected patients (n=256) from one tertiary hospital in the United States, who were tested for LAC and other antiphospholipid (aPL) antibodies from a 2 month period in 2006. The laboratory results (PT/INR, aPTT, dilute Russell's viper venom time (DRVVT), STACLOT and platelet neutralization (PNP)) were evaluated. The profile included 3 separate PL -D assays (DRVVT confirm, STACLOT, PNP). Samples containing heparin (>0.1U/ml) were pre-treated with Hepadsorb. The LAC profile was considered indeterminate if PL test results were positive, but without a positive aPTT or DRVVT mixing study. The initial cohort included 83 patients with indeterminate results. From this group, 18 patients were excluded: Four due to incomplete data, 2 due to high heparin level (anti Xa>1.0 U/ml), 5 due to other prothrombotic etiologies and 7 with other positive aPL antibodies. For an assessment of thrombotic history, we performed retrospective chart reviews and tabulated all Sapporo clinical features, malignancy and auto-immune disorders within 5 years before and 2 years after the index laboratory testing. Events that did not fulfill diagnostic criteria for thrombosis, ischemic events or obstetrical complications were excluded. The final analysis sample included 65 patients with indeterminate LAC, 106 with negative and 27 with positive LAC. Results The final indeterminate LAC cohort included 65 patients, with mean follow-up of 18 months. Malignancy was present in 29% and autoimmune disease in 25% of patients. The most common thrombotic events were deep vein thrombosis (DVT) (28%), cerebral ischemic stroke (14%) and pulmonary embolism (14%). When compared to those with negative tests, indeterminate group patients were more likely males, relatively older, and more likely to have DVT, superficial thrombosis (ST) or myocardial infarction (MI) (P= 0.049, 0.021, 0.044, 0.005 and 0.045 respectively). Concurrent coumadin (warfarin) therapy was more prevalent in the indeterminate group, but it did not reach statistical significance (p=0.15). There was no statistical significant difference in the prevalence of cancer or autoimmune disease (P=0.19 and 0.48 respectively). In the multivariate analysis model none of the previous variables reached any statistical significance between the two groups. When compared the above clinical variables between indeterminate results and positive LAC results groups from the same cohort, we failed to show any major statistically significant differences. We noticed very poor retesting rate in the indeterminate group during the follow up period of 2 years (15% only). Conclusions Indeterminate results are common among patients referred for LAC testing. When compared to those with negative results, patients with indeterminate results are more likely to have a history of DVT, superficial thrombosis or MI, but none of the clinical variables reached statistical significance in a multivariate model. On the other hand, patients in the indeterminate group shared demographic and clinical profiles with those in the positive results group. This further highlights the need to study the clinical significance of indeterminate LAC results in a prospective study. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3382-3382
Author(s):  
Junka Haku ◽  
Kenichi Ogiwara ◽  
Tomoko Matsumoto ◽  
Koji Yada ◽  
Midori Shima ◽  
...  

Abstract Abstract 3382 There is no optimal assay to monitor the hemostatic effect of bypassing therapy for hemophilia A patients (HA) with inhibitor. Clot waveform analysis (CWA) is a convenient method of assessing global clotting function based on the continuous monitoring of light transmittance or absorbance during routine coagulation tests such as aPTT. We attempted to optimize the aPTT based CWA for hemostatic monitoring during the bypassing therapy in patients of HA with inhibitor. An automated laboratory system, MDA-II® (Trinity Biotech) was used for CWA. Clot waveform was plotted from transmittance change during the clot formation. By the first and second derivation of the waveform, parameters such as clotting time, the maximum velocity (|min1|) and acceleration (|min2|) were calculated. Three trigger reagents were used; [R1] ellagic acid (E) and phospholipid (PL) that is used as an aPTT reagent, [R2] tissue factor (TF, Innovin®) and PL as a 17,000-diluted PT reagent, [R3] the mixed condition of E, TF and PL, based on our recent report on thrombin generation test optimized for HA (Matsumoto, IJH, 2009). Results were expressed as a percentage (% of control) relative to control pooled plasma. The great difference in all parameters between factor (F)VIII-deficient and control plasma resulted in R1>R3>>R2 in order (|min2| in R1, R2, R3 were 13, 119, 21% of control, respectively). Since significant difference between both plasma was not observed, we excluded R2. By the addition of clinically therapeutic concentration of recombinant FVIIa (rFVIIa, NovoSeven®) (25 nM), parameters in R3 were more improved than R1 (|min2| in R1, R3 were 18, 47% of control, respectively). Similar results were observed by the addition of activated prothrombin complex concentrates (APCC, FEIBA®) (1 U/ml) (|min2| in R1, R3 was 32, 71% of control, respectively). According to these in vitro experiments, R3 (E/TF/PL) was regarded as most optimal reagent. Next, in order to confirm the usefulness of the R3-CWA system in vivo, six patients with HA with inhibitor to whom rFVIIa (n=6, 37 (12–59) BU/ml, 105 (91–175) ƒÊg/kg dosage) or APCC (n=3, 25 (12–49) BU/ml, 91 (91–100) U/kg dosage) was administrated, were evaluated. All patients showed the clinical hemostatic efficacy by each bypassing agent. As shown in Table 2 and Fig. 1, the hemostatic effects by bypassing agents were confirmed by improvement in all R3-CWA parameters. APTT based CWA system should be promising method for quantitative monitoring during the bypassing therapy with routine automated clotting machine and only with the modified reagents such as well-balanced mixtures of E, TF and PL. Table 1. Parameters of CWA using three reagents in FVIII-deficient plasma with bypassing agents in vitro Parameter Reagent R1 R2 R3 Clotting time % of control     FVIII-def plasma 372 86 147     +rFVIIa (25 nM) 271 – 100     +APCC (1 U/ml) 122 – 69 |min1| % of control     FVIII-def plasma 16 90 33     +rFVIIa (25 nM) 20 – 54     +APCC (1 U/ml) 27 – 69 |min2| % of control     FVIII-def plasma 13 119 21     +rFVIIa (25 nM) 18 – 47     +APCC (1 U/ml) 32 – 71 Values are shown as median of triplicate. Table 2. Effect of bypassing agents in vivo on parameters of R3-triggered CWA Parameter rFVIIa (n=6) APCC (n=3) Pre Post Pre Post % of control % of control Clotting time 202 103 135 99 (129–268) (91–122) (127–159) (89–104) |min1| 37 56 41 60 (19–48) (40–78) (30–42) (46–75) |min2| 19 53 23 41 (13–32) (44–75) (16–24) (34–60) Values are shown as median (range). Fig. 1 Representative data of R3-triggered CWA in a case of HA with inhibitor on bypassing therapy using rFVIIa (dotted line) or APCC (solid line). Fig. 1. Representative data of R3-triggered CWA in a case of HA with inhibitor on bypassing therapy using rFVIIa (dotted line) or APCC (solid line). Disclosures: Shima: Chugai Pharmaceutical Co., Ltd.: Consultancy, Honoraria, Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 18-19
Author(s):  
Evan C. Chen ◽  
William J. Gibson ◽  
Paula Temoczko ◽  
Nathan T. Connell ◽  
Robert Handin ◽  
...  

Background Acquired hemophilia A is a rare bleeding disorder caused by autoantibodies that inhibit coagulation factor VIII (FVIII). The disorder is understudied given its rarity and there are no randomized prospective trials to guide therapy. In practice, treatment involves attaining hemostasis and eliminating the FVIII inhibitor, typically with high-dose steroids (1 mg/kg daily) and either cyclophosphamide or rituximab. However, current approaches carry risk of significant adverse events and delayed or inadequate responses. Emicizumab is a bispecific antibody that targets coagulation factors IXa and X to recapitulate the function of endogenous FVIII. We present a case series of patients with acquired hemophilia A who were successfully treated with a regimen consisting of rituximab and emicizumab. Methods We identified patients >18 years who were diagnosed with acquired hemophilia A and received treatment with rituximab and emicizumab at Brigham and Women's Hospital between 2019 and 2020. We performed a retrospective chart review. Data collected included the patients' clinical presentation, laboratory studies (including coagulation testing, FVIII activity, and FVIII inhibitor titer), and treatments received (including systemic therapies, recombinant factor VIIa [rFVIIa], red blood cell [RBC] transfusions, and vascular embolization). We recorded the time to normalization of the activated partial thromboplastin time (aPTT) and chromogenic FVIII activity following emicizumab and rituximab initiation, respectively. Activated prothrombin complex concentrate was avoided given the use of emicizumab. Results We identified 8 patients with acquired hemophilia A who received treatment with emicizumab and rituximab. The median patient age was 81 (range 47-93). All patients sought medical attention for extensive ecchymoses or bleeding and were found to have prolonged aPTT leading to FVIII inhibitor identification (Table 1). The median inhibitor titer was 18 Bethesda units (range 9.2-107.5). Patients concurrently received 4 weekly doses of rituximab 375mg/m2 and 4 weekly loading doses of emicizumab 3mg/kg. Patient (Pt) #1 continued emicizumab 3mg/kg every two weeks to complete three months of treatment. Pts #2, #3, and #8 received high-dose prednisone (1mg/kg) at the start of treatment for a range of 10-14 days. Pt #8 received 7 additional days of prednisone for an initial aPTT of 60.7 seconds before starting emicizumab and rituximab; she had no clinical response when treated with prednisone alone. Pts #2, #5, and #7 required vascular embolization. 7 patients (Pts #2 through #8) had aPTT retested within 1 week of starting emicizumab, and the aPTT for these patients normalized within 10 days of starting emicizumab (i.e. after only 1-2 doses; Figure 1). Except for Pt #5 who had recurrent hematuria from a persistent anatomic bladder defect that eventually required prostatic artery embolization, patients did not require rFVIIa or RBC transfusions for more than 7 days after starting emicizumab. Except for Pt #5 who required 28 doses of rFVIIa and 3 units of RBC transfusions after starting emicizumab, the median number of rFVIIa doses and RBC units given to the remaining 7 patients was zero (range 0-6 doses) and zero (range 0-4 units), respectively. Pts #2 and #3 had chromogenic FVIII levels obtained >30 days after starting rituximab with improvement in FVIII activity to 29% (day 71) and 86% (day 91), respectively. During a median follow-up of 102 days, no patients experienced recurrent bleeding. However, Pt #3 exhibited a slowly increasing aPTT that reached 46.3 seconds on day 233 of follow-up without symptoms; further diagnostic testing is pending. Conclusion Our case series demonstrates that the combination of rituximab and emicizumab can be an effective and safe regimen for the treatment of acquired hemophilia A. No thrombotic events or thrombotic microangiopathy occurred. Treatment with weekly emicizumab led to aPTT normalization after 1-2 doses and facilitated hemostasis, as reflected by a median usage of zero rFVIIA doses and zero RBC transfusions after starting emicizumab when excluding one patient with hematuria from an anatomic defect. This compares favorably to historical reports. While no patient has had recurrent bleeding, additional chromogenic FVIII activity testing for patients is needed to confirm long-term normalization of FVIII activity. Disclosures Gibson: Ampressa therapeutics: Current equity holder in private company; nference: Consultancy, Current equity holder in private company; ImmPACT-Bio: Consultancy; Boston Clinical Research Institute: Consultancy. Parnes:Bayer: Consultancy; I-Mab: Consultancy; Sunovion: Consultancy; UniQure: Consultancy; Sigilon: Consultancy; Shire/Takeda: Consultancy, Research Funding; Genentech: Research Funding; Geron: Current equity holder in publicly-traded company. OffLabel Disclosure: Emicizumab is used off-label in our case series for the treatment of acquired hemophilia A.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1122-1122
Author(s):  
Zekun Li ◽  
Zhenping Chen ◽  
Xiaoling Cheng ◽  
Xinyi Wu ◽  
Li Gang ◽  
...  

Background: Low-dose immune tolerance induction (ITI) +/- immunosuppression as a practical ITI strategy in China showed a relatively satisfactory success rate and economic advantages in pilot study. However, the outcome still needs to be verified by larger cohort. Aim: To report the efficacy of this low-dose ITI +/- immunosuppression strategy in hemophilia A children ≥ 10 BU. Methods: This was a single center, prospective study in 53 hemophilia A subjects from Sep 2016 to Apr 2019. All subjects having ≥ 10 BU receiving ~50IU/kg FVIII every other day using domestic intermediate purity pdFVIII/VWF products, either alone or in combination with rituximab and prednisone judging by inhibitors and ITI response. Results: Finally, 46 subjects received this strategy at a median of 3.2 (IQR, 2.3-6.5) years old, their pre-ITI inhibitor titer was median 30.0 (range, 10.1-416) BU. Analysis at median 15.1 (range 3.0-34.4) months follow-up, success (inhibitor <0.6BU) was achieved in 32 (69.6%) subjects, partial success (inhibitor <5BU but >0.6BU) in 11 (23.9%) subjects, and failure in 5 (10.9%) subjects. Between subjects administered ITI-alone and ITI- immunosuppression, no significant difference was observed in time to success (median 8.5; IQR 6.7-11.7 vs 10.2; IQR 5.1-25.1, P=0.164). The mean monthly bleeding rate on ITI was 0.49 which declined 59.3% compared with pre-ITI period. Subjects administered ITI-immunosuppression (0.54 ± 0.46) was higher than ITI-alone (0.42 ± 0.69) although with no significantly difference (P=0.089). Seven (21.9%) subjects experienced inhibitor recurrence, 4 subjects treated with ITI-alone, 3 with ITI-immunosuppression. Recurrence occurred at a median of 4.8 (range, 2.8-10.8) months after successful ITI with inhibitor titer transiently rising to median 0.7 (range, 0.7-1.5) BU. Conclusion: This low-dose ITI +/- immunosuppression therapy in subjects with pre-ITI inhibitor ≥ 10 BU showed a success rate similar to other high/intermediate-dose regimen for the whole inhibitor patients. The subjects treated with ITI-immunosuppression did not showed higher recurrence at present, while a longer time follow-up is still needed. Disclosures Poon: Roche: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bioverativ/Sanofi: Consultancy, Membership on an entity's Board of Directors or advisory committees; World Federation of Hemophilia: Other: Not-for-profit organization affiliation: volunteer ; Novo Nordisk: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Participation in sponsored research; CSL-Behring: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Grant Funding; Bayer: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Grant Funding; Takeda/Shire: Consultancy, Membership on an entity's Board of Directors or advisory committees; Octapharma: Consultancy, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3091-3091
Author(s):  
Karin van Dijk ◽  
Johanna G. van der Bom ◽  
Eveline P. Mauser-Bunschoten ◽  
Goris Roosendaal ◽  
Peter J. Lenting ◽  
...  

Abstract Introduction Patients with severe hemophilia A have considerably different factor VIII half-lives. Whether this is associated with clinical characteristics has not been reported. The aim of this study was to describe the effect of half-life on the clinical characteristics of patients with severe hemophilia. Patients and Methods Patients were selected from a single-centre cohort of 214 patients with severe hemophilia, born between 1944 and 1995. To improve efficiency we measured factor VIII half-life in the patients with the most severe and the mildest clinical phenotypes of severe hemophilia. Patients were selected according to age at first joint bleed, annual joint bleed frequency, clotting factor consumption and radiological Pettersson scores. A first blood sample was taken after a period of 72 hours in which the patient did not use factor VIII. After infusion with 50 IU factor VIII/kg, blood was collected at 15, 30 minutes and 1, 3, 5, 24, 30, 48 and 60 hours. From 1972 onwards, data on joint bleed frequency, clotting factor use and age at first joint bleed were collected from the patients’ files. Pettersson scores were performed at five-year intervals. For calculations of annual clotting factor use (IU/kg/yr) and number of joint bleeds per year, the last 5 years of follow-up were used. Linear regression analysis was used to assess the relation between clinical characteristics and factor VIII half-life. Results Factor VIII half-life was measured in 42 patients and ranged from 7.4–20.4 hours, with a median of 11.8 hours. One hour increase in factor VIII half life was associated with a decrease of 96 (SD 45) IU clotting factor use per kg per year (p&lt;0.05). Joint bleed frequency was similar in patients with a shorter and a longer factor VIII half-life. Median number of joint bleeds was 2.9 per year (interquartile range (IQR) 1.1–4.4) in patients with a factor VIII half-life shorter than 12 hours and 2.6 per year (IQR 1.0–4.8) in patients with a factor VIII half-life longer than 12 hours (p=0.84). Patients with a factor VIII half-life shorter than 12 hours had a median Pettersson score of 52 points (IQR 12–61) and patients with a factor VIII half-life longer than 12 hours had a median Pettersson score of 29 points (IQR 16–52; p=0.90). Conclusion: Patients with a shorter factor VIII half-life need more clotting factor to prevent joint bleeds and subsequent arthropathy than patients with a longer factor VIII half-life.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4781-4781 ◽  
Author(s):  
Mauricio A Alzate ◽  
Susana S Meschengieser ◽  
Alicia Blanco ◽  
Silvia Grosso ◽  
María A Lazzari ◽  
...  

Introduction Acquired hemophilia A is a rare and serious autoimmune disease. Morbidity and mortality are associated with advanced age, comorbidities, toxicity of treatment and bleeding severity. Treatment goals are control of the bleeding and eradication of the inhibitor, while treating the underlying condition if it is present. Objective To describe the baseline characteristics of acquired hemophilia A patients and to assess the response to treatment. Patients and Methods Between November 1991 and April 2013, 27 patients were diagnosed with acquired hemophilia A (mean age 59, range 21-86; 18 women - 66%) in the Departamento de Hemostasia y Trombosis. Five patients were lost from follow-up. APTT mixing studies with normal plasma (1:1) and time-temperature dependent effect on the APTT were performed for a-FVIII diagnosis. Whenever possible, inhibitor activity was titrated by Bethesda method at diagnosis (BU/mL). Medical records were reviewed to evaluate the initial symptoms, underlying diseases, treatments and outcome. Results The mean follow-up was 86 weeks (range 1-640). Underlying etiologies included: idiopathic 70.4%, postpartum 14.8%, malignancy 11.1%, autoimmune disease 3.7%. All patients had bleeding at diagnosis. The most frequent sites of bleeding were: muscular 32%, soft tissue 18%, urinary tract 9%, gastrointestinal tract 6%; being from multiple sites in 9%. At diagnosis, the mean value for FVIII was 6% (range 1-40), and inhibitor titer 220 BU/mL (range 2.2-1173). Initial therapeutic scheme included glucocorticoids in 97% of the patients, 13 in monotherapy (mean age 53 years ± 19), 13 with cyclophosphamide (63 years ± 18) (p= ns), and human immunoglobulin in 1 patient. This last patient died after 1 week of diagnosis due to uncontrolled gastrointestinal bleeding (previous to the era of rVIIa). As a second-line therapy, rituximab was used in 3 patients. Sixty-three (63%) patients achieved complete response (CR) (inhibitor titer < 0.6 BU/mL without bleeding episodes), and 23% achieved partial response (PR) (reduction in inhibitor titer > 50% without bleeding episodes), without differences between monotherapy or combined. Overall, women responded more frequently than men (93.3% vs. 71.4%, p= ns). All patients that received rituximab achieved CR. Conclusions In this study, the overall response rate was higher than 80%. In most cases, the disease has a prolonged course like other autoimmune diseases, with remissions and relapses. Disclosures: No relevant conflicts of interest to declare.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e17006-e17006
Author(s):  
Giacinto La Verde ◽  
Antonella Ferrari ◽  
Vincenzo Ziparo ◽  
Virginia Naso ◽  
Maria Paola Bianchi ◽  
...  

e17006 Background: Acquired hemophilia A is caused by the development of factor (F)VIII autoantibodies and results in more serious haemorrhagic symptoms than in congenital severe HA. About 50% of cases, not identified as idiopathic, are related to autoimmune diseases, viral infections, pregnancy and also neoplasms. To treat bleeds recombinant factor VIIa and activated prothrombin complex concentrate are equally efficacious while immunosuppression with steroids alone or combined with cytotoxic agents should be started as soon as the diagnosis is made. Methods: In February 2010 a woman of 58 years with acquired hemophilia A, previously treated in 1978 with cyclophosphamide and steroids without clinical benefits, was admitted to the Hematology Department of Sant’Andrea Hospital in Rome for a recent episode of spontaneous massive enterorrhagia. The diagnostic exams revealed a colorectal carcinoma (moderately differentiated G3) associated with papillary urothelial neoplasia with low malignant potential. A coagulation panel showed an aPTT ratio of 3.75 (normal range 0.8-1.2), INR 1.03 (normal range 0.9-1.2), fibrinogen 327 mg/dL (normal range 238-500), FVIII activity level of 3% (normal reference 50-150%) and FVIII inhibitor titer > 100 Bethesda Units. Results: The patient was treated with left hemicolectomy and removal of the bladder lesion associated with chemotherapy XELOX (oxaliplatin 130 mg/sm day 1 q21, capecitabine 1000 mg/sm orally day 1-14 q21). From day -1 to day +15 post surgery, the patient was treated with intravenous recombinant human activated FVII at standard dose (NovoSeven®, 90 mcg/kg every 4 hours). Our treatment avoided both intra and post-operative surgical bleeding complications, while not changing the titer of the inhibitor nor by normalizing the aPTT ratio. Actually the patient is in good clinical conditions with no further hemorrhagic episodes, although the FVIII inhibitor titer still remains high. Conclusions: We have described this case to emphasize that the presence of acquired hemophilia A in cancer patients, thanks to its specific prophylactic treatment, cannot be a limit in performing routine diagnostic and therapeutic procedures, especially surgical ones.


2020 ◽  
Vol 26 ◽  
pp. 107602962097881
Author(s):  
Daiki Shimomura ◽  
Tomoko Matsumoto ◽  
Kana Sugimoto ◽  
Tokio Takata ◽  
Aya Kouno ◽  
...  

Clot waveform analysis based on activated partial thromboplastin time (aPTT) is reported to be a useful assay. We attempted to find beneficial parameters with the first-derivative curve. We examined 106 plasma samples with prolonged aPTT and analyzed the first-derivative curve statistically by dividing it into 6 groups (Lupus anticoagulant, Heparin, Direct oral anticoagulants, Factor VIII inhibitor, Hepatic dysfunctions and Factor deficiency). We obtained 7 coordinates for parameter measurement by analyzing the first-derivative curve and set 20 parameters including the velocity axis, the time axis, and area parameters. The distribution was checked by extracting each parameter that showed the most significant difference in the 6 groups. As a result, it was revealed that we could classify aPTT prolongation by using a combination of 3 parameters, the initial-to-peak gradient, the ratio initial-to-intermediate velocity/intermediate-to-peak velocity, and the initial-to-peak area size. We constructed a flowchart combining these 3 parameters and were able to discriminate 75% of the specimens. These parameters derived from the first-derivative curve of clot waveform analysis are useful tools to discriminate aPTT prolongation.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 611-611 ◽  
Author(s):  
Giang N. Nguyen ◽  
John K. Everett ◽  
Hayley Raymond ◽  
Samita Kafle ◽  
Elizabeth P. Merricks ◽  
...  

Hemophilia is an X-linked bleeding disorder caused by a deficiency in clotting factor VIII (FVIII)(hemophilia A, HA) or factor IX (FIX)(hemophilia B, HB). While early clinical trials of AAV delivery of FIX for HB have demonstrated stable FIX expression for &gt;8 years, an ongoing clinical trial of AAV-FVIII delivery for HA achieved high levels of transgene expression that unexpectedly declined after 1 year. Here we describe preclinical studies of AAV-canine FVIII (cFVIII) delivery in nine HA dogs with sustained FVIII expression for the duration of the study, as long as 10 years. FVIII was delivered using two delivery approaches: (1) co-administration of two AAV vectors encoding separate cFVIII heavy and light chains driven by the thyroxine binding globulin (TBG) promoter (Two chain approach)(TC) (n=5) at two AAV doses (2.5 x 1013vg/kg; F24, Woodstock, J60) and (1.2 x 1013vg/kg; Linus, H19) or (2) delivery of cFVIII as a single chain driven by the human alpha-1 anti-trypsin (hAAT) promoter (Single chain approach)(SC)(n=4) at two AAV doses (4 x 1013 vg/kg; M50, M06) and (2 x 1013vg/kg; M66, L51) (Sabatino 2011). We demonstrated that both strategies were efficacious; preventing &gt;95% of spontaneous bleeding episodes without toxicity. We now report the long-term follow-up of between 2.2 and 10.1 years for these treated dogs. Dose-dependent cFVIII:C (Coatest SP4 FVIII) was observed. At the final time point, the cFVIII:C was 2.7% (F24), 7.1% (Woodstock), 4.5% (J60), 11.3% (Linus) and 2.5% (H19) for TC dogs. For the SC dogs, the cFVIII:C was 9.4% (M06), 10.3% (M50), 1.9% (L51) and 3.7% (M66). Stable FVIII expression was maintained for seven of the dogs over the course of the study. Two dogs (Linus, M50) had a gradual increase in FVIII:C that began about three years after vector administration and continued for an additional seven years (Linus) and four years (M50), until the termination of the study. Liver function tests, serum alpha-fetoprotein concentrations, fibrinogen levels as well as liver pathology did not suggest altered liver function or tumor development in Linus and M50 compared to the other dogs. Clinically, there was no evidence of malignancy and no tumors were detected at the time of necropsy in any dog. One of the safety concerns for AAV-mediated gene therapy approaches is the potential for AAV integration events to be genotoxic and lead to tumorigenesis. While recombinant AAV primarily remains as an episome, integration events have been observed in mouse models and hepatocellular carcinoma has been observed after neonatal delivery of AAV vectors. In addition, the increase in FVIII expression in Linus and M50 prompted us to investigate integration and clonal expansion as a potential mechanism for these observations. Vector copy number (VCN) analysis was performed on liver samples (5-29 per dog, n=8 dogs) by Q-PCR and detected DNA copy numbers between 0.0 and 7.8 per diploid genome (Fig 1A). We performed integration target site analysis on liver samples (n=3/dog) from six of the AAV-treated HA dogs and naïve HA dogs (n=2) by ligation-mediated PCR, Ilumina paired-end sequencing and analysis using the custom software pipeline, AAVenger. Analysis of the 20 samples identified &gt;2,000 unique AAV integration events (IE). There was a correlation between the DNA copy number and the number of integration events detected. Clonal abundances were estimated by counting the unique genome breaks associated with integration positions, which showed that the maximum clonal abundance ranged from 1 to 138. The integration events were distributed across the canine genome. Clonal expansions were observed with integration near genes previously associated with growth control and transformation in humans (Fig 1B) with the most abundant clones located in DLEU2L (Linus), PEBP4 (J60) and EGR3 (M50). Integration events in EGR3, EGR2, CCND1, LTO1 and ZNF365 were detected in multiple dogs. Validation of integration sites in the most abundant clones was performed using targeted PCR to isolate junction fragments followed by Sanger sequencing. While AAV integration and clonal expansion was observed, the dogs had no evidence for tumorigenesis and it is not clear if the increase in FVIII expression is associated with the clonal expansions detected. Overall, these studies demonstrate long-term sustained FVIII expression for up to 10 years with clonal expansion, but without clinical adverse events after AAV-mediated gene therapy for hemophilia. Disclosures Sabatino: Spark Therapeutics: Patents & Royalties.


Sign in / Sign up

Export Citation Format

Share Document