scholarly journals Serum TNF-α Level as a Possible Predictor of Inhibitor Levels in Severe Hemophilia A

2021 ◽  
Vol 2021 ◽  
pp. 1-10
Author(s):  
Susi Susanah ◽  
Harry Raspati ◽  
Nur Melani Sari ◽  
Lulu Eva Rakhmilla ◽  
Yunia Sribudiani ◽  
...  

Background. The development of factor VIII (FVIII) inhibitor in patients with hemophilia A (PWHA) is a great challenge for hemophilia care. Both genetic and environmental factors led to complications in PWHA. The development of inhibitory antibodies is usually induced by the immune response. Tumor necrosis factor α (TNF-α), one of the cytokines, might contribute to its polymorphism. In this study, we investigated the clinical factors, level of serum TNF-α, and polymorphism of c . − 308 G > A   TNF − α gene in inhibitor development in severe PWHA. Methods. A cross-sectional study was conducted among all PWHA in West Java province. The clinical parameters, FVIII, FVIII inhibitor, and serum TNF-α level were assessed. The genotyping of − 380 G > A TNF-α gene polymorphism was performed using polymerase chain reaction and Sanger sequencing. Results. Among the 258 PWHA, 216 (83.7%) were identified as severe PWHA. The FVIII inhibitor was identified in 90/216 (41.6%) of severe PWHA, consisting of 45 high-titer inhibitors (HTI) and 45 low-titer inhibitors (LTI). There was a significant correlation between serum TNF-α level and the development of HTI ( p = 0.043 ). The cutoff point of serum TNF-α level, which can be used to differentiate between HTI and LTI, was 11.45 pg/mL. The frequency of FVIII replacement therapy was significant only in HTI of severe PWHA regarding serum TNF-α level ( p = 0.028 ). There is no correlation between polymorphisms of − 380 G > A TNF-α gene and inhibitor development ( p = 0.645 ). Conclusions. The prevalence of FVIII inhibitor in severe PWHA in West Java, Indonesia, was 41.6%. The frequency of replacement therapy is a risk factor for inhibitor development. Serum TNF-α level might be used to differentiate between high and low inhibitor levels in severe hemophilia A, and this might support decision making regarding treatment options for inhibitor in severe hemophilia A.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 330-330
Author(s):  
Flora Peyvandi ◽  
Antonino Cannavò ◽  
Isabella Garagiola ◽  
Roberta Palla ◽  
Frits Rosendaal ◽  
...  

Abstract BACKGROUND: Administration of factor VIII (FVIII) concentrates to patients with severe hemophilia A has nearly normalized their life expectancy; however, a substantial number of previously untreated patients (PUPs) develop neutralizing alloantibodies (inhibitors) against FVIII that complicate the benefits of treatment and make the management of these patients difficult and costly. Recently a randomized clinical trial (SIPPET) showed that the risk of inhibitor development is nearly doubled in previously untreated and minimally treated patients with severe hemophilia A treated with recombinant products (rFVIII) compared with plasma-derived (pdFVIII) products containing von Willebrand factor (VWF). AIM: The aim of this post-hoc analysis of the SIPPET study was to assess the early risk of inhibitor development every 5 exposure days (EDs) and to see whether or not there was a difference between the two arms of plasma-derived FVIII and recombinant FVIII products over the course of time of FVIII exposure. METHODS: 251 children were enrolled in the SIPPET study and randomized to receive a single pd or rFVIII (4 different brands of each). The outcomes were any FVIII inhibitor levels ≥ 0.4 Bethesda Units (BU) and high-titer inhibitors (peak levels > 5 BU). Patients were tested for inhibitors before entry and at regular intervals during 50 EDs, 3 years or the development of an inhibitor. Patients who had not reached 50 EDs by the time of study termination were censored. To study the risk over time, survival analysis by Kaplan-Meier and Cox regression were repeated for every 5 EDs to assess the association of source of FVIII with inhibitor development over time. RESULTS: 76 out of 251 patients developed an inhibitor and of those 50 were high-titer; all occurred before 40 EDs. Over the complete observation period, users of rFVIII products had a 87% higher rate of inhibitor development than users of pdFVIII (hazard ratio (HR) 1.87; 95% confidence interval (CI95) 1.17-2.96). The hazard ratio was particularly high during the first 5 EDs, both for all (HR 3.14, CI95% 1.01-9.74) and high-titer inhibitor development (HR 4.19, CI95% 1.18-14.8). After the first 5 EDs, the difference between two arms was less pronounced and quickly dropped to the overall difference of 1.87. Table 1 shows the cumulative incidence per time interval for each of the product classes. CONCLUSION : The risk of inhibitor development during replacement therapy occurs more early (0-5 EDs) in patients treated with rFVIII than in those treated with pdFVIII, and remains high until 15 EDs. For pdFVIII, the highest risk came later (6-10 EDs) and this peak lasted shorter. The results of this analysis shows the relevance of the early exposure days for inhibitor development. Table 1. Table 1. Disclosures Peyvandi: Alexion: Other: research funding paid to Luigi Villa Foundation, Research Funding; Novo Nordisk: Other: research funding paid to Luigi Villa Foundation, Research Funding, Speakers Bureau; Bayer: Speakers Bureau; Kedrion Biopharma: Consultancy, Other: research funding paid to Luigi Villa Foundation, Research Funding; LFB: Consultancy; CSL Behring: Speakers Bureau; Biotest: Other: research funding paid to Luigi Villa Foundation, Research Funding, Speakers Bureau; Octapharma: Consultancy; Ablynx: Membership on an entity's Board of Directors or advisory committees, Other: research funding paid to Luigi Villa Foundation, Research Funding; SOBI: Speakers Bureau; Grifols: Speakers Bureau. Palla:Pfizer: Other: travel support . Mannucci:Bayer: Speakers Bureau; Grifols: Speakers Bureau; Kedrion: Speakers Bureau; NovoNordisk: Speakers Bureau.


Blood ◽  
2012 ◽  
Vol 119 (12) ◽  
pp. 2922-2934 ◽  
Author(s):  
Samantha C. Gouw ◽  
H. Marijke van den Berg ◽  
Johannes Oldenburg ◽  
Jan Astermark ◽  
Philip G. de Groot ◽  
...  

Abstract This systematic review was designed to provide more precise effect estimates of inhibitor development for the various types of F8 gene mutations in patients with severe hemophilia A. The primary outcome was inhibitor development and the secondary outcome was high-titer-inhibitor development. A systematic literature search was performed to include cohort studies published in peer-reviewed journals with data on inhibitor incidences in the various F8 gene mutation types and a mutation detection rate of at least 80%. Pooled odds ratios (ORs) of inhibitor development for different types of F8 gene mutations were calculated with intron 22 inversion as the reference. Data were included from 30 studies on 5383 patients, including 1029 inhibitor patients. The inhibitor risk in large deletions and nonsense mutations was higher than in intron 22 inversions (pooled OR = 3.6, 95% confidence interval [95% CI], 2.3-5.7 and OR = 1.4, 95% CI, 1.1-1.8, respectively), the risk in intron 1 inversions and splice-site mutations was equal (pooled OR = 0.9; 95% CI, 0.6-1.5 and OR = 1.0; 95% CI, 0.6-1.5), and the risk in small deletions/insertions and missense mutations was lower (pooled OR = 0.5; 95% CI, 0.4-0.6 and OR = 0.3; 95% CI, 0.2-0.4, respectively). The relative risks for developing high titer inhibitors were similar.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 218-218
Author(s):  
Jay N. Lozier ◽  
Idan Menashe ◽  
James J. Goedert ◽  
Philip S. Rosenberg

Abstract Abstract 218 Introduction: Inhibitor antibodies to FVIII develop in ∼20% of patients with severe hemophilia A, and are the most important adverse events associated with FVIII replacement therapy. Mutations in the FVIII gene are the major determinant of inhibitor risk, but variations in immune response genes, such as IL10 or TNFα, (Astermark, et al, 2006a, 2006b) may also confer risk of inhibitor development and variations in the CTLA4 gene may protect against inhibitors (Astermark et al, 2006c). Specific Objective: Using a case-control study design we sought to confirm and extend previous observations of inhibitor risk modifying genes seen in family studies. Candidate genes were selected based on previous clinical or animal studies and included cytokines involved in the TH1/TH2 immune responses, and genes thought to decrease demand for FVIII therapy (e.g., FV Leiden and prothrombin 20210 polymorphisms). Materials and Methods: We used the CEU population data in the HapMap database to select haplotype tagging SNPs in IL1α/β, IL2, IL4, IL6, IL10, IL12A/B, IL13, IL17A, IL22, TNFα, CTLA4, interferon-γ, TGFβ, zinc α-2 glycoprotein I, IL1RN, and the FVIII gene, as well as the FV Leiden and prothrombin 20210 gene polymorphisms. A total of 366 tagging SNPs were selected with a goal of covering the entire coding and regulatory regions of the genes (spanning from 20kb 5' to 10kb 3' of the gene's coding sequence), resulting in 100% coverage of potential haplotypes (r2 = 1.0). DNA was purified from 915 Caucasian, severe hemophilia A patients (282 inhibitor cases and 633 non-inhibitor controls) who participated in the Multicenter Hemophilia Cohort Studies I & II. Subjects were classified as having an inhibitor to FVIII on the basis of at least one inhibitor titer ≥1.0 Bethesda unit. SNP genotypes were determined by Sequenom MALDI-TOF spectroscopy. Haplotype frequencies were estimated using expectation maximization (EM) algorithm, and generalized linear models (GLM) were used to assess the marginal effect of SNPs and haplotypes on FVIII inhibitor development risk after adjusting for HIV infection and HCV persistence (prevalence in controls, 46% and 77%, respectively). Results: Of the 366 SNPs, 298 (81%) had unambiguous genotypes in >80% of the subjects and consequently qualified to the association analyses. Significant associations were seen between loci in the IL10, IL12A, IL1, IL2, TNFα, & IL17A genes and inhibitor development. Particularly, individuals carrying an 8 SNP haplotype located ∼10 kb 5' to the IL10 initiation start site were at higher risk to develop inhibitors than individuals with the most common haplotype (OR:1.54, 95% CI:1.15–2.06, p-value = 0.004). This haplotype covers a region that includes the IL10G microsatellite previously studied by Astermark et al (2006a), as well as the nearby IL10R microsatellite. Interestingly, the effect of this haplotype on FVIII inhibitor development was larger among HIV+ subjects in our study (OR: 1.81, 95% CI: 1.18–2.77 vs. OR: 1.28, 95% CI: 0.85–1.93 for HIV+ and HIV- subjects, respectively). A similar phenomenon was seen with haplotypes in the IL12A and IL2 genes. Additional haplotypes in genes for IL1α, IL1β, TNFα, and IL17A significantly increased or decreased risk for inhibitor development. No association was seen between FVIII haplotypes and inhibitors in this group of Caucasian subjects with hemophilia A, in contrast to the findings in African-Americans by Viel et al (2009). Also, no significant effect on inhibitor risk was seen for the factor V Leiden or prothrombin 20210 polymorphisms. Conclusions and Future Directions: Our large case-control study confirms the findings in family studies that IL10 and TNFα genotypes confer risk for inhibitor development in Caucasians with severe hemophilia A, which differed by HIV status and were of lower magnitude than seen previously in family studies. We identified four other genes in which certain haplotypes alter inhibitor risk. More work is needed to identify inhibitor associations in non-Caucasian populations and to corroborate and more specifically define our novel associations in Caucasians. Moreover, whole genome association and other studies should be considered to identify additional modifier genes and potential targets for intervention to prevent inhibitors. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 109 (11) ◽  
pp. 4693-4697 ◽  
Author(s):  
Samantha C. Gouw ◽  
Johanna G. van der Bom ◽  
Günter Auerswald ◽  
Carmen Escuriola Ettinghausen ◽  
Ulf Tedgård ◽  
...  

Abstract It has been suggested that plasma-derived factor VIII products induce fewer inhibitors than recombinant factor VIII products. We investigated the relationship of factor VIII product type and switching between factor VIII products with the risk to develop inhibitors. This multicenter retrospective cohort study included 316 patients with severe hemophilia A born between 1990 and 2000. The outcome was clinically relevant inhibitor development, defined as the occurrence of at least 2 positive inhibitor titers with decreased recovery. The risk of inhibitor development was not clearly lower in plasma-derived compared with recombinant factor VIII products (relative risk [RR], 0.8; 95% confidence interval [CI], 0.5-1.3). Among high-titer inhibitors, the possible reduction in risk was even less pronounced (RR, 0.9; CI, 0.5-1.5). Plasma-derived products with considerable quantities of von Willebrand factor (VWF) carried the same risk for inhibitor development as recombinant factor VIII products (RR, 1.0; CI, 0.6-1.6). Switching between factor VIII products did not increase the risk for inhibitors (RR, 1.1; CI, 0.6-1.8). In conclusion, our findings support neither the notion that plasma-derived factor VIII products with considerable concentrations of VWF confer a lower risk to develop inhibitory antibodies than recombinant factor VIII products, nor that switching between factor VIII product brands increases inhibitor risks in previously untreated patients with severe hemophilia A.


1998 ◽  
Vol 80 (11) ◽  
pp. 779-783 ◽  
Author(s):  
Y. Laurian ◽  
E. P. Satre ◽  
A. Borel Derlon ◽  
H. Chambost ◽  
P. Moreau ◽  
...  

SummaryFifty French previously untreated patients with severe hemophilia A (factor VIII <1%), treated with only one brand of recombinant factor VIII (rFVIII), were evaluated for inhibitor development, assessment of risk factors and outcome of immune tolerance regimen. The median period on study was 32 months (range 9-74) since the first injection of rFVIII. Fourteen patients (28%) developed an inhibitor, four of whom (8%) with a high titer (≥10 BU). All inhibitor patients but one continued to receive rFVIII either for on-demand treatment or for immune tolerance regimen (ITR). Among these patients, inhibitor was transient in 2 (4%), became undetectable in 6 and was still present in 6. The prevalence of inhibitor was 12%. Presence of intron 22 inversion was found to be a risk factor for inhibitor development. Immune tolerance was difficult to achieve in our series despite a follow-up period of 16 to 30 months: immune tolerance was complete in only one out of the 3 patients undergoing low dose ITR and in one out of the 5 patients with high dose ITR.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3185-3185
Author(s):  
Robert F. Sidonio ◽  
Alexis A. Thompson ◽  
Flora Peyvandi ◽  
Canan Albayrak ◽  
Seoh Leng Yeoh ◽  
...  

Abstract Background The development of inhibitors to exogenous factor VIII (FVIII) is a serious treatment complication in patients with hemophilia A. Immune tolerance induction (ITI) is the only proven method for the eradication of FVIII inhibitors. This prospective, multicenter, open-label, phase 3 study (NCT02615691) is being conducted to determine the safety, immunogenicity, and efficacy of the extended half-life (EHL) recombinant FVIII rurioctocog alfa pegol (Adynovate ®; Baxalta US Inc., a Takeda company, Lexington, MA, USA) in previously untreated patients (PUPs) with severe hemophilia A. The data presented here aims to evaluate the efficacy and safety of ITI therapy with rurioctocog alfa pegol in patients who developed FVIII inhibitors. Methods Eligible patients were ˂6 years of age with severe hemophilia A (FVIII &lt;1%) and &lt;3 exposure days (ED) to rurioctocog alfa pegol, octocog alfa, or plasma transfusion at any time prior to screening. Patients with detectable FVIII inhibitory antibodies at screening or a history of FVIII inhibitors prior to screening (≥0.6 Bethesda units [BU]) were excluded from the study. Patients received intravenous rurioctocog alfa pegol as prophylaxis (25-50 IU/kg, up to 80 IU/kg ≥1 × weekly) and/or on-demand therapy (10-50 IU/kg, up to 80 IU/kg depending on bleed severity). Patients who developed a high-titer FVIII inhibitor (&gt;5.0 BU) or low-titer FVIII inhibitor (≥0.6 BU to ≤ 5.0 BU) plus poorly controlled bleeding despite increased FVIII doses and/or bypassing agents, were eligible for ITI therapy. Dosing for ITI therapy ranged between 50 IU/kg 3 × weekly (low dose) and 100-200 IU/kg daily (high dose) at investigator discretion. This protocol-specified interim analysis was conducted after 50 patients had completed ≥50 EDs without developing confirmed inhibitors to rurioctocog alfa pegol or had developed a confirmed FVIII inhibitor at any time. The data cut-off was 30 August 2019. The primary endpoint of this study was the success rate of ITI with rurioctocog alfa pegol. Success was defined as an inhibitor titer persistently &lt;0.6 BU, FVIII incremental recovery (IR) ≥66% of baseline following 84- to 96-hour wash-out, and FVIII half-life ≥6 hours (dependent on protocol version). Secondary endpoints included the rates of partial success and failure of ITI, and annualized bleeding rate (ABR) during ITI. The number and percentage of patients reporting adverse events (AEs) and serious AEs (SAEs) were recorded for patients treated with ITI. Informed consent and ethics approval were obtained. Results As of the data cut-off, 59 (73.8%) of 80 enrolled patients had received ≥1 dose of rurioctocog alfa pegol; 18 patients did not meet the eligibility criteria (screen failures) and 4 discontinued prior to treatment. 10 patients developed an inhibitor to rurioctocog alfa pegol (high titer: n=5; low titer: n=5), of these, 6 patients were enrolled to receive ITI and only 5 of these (83.3%) actually received ≥1 dose of rurioctocog alfa pegol for the treatment of FVIII inhibitors (low dose: n=3; high dose: n=2). Of these 5 patients, 1 completed high-dose ITI therapy and this was successful (based on negative inhibitor titer and IR ≥66% of baseline). The remaining 4 patients were continuing in the study at the time of the data cut-off. Of the 5 patients who received ≥1 dose of ITI, 4 (80.0%) had a total of 17 AEs, 3 (60.0%) experienced 8 SAEs, and 1 experienced a treatment-related SAE of FVIII inhibition. It is important to note that the onset date of FVIII inhibitor development in this patient occurred prior to initiation of ITI. One patient experienced 2 catheter-related AEs, both of which resolved, and no patients experienced thrombotic AEs, study procedure-related AEs, or AEs leading to discontinuation of treatment. Discussion This is the first prospective study of the EHL recombinant FVIII rurioctocog alfa pegol for the treatment of PUPs with severe hemophilia A. These preliminary results demonstrate that rurioctocog alfa pegol has a safety profile consistent with previous studies. In addition, these interim data suggest that using a high-dose regimen for ITI therapy is potentially efficacious in PUPs who have developed FVIII inhibitors, although only 1 patient had completed ITI at the time of this interim analysis. Disclosures Sidonio: Pfizer: Consultancy; Octapharma: Consultancy, Research Funding; Catalyst: Consultancy; Novo Nordisk: Consultancy; Bayer: Consultancy; Guardian Therapeutics: Consultancy; Genentech: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; Biomarin: Consultancy. Thompson: Global Blood Therapeutics: Current equity holder in publicly-traded company; CRISPR Therapeutics: Research Funding; Vertex: Research Funding; Editas: Research Funding; Graphite Bio: Research Funding; Novartis: Research Funding; Agios: Consultancy; Beam: Consultancy; Celgene/BMS: Consultancy, Research Funding; Biomarin: Research Funding; Baxalta: Research Funding; bluebird bio, Inc.: Consultancy, Research Funding. Peyvandi: Bioverativ: Honoraria; Sanofi: Consultancy, Honoraria; Sobi: Consultancy, Honoraria; Spark: Honoraria; Takeda: Honoraria; Roche: Honoraria; Grifols: Honoraria. Yeoh: Grifols: Honoraria; Roche: Honoraria; Pfizer: Honoraria; Takeda: Honoraria. Lam: Takeda: Consultancy, Honoraria; Roche: Honoraria; Bayer: Honoraria; Pfizer: Consultancy, Honoraria. Maggiore: IQVIA: Current Employment. Engl: Takeda: Current equity holder in publicly-traded company; Baxalta Innovations GmbH, a Takeda company: Current Employment. Allen: Takeda: Current equity holder in publicly-traded company; Takeda Development Center Americas, Inc.: Current Employment. Tangada: Takeda Development Center Americas, Inc: Current Employment; Takeda: Current equity holder in publicly-traded company.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1211-1211
Author(s):  
Alessandra N L Prezotti ◽  
Monica H Cerqueira ◽  
Marilia Renni ◽  
Clarissa Ferreira ◽  
Ieda S. Pinto ◽  
...  

Abstract Introduction: One of the most important complications in the treatment of patients with hemophilia A is the formation of neutralizing antibodies (inhibitors) interfering in the coagulant activity of factor VIII (FVIII). The presence of inhibitor causes a direct impact on mortality and morbidity in these patients and considerably increases the cost of treatment. Among the non-genetic risk factors for inhibitor development, the influence of the type of factor concentrate used in replacement therapy (recombinant or plasma-derived) remains controversial. Thus, the evaluation of an additional population in the real world setting may contribute to elucidate this problem. Since August 2013, almost all previously untreated patients (PUPs) with hemophilia A in Brazil have been receiving exclusively the same third-generation recombinant FVIII (rFVIII) (Advate®, Shire). Objective: The aim of this study is to evaluate the immunogenicity of rFVIII (Advate®). In this context, we analyzed the occurrence of inhibitor among severe and moderately severe hemophilia A PUPs during the first 50 exposure days (EDs) to Advate®. Methods: This is an open-label, multicenter, prospective/retrospective, uncontrolled, observational study conducted in eight reference hemophilia treatment centers from distinct geographic areas in Brazil. The inclusion criteria were (a) diagnosis of severe or moderately severe hemophilia A (FVIII:C <2 IU/dL), (b) absence of previous exposure to other FVIII concentrates, except a maximum of 5 previous exposures to any blood components (whole blood, fresh-frozen plasma, packed red cells, platelets, or cryoprecipitate), and (c) exclusive treatment with Advate® until the 50th ED or until inhibitor development (primary endpoint). Positive inhibitor was defined as at least two consecutive plasma samples with Bethesda-Nijmegen assay results ≥0.60 BU/mL. Patients were considered as having low-titer inhibitors when peak titers were <5 BU/mL, and high-titer inhibitors if inhibitor titer was ≥5 BU/mL on at least one occasion. Any clinical information considered relevant for the risk of inhibitor development was analyzed when available, and included family history of inhibitor, F8 genotype, ethnicity (defined according physical traits and ancestry ethnic background in the last three generations), age at first rFVIII exposure, treatment regimens (prophylaxis or episodic), doses, occurrence of a severe bleeding episode, surgery, and use of FVIII concentrate simultaneously to infection or vaccination. Results: So far, 122 patients were enrolled, and 100 patients reached the 50th ED to rFVIII or developed inhibitor. Twenty-two are still on Advate® and have not achieved 50EDs (7 patients: 20 to 50EDs; 15 patients: <20ED). Overall, the median age at first exposure to Advate® was 11.9 months (interquartile range (IQR): 7.5-16.7), and most patients were African-descendants (48%), followed by Caucasians (45%). Positive inhibitor was detected in 35 of the 100 patients (35%), and 71% occurred during the first 20EDs. Most inhibitors were detected during prophylactic treatment (29 of 35; 82.9%). Twenty-five (25%) patients had high-titer inhibitor. Although not statistically significant, 19/48 (39.6%) of the African-descendants patients developed inhibitor, in contrast to 15/45 (33%) of the Caucasians. Interestingly, inhibitor was detected in only 1/7 (14.6%) of the patients with the indigenous background (native population). The influence of other risk factors, as severe bleeding episodes, presence of infection, surgery and history of blood transfusion, were not statistically significant. Conclusions: Overall inhibitor development in this cohort is consistent with results reported in other PUP studies with recombinant products. The majority of inhibitors developed during the first 20EDs. However, no other risk factor as intensive treatment was statistically significant, due to the small number of events observed. Although observational studies have limitations to assess the immunogenicity of FVIII products, our study contributes to this knowledge, since it evaluates a single third-generation rFVIII in a distinct population, with similar access to factor concentrate and same treatment regimen. Disclosures Prezotti: Shire: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pfizer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Bioverative: Membership on an entity's Board of Directors or advisory committees; Novo Nordisk: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Bayer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Roche: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Medina:Shire: Speakers Bureau. Ozelo:Novo Nordisk: Honoraria, Research Funding, Speakers Bureau; BioMarin: Honoraria, Speakers Bureau; Shire: Honoraria, Research Funding, Speakers Bureau; Pfizer: Honoraria, Research Funding, Speakers Bureau; Bioverativ: Honoraria, Research Funding; Grifols: Honoraria.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3665-3665
Author(s):  
Lulu Zhang ◽  
Ziqiang Yu ◽  
Wei Zhang ◽  
Lijuan Cao ◽  
Changgeng Ruan

Abstract Abstract 3665 Introduction: The inhibitor against factor VIII would develop in one-third to one-fourth patients with hemophilia A, which is the severe complication during replacement therapy. Although mutation in factor VIII gene is the main determinant, HLA alleles and gene polymorphism of cytokines, such as TNF-α, CTLA-4 and IL-10, are also associated with inhibitor development. The polymorphisms of TNF-α-308G>A, CTLA-4 -318C>T and one of the CA repeat microsatellites in IL-10 gene were analyzed in 140 Chinese patients with hemophilia A treated with plasma-derived F‡[ concentrates and in 108 normal controls, in order to evaluate their contribution to inhibitor development. Methods: Genomic DNA was extracted from citrate-preserved blood from all patients and normal controls. The promoter region of TNF-α and CTLA-4 were amplified by PCR, then the PCR products of TNF-α and CTLA-4 were digested by NcoI or MseI, respectively, then subjected to 2% agarose gel electrophoresis. PCR products were also directly sequenced to identify the genotype. The short tandem repeat in the promotor region of IL-10 was amplified by PCR, then analyzed by capillary electrophoresis. The inhibitor activities against factor VIII in plasma of patients with hemophilia A were measured by modified-Nijmegen assay simultaneously. Results: The frequencies of -308 G allele and A allele in patients with hemophilia A were 90.7% and 9.3% respectively, compared with 90.3% and 9.7% in normal controls. The frequencies of -318 C allele and T allele in patients with hemophilia A were 87.9% and 12.1% respectively, compared with 86.6% and 13.4% in normal controls. The allele 134bp positive and allele 134bp negative in patients with hemophilia A were 27.9% and 72.1% respectively, compared with 29.6 % and 71.4% in normal controls. There were not significant difference of -308G/A allele frequencies ƒ A-318C/T allele frequencies and 134bp allele frequencies between two groups. The F‡[ inhibitor activity was identified in 34/140 (24.3%) patients. In the patients with inhibitor, 13/34(38.2%) patients carried the -308 A allele, the frequencies of -308 G allele and A allele in these patients were 75% and 25% respectively. The patients with hemophilia A who carried -308 A allele had a high risk of inhibitor development (OR,7.519; 95%CI,3.168 ‘17.844), especially for the severe patients with hemophilia A (OR,8.163; 95%CI,2.521 ‘26.434). Also in the patients with inhibitor, 10/34(29.4%) patients carried the -318 T allele, the frequencies of -318 C allele and T allele in these patients were 83.8% and 16.2% respectively. There is no statistical significance regarding the risk of inhibitor development between the patients who were carrier of C allele and T allele (OR,1.586,95%CI,0.729 ‘3.450). The frequencies of allele 134bp positive and allele 134bp negative in patients with inhibitor were 44% and 56% respectively, there was no statistical significance regarding the risk of inhibitor development between the patients who were the allele 134bp positive and those who were allele 134bp negative (OR,1.769,95%CI,0.676 ‘4.627). Conclusion: TNF-α-308G>A polymorphism is significantly associated with factor VIII inhibitor development in Chinese patients with hemophilia A. TNF-αgene may be a useful marker and potential modulator of the immune response to replacement therapy in patients with hemophilia A. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 109 (11) ◽  
pp. 4648-4654 ◽  
Author(s):  
Samantha C. Gouw ◽  
Johanna G. van der Bom ◽  
H. Marijke van den Berg

Abstract The CANAL Study (Concerted Action on Neutralizing Antibodies in severe hemophilia A) was designed to describe the relationship between treatment characteristics and inhibitor development in previously untreated patients with severe hemophilia A. This multicenter retrospective cohort study investigated 366 consecutive patients born between 1990 and 2000. The outcome was clinically relevant inhibitor development, defined as the occurrence of at least 2 positive inhibitor titers combined with a decreased recovery. Eighty-seven (24%) patients developed inhibitors (69 high titer [19%]). The incidence of inhibitors appeared to be associated with age at first treatment, decreasing from 41% for those treated within the first month of age to 18% in those treated after 18 months; after adjustment for treatment intensity, this association largely disappeared. Surgical procedures and peak treatment moments at start of treatment increased inhibitor risk (relative risk [RR], 3.7; 95% confidence interval [CI], 2.0-7.1; and RR, 3.3; CI, 2.1-5.3, respectively). Regular prophylaxis was associated with a 60% lower risk than on-demand treatment (RR, 0.4; CI, 0.2-0.8). Our findings suggest that the previously reported associated between an early age at first exposure and the risk of inhibitor development is largely explained by early, intensive treatment. The latter appears to be an independent risk factor for inhibitor development. In addition, early, regular prophylaxis may protect patients with hemophilia against the development of inhibitors.


Sign in / Sign up

Export Citation Format

Share Document