scholarly journals Recombinant factor XIII: a safe and novel treatment for congenital factor XIII deficiency

Blood ◽  
2012 ◽  
Vol 119 (22) ◽  
pp. 5111-5117 ◽  
Author(s):  
Aida Inbal ◽  
Johannes Oldenburg ◽  
Manuel Carcao ◽  
Anders Rosholm ◽  
Ramin Tehranchi ◽  
...  

Congenital factor XIII (FXIII) deficiency is a rare, autosomal-recessive disorder, with most patients having an A-subunit (FXIII-A) deficiency. Patients experience life-threatening bleeds, impaired wound healing, and spontaneous abortions. In many countries, only plasma or cryoprecipitate treatments are available, but these carry a risk for allergic reactions and infection with blood-borne pathogens. The present study was a multinational, open-label, single-arm, phase 3 prophylaxis trial evaluating the efficacy and safety of a novel recombinant FXIII (rFXIII) in congenital FXIII-A subunit deficiency. Forty-one patients ≥ 6 years of age (mean, 26.4; range, 7-60) with congenital FXIII-A subunit deficiency were enrolled. Throughout the rFXIII prophylaxis, only 5 bleeding episodes (all trauma induced) in 4 patients were treated with FXIII-containing products. The crude mean bleeding rate was significantly lower than the historic bleeding rate (0.138 vs 2.91 bleeds/patient/year, respectively) for on-demand treatment. Transient, non-neutralizing, low-titer anti-rFXIII Abs developed in 4 patients, none of whom experienced allergic reactions, any bleeds requiring treatment, or changes in FXIII pharmacokinetics during the trial or follow-up. These non-neutralizing Abs declined below detection limits in all 4 patients despite further exposure to rFXIII or other FXIII-containing products. We conclude that rFXIII is safe and effective in preventing bleeding episodes in patients with congenital FXIII-A subunit deficiency. This study is registered at http://www..clinicaltrials.gov as number NCT00713648.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 20-20 ◽  
Author(s):  
Aida Inbal ◽  
Johannes Oldenburg ◽  
Manuel Carcao ◽  
Anders Rosholm ◽  
Ramin Tehranchi ◽  
...  

Abstract Abstract 20 Congenital Factor XIII (FXIII) deficiency is a rare autosomal recessive disorder affecting approximately 1 in 2–5 million individuals without gender or ethnic predilection. The plasma form of FXIII is a heterotetramer [A2B2] comprising two FXIII-A subunits and two FXIII-B subunits. A-subunit deficiency predominates (95%). FXIII deficient patients have a high risk of life-threatening bleeds, notably spontaneous intracranial haemorrhage. They experience impaired wound healing, and recurrent first trimester spontaneous abortions. Given the severity of the disorder there is a clinical need for providing effective haemostatic replacement therapy. Currently, only plasma derived products are approved for use in the United States. These may carry a risk of blood borne infection, sensitization and allergic reactions. A multi-center, multi-national, open-label, single-arm, multiple dosing phase 3 (prophylaxis) trial was undertaken to evaluate the efficacy and safety of a novel recombinant FXIII (rFXIII) in the prevention of bleeds in congenital FXIII-A subunit deficiency. This rFXIII-A2 dimer is expressed in Saccharomyces Cerevisiae and is an exact copy of the human A-subunit, which is the active cross-linking enzyme. When rFXIII is administered, it immediately binds to the B subunit that is present in the bloodstream. Forty-one patients aged ≥7 years [mean: 26.4 (range: 7–60); 23 males; 18 females] with a diagnosis of severe congenital FXIII-A subunit deficiency were enrolled. Patients entered a 4-week run-in period followed by monthly treatment with 35 IU/kg of rFXIII for 52 weeks. Informed consent was obtained prior to any trial-related activities. Bleeding episodes and adverse events were recorded. Testing was conducted monthly to measure pre- and post-dose FXIII activity and concentration, 5M urea clot solubility and anti-FXIII antibodies. Where antibodies were found, detailed testing for neutralizing potential was performed. Data were compared with bleeding rates based on historical data. During the treatment period with rFXIII (466 patient months) five bleeding episodes treated with FXIII-containing products were observed in four patients. All five were associated with trauma. No spontaneous bleeds that require treatment or intracranial haemorrhage occurred during rFXIII treatment period in any of the patients. The annual rate of bleeds requiring treatment was estimated via a Poisson model to be 0.048 bleeds/patient/year (95% CI: [0.0094; 0.2501] - significantly lower than the historic bleeding rate of 2.91 (p<0.0001). The crude mean rate of bleeding was 0.138 bleeds/patient/year. The occurrence of bleeds requiring treatment was not associated with low FXIII activity levels in study patients. The average FXIII activity at 1 hr post-dose was 77.0 ± 20.3% (mean±SD). The mean incremental increase in activity from pre-dose to 1 hr post-dose was 1.68 ± 0.51% /IU/ kg. Four patients developed transient, non-neutralizing, low-titer anti-rFXIII antibodies. None of these patients developed anaphylactic or allergic reactions, bleeding episodes or changes in FXIII pharmacokinetics at any time. Furthermore, the non-neutralizing antibodies declined below the limit of detection in all patients despite repeated exposure to rFXIII or other FXIII containing products. Consequently, these low titer, non-neutralizing antibodies appear to be clinically insignificant. No clinical safety issues, thromboembolic events, or fatal adverse events were recorded. In conclusion, rFXIII-A2 is safe and effective prophylaxis for preventing bleeding episodes in patients with congenital FXIII-A subunit deficiency. Disclosures: Inbal: NovoNordisk: Honoraria, Research Funding. Oldenburg:novonordisk: Research Funding. Carcao:NovoNordisk: Research Funding. Rosholm:NovoNordisk: Employment. Tehranchi:NovoNordisk: Employment. Nugent:NovoNordisk: Consultancy, Research Funding.


2006 ◽  
Vol 95 (01) ◽  
pp. 77-84 ◽  
Author(s):  
Verena Schroeder ◽  
Esther Meili ◽  
Trinh Cung ◽  
Peter Schmutz ◽  
Hans Kohler

SummaryIn 1960, the first case report on factor XIII deficiency was published describing a seven-year-old Swiss boy with a so far unknown bleeding disorder. Today, more than 60 mutations in the factor XIIIA- and B-subunit genes are known leading to congenital factor XIII deficiency. In the present study, we describe six novel mutations in the factor XIII A-subunit gene. Additionally, we present the molecular characterisation of the first described patient with congenital factor XIII deficiency. The six novel mutations include a small deletion, Glu202 del G, leading to a premature stop codon and truncation of the protein, and a splice site mutation at the exon 10/intron 10 boundary, +1G/A, giving rise to an incorrect spliced mRNA lacking exons 10 and 11. The remaining four mutations are characterised by the single amino acid changes Met159Arg, Gly215Arg, Trp375Cys, and His716Arg, and were expressed in COS-1 cells. Antigen levels and activity of the mutants were significantly reduced compared to the wild-type. The patient described in 1960 also shows a single amino acid change, Arg77Cys. Structural analysis of all mutant enzymes suggests several mechanisms leading to destabilisation of the protein.


2011 ◽  
Vol 106 (11) ◽  
pp. 908-913 ◽  
Author(s):  
Vera Tiedje ◽  
Daniela Dunkler ◽  
Cihan Ay ◽  
Birgit Horvath ◽  
Peter Quehenberger ◽  
...  

SummaryVenous thromboembolism (VTE) is a life-threatening complication in cancer patients. Identification of risk factors has been in focus in the past years. Functional single nucleotide polymorphisms (SNP) of coagulation factors known to influence the concentration or function may be considered to influence the risk of VTE in cancer patients. We evaluated the influence of fibrinogen plasma levels, the –455G>A SNP in the fibrinogen beta gene and the Val34Leu (163G>T) SNP in the factor XIII A-subunit (FXIII-A) gene on the risk of VTE. In 1,079 tumour patients recruited for the prospective Vienna Cancer and Thrombosis Study (CATS) fibrinogen levels were determined by the Clauss method. The FXIII-A Val34Leu and the fibrinogen –455G>A SNPs were tested by allele-specific PCR. The median follow-up time was 604 days, 83 thrombotic events occurred. The median fibrinogen level was 381 mg/dl (25th-75th percentile: 312 to 467). In a multivariable Cox model adjusted to chemotherapy, surgery, radiotherapy, age and sex, neither the fibrinogen concentration (hazard ratio [HR] =1.05, confidence interval [CI] 0.839–1.310 p=0.68), nor the –455G>A SNP (HR=0.77, 95%CI 0.491–1.197; p=0.24), nor the Val34Leu SNP (HR=0.99, 95%CI 0.646–1.542 p=0.99) were associated with occurrence of VTE. The fibrinogen concentration was not significantly different among the fibrinogen –455G or A genotype carriers (p = 0.33). Disseminated intravascular coagulation was observed in only five patients, none of these developed VTE. In conclusion, fibrinogen plasma levels, the fibrinogen –455G>A and the FXIII-A Val34Leu polymorphisms were not associated with VTE in our study.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2149-2149
Author(s):  
Vytautas Ivaskevicius ◽  
Rainer Seitz ◽  
Hans P. Kohler ◽  
Laszlo Muszbek ◽  
Robert A.S. Ariens ◽  
...  

Abstract Inherited factor XIII (FXIII) deficiency is a rare autosomal recessive disorder affecting approximately one out of one to three million people. FXIII deficiency is characterized by a lifelong bleeding tendency, impaired wound healing and spontaneous abortions in females. In 1993, the European Thrombosis Research Organization (ETRO) Working Party on FXIII initiated a Europe-wide questionnaire on inherited FXIII deficiency. Since 2005, the registry has been endorsed by the Factor XIII Subcommittee of the Scientific and Standardization committee (SSC) of the ISTH. The analysis of 104 European patients demonstrated that the most common bleeding symptoms were subcutaneous bleeding (57%) followed by delayed umbilical cord bleeding (56%), muscle hematoma (49%), hemorrhage after surgery (40%), hemarthrosis (36%), and intracerebral bleeding (34%). Prophylactic treatment was initiated in about 70% of all patients. FXIII-B subunit-deficient patients had a milder phenotype than patients with FXIII-A subunit deficiency.The most frequent mutation affecting the F13A gene was a splice site mutation in intron 5 (IVS5-1G&gt;A).This mutation was found in eight (17%) of 46 analyzed families.The haplotype analysis of patients carrying the IVS5-1A allele was consistent with a founder effect. Recently, we created a new FXIII database website (http://www.f13-database.de) with information about FXIII proteins, genes, mutations and polymorphisms. This website also includes a new questionnaire. Information provided by this questionnaire will allow better understanding of the differences of diagnostic and treatment possibilities in various parts of the world, and it will help to understand the impact of reduced FXIII activity in heterozygous relatives and finally, it will generally increase our knowledge on this rare disease. We hope that our initiative to establish a new international FXIII registry will be actively supported by the community involved in caring for FXIII deficient patients.


2018 ◽  
Vol 118 (03) ◽  
pp. 451-460 ◽  
Author(s):  
Manuel Carcao ◽  
Carmen Altisent ◽  
Giancarlo Castaman ◽  
Katsuyuki Fukutake ◽  
Bryce Kerlin ◽  
...  

AbstractRecombinant factor XIII-A2 (rFXIII-A2) was developed for prophylaxis and treatment of bleeds in patients with congenital FXIII A-subunit deficiency. mentor™2 (NCT00978380), a multinational, open-label, single-arm, multiple-dosing extension to the pivotal mentor™1 trial, assessed long-term safety and efficacy of rFXIII-A2 prophylaxis in eligible patients (patients with severe [<0.05 IU/mL] congenital FXIII subunit A deficiency) aged ≥6 years. Patients received 35 IU/kg rFXIII-A2 (exact dosing) every 28 ± 2 days for ≥52 weeks. Primary endpoint was safety (adverse events including immunogenicity); secondary endpoints were rate of bleeds requiring FXIII treatment, haemostatic response after one 35 IU/kg rFXIII-A2 dose for breakthrough bleeds and withdrawals due to lack of rFXIII-A2 efficacy. Steady-state pharmacokinetic variables were also summarized. Elective surgery was permitted during the treatment period. Sixty patients were exposed to rFXIII-A2; their median age was 26.0 years (range: 7.0–77.0). rFXIII-A2 was well tolerated without any safety concerns. No non-neutralizing or neutralizing antibodies (inhibitors) against FXIII were detected. Mean annualized bleeding rate (ABR) was 0.043/patient-year. Mean spontaneous ABR was 0.011/patient-year. No patients withdrew due to lack of efficacy. Geometric mean FXIII trough level was 0.17 IU/mL. Geometric terminal half-life was 13.7 days. rFXIII-A2 prophylaxis provided sufficient haemostatic coverage for 12 minor surgeries without the need for additional FXIII therapy; eight procedures were performed within 7 days of the patient's last scheduled rFXIII-A2 dose, and four were performed 10 to 21 days after the last dose.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1014-1014
Author(s):  
Jeanne M. Lusher ◽  
Steven W. Pipe ◽  
Sarah Alexander ◽  
Diane J. Nugent

Abstract In an ongoing Phase III study we are evaluating the prophylactic efficacy and long-term safety of Fibrogammin® P, a purified, pasteurized concentrate of Factor XIII, in patients (pts) with congenital Factor XIII deficiency (an extremely rare, hereditary, life-threatening bleeding disorder). The study is being conducted at 37 sites and has enrolled 61 pts; these 61 represent approximately 2/3 of US patients with FXIII deficiency. For this analysis, a retrospective chart review was undertaken to compare spontaneous bleed-event rates 24 months prior to (when possible) and after the initiation of therapy in a subset of 7 study subjects who did not receive any prophylactic therapy prior to entering the study. The table below presents data for each of these pts on dosing, bleeding episodes and treatment (Rx) during both observation periods (Obs Pd). An additional 4 pts had been treated prophylactically with regular infusions of FFP or cryoprecipitate and transitioned successfully to FXIII therapy; there were no seroconversions to HIV, Hepatitis B/C in any study pts. Pre-Fibrogammin P Therapy On Fibrogammin P Therapy Pt # Age ♦ Obs Pd (mos) / # Bleeds; Bleed Type Rx Obs Pd mos; (# inf) / # bleeds Bleed Type Dose/Kg† ♦ Age at onset of Fibrogammin Prophylactic therapy; † Pts were dosed on a unit basis (1250 unit vials), at 5-wk intervals, variability in weight and vial size lead to a range of per Kg doses; ⋄ p = .01 vs pre-therapy 504 5 yr 24 / 2 Hematomas none 12 (12) / 0 27 2703 10 mo 9 / 2 Umbilical cord; extensive bruising FFP 25 (26) / 1 Head trauma and bruising 29.7 – 44.9 2704 20 mo 6 / 4 Forehead × 2; frenulum × 2; other FFP × 5 26 (26) / 2 Traumatic foot; traumatic head (extracranial) 18.3 – 43.1 2705 9 yr 24 / 3 Lip; ankle; forehead FFP 13 (12) / 0 none 19.9 – 29.7 3301 19 yr 24 / 5 Large hematomas none 9.5 (11) / 0 none 18.9 3302 17 yr 24 / 0 none none 9.5 (12) / 0 none 24.3 3303 14 yr 24 / 8 Intracranial; others × 7 FFP, Cryoppt 10 (11) / 0 none 15.6 Mean 2.5 bleeds/yr 0.2 bleeds/yr⋄ As shown in the table, these children had 2.5 bleeds per year in the period before Fibrogammin prophylaxis, and only 0.2 per year while on Fibrogammin. These results demonstrate a consistent and clinically significant reduction in spontaneous bleeding with prophylactic use of Fibrogammin® P in pts with symptomatic congenital Factor XIII deficiency.


2014 ◽  
Vol 12 (12) ◽  
pp. 2038-2043 ◽  
Author(s):  
B. Kerlin ◽  
B. Brand ◽  
A. Inbal ◽  
S. Halimeh ◽  
D. Nugent ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document