Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Blood ; 142(3): 290-305, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37192286

ABSTRACT

Despite >80 years of clinical experience with coagulation factor VIII (FVIII) inhibitors, surprisingly little is known about the in vivo mechanism of this most serious complication of replacement therapy for hemophilia A. These neutralizing antidrug alloantibodies arise in ∼30% of patients. Inhibitor formation is T-cell dependent, but events leading up to helper T-cell activation have been elusive because of, in part, the complex anatomy and cellular makeup of the spleen. Here, we show that FVIII antigen presentation to CD4+ T cells critically depends on a select set of several anatomically distinct antigen-presenting cells, whereby marginal zone B cells and marginal zone and marginal metallophilic macrophages but not red pulp macrophages (RPMFs) participate in shuttling FVIII to the white pulp in which conventional dendritic cells (DCs) prime helper T cells, which then differentiate into follicular helper T (Tfh) cells. Toll-like receptor 9 stimulation accelerated Tfh cell responses and germinal center and inhibitor formation, whereas systemic administration of FVIII alone in hemophilia A mice increased frequencies of monocyte-derived and plasmacytoid DCs. Moreover, FVIII enhanced T-cell proliferation to another protein antigen (ovalbumin), and inflammatory signaling-deficient mice were less likely to develop inhibitors, indicating that FVIII may have intrinsic immunostimulatory properties. Ovalbumin, which, unlike FVIII, is absorbed into the RPMF compartment, fails to elicit T-cell proliferative and antibody responses when administered at the same dose as FVIII. Altogether, we propose that an antigen trafficking pattern that results in efficient in vivo delivery to DCs and inflammatory signaling, shape the immunogenicity of FVIII.


Subject(s)
CD4-Positive T-Lymphocytes , Factor VIII , Hemophilia A , Hemostatics , Animals , Mice , Dendritic Cells/metabolism , Factor VIII/immunology , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemostatics/immunology , Hemostatics/therapeutic use , Ovalbumin/immunology
2.
Front Immunol ; 10: 2991, 2019.
Article in English | MEDLINE | ID: mdl-31998296

ABSTRACT

Development of neutralizing antibodies against biotherapeutic agents administered to prevent or treat various clinical conditions is a longstanding and growing problem faced by patients, medical providers and pharmaceutical companies. The hemophilia A community has deep experience with attempting to manage such deleterious immune responses, as the lifesaving protein drug factor VIII (FVIII) has been in use for decades. Hemophilia A is a bleeding disorder caused by genetic mutations that result in absent or dysfunctional FVIII. Prophylactic treatment consists of regular intravenous FVIII infusions. Unfortunately, 1/4 to 1/3 of patients develop neutralizing anti-FVIII antibodies, referred to clinically as "inhibitors," which result in a serious bleeding diathesis. Until recently, the only therapeutic option for these patients was "Immune Tolerance Induction," consisting of intensive FVIII administration, which is extraordinarily expensive and fails in ~30% of cases. There has been tremendous recent progress in developing novel potential clinical alternatives for the treatment of hemophilia A, ranging from encouraging results of gene therapy trials, to use of other hemostatic agents (either promoting coagulation or slowing down anti-coagulant or fibrinolytic pathways) to "bypass" the need for FVIII or supplement FVIII replacement therapy. Although these approaches are promising, there is widespread agreement that preventing or reversing inhibitors remains a high priority. Risk profiles of novel therapies are still unknown or incomplete, and FVIII will likely continue to be considered the optimal hemostatic agent to support surgery and manage trauma, or to combine with other therapies. We describe here recent exciting studies, most still pre-clinical, that address FVIII immunogenicity and suggest novel interventions to prevent or reverse inhibitor development. Studies of FVIII uptake, processing and presentation on antigen-presenting cells, epitope mapping, and the roles of complement, heme, von Willebrand factor, glycans, and the microbiome in FVIII immunogenicity are elucidating mechanisms of primary and secondary immune responses and suggesting additional novel targets. Promising tolerogenic therapies include development of FVIII-Fc fusion proteins, nanoparticle-based therapies, oral tolerance, and engineering of regulatory or cytotoxic T cells to render them FVIII-specific. Importantly, these studies are highly applicable to other scenarios where establishing immune tolerance to a defined antigen is a clinical priority.


Subject(s)
Factor VIII/pharmacology , Factor VIII/therapeutic use , Animals , Antibodies, Neutralizing/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Factor VIII/immunology , Hemophilia A/drug therapy , Hemophilia A/immunology , Hemostatics/immunology , Humans , Immune Tolerance/drug effects , Immune Tolerance/immunology , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/therapeutic use , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
3.
Eur J Pharm Sci ; 123: 531-538, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30077714

ABSTRACT

Historically, clinical trials of haemophilia with inhibitors (HwI) have been challenged by the small patient population. New approaches to clinical trial methodology and statistical modelling could potentially be used for study optimization. The aim of this work was to evaluate the impact of different trial designs and study conditions on the estimated drug potency and power, and compare traditional statistical methods with repeated time-to-event (RTTE) modelling in terms of power. Bleeding information from a clinical trial of 23 haemophilia patients with inhibitors treated on-demand was used to develop a baseline RTTE model using NONMEM. Clinical trial simulations for a hypothetical anti-haemophilic drug were performed, by adding a drug effect and a literature-derived placebo effect to the baseline RTTE model, using different trial designs (parallel-group, placebo-controlled parallel-group, crossover and placebo-controlled crossover designs) and study conditions, including sample size, study duration and doses. The precision and accuracy of the estimated drug potency (EC50) and power for different trial designs, study conditions and statistical methods (RTTE modelling, t-test and negative binomial regression) were evaluated. The developed baseline RTTE model accurately described the clinical data. The crossover designs displayed up to four-fold higher precision of the estimated EC50 and three-fold higher power relative to the parallel-group trial designs. Furthermore, RTTE modelling provided a higher power relative to the traditional statistical tests. We found that crossover designs in combination with RTTE modelling can reduce the required sample size and study duration, while ensuring high power and precise estimation of EC50, in clinical trials of HwI.


Subject(s)
Antibodies/blood , Clinical Trials as Topic/methods , Factor VIII/administration & dosage , Hemophilia A/drug therapy , Hemostatics/administration & dosage , Research Design , Adolescent , Adult , Antibodies/immunology , Clinical Trials as Topic/statistics & numerical data , Computer Simulation , Cross-Over Studies , Data Interpretation, Statistical , Factor VIII/adverse effects , Factor VIII/immunology , Hemophilia A/blood , Hemophilia A/diagnosis , Hemophilia A/immunology , Hemostatics/adverse effects , Hemostatics/immunology , Humans , Male , Middle Aged , Recombinant Proteins/administration & dosage , Research Design/statistics & numerical data , Sample Size , Time Factors , Treatment Outcome , Young Adult
4.
Semin Thromb Hemost ; 44(2): 142-150, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28905351

ABSTRACT

The role of tissue factor (TF) as the major initiator of hemostatic blood coagulation is well recognized. The ability to form an adequate hemostatic clot is essential to the normal healing of an injury by staunching bleeding, stabilizing the injured tissue, and serving as a scaffold for repair processes. Also, some molecules produced during hemostasis, particularly thrombin, have cytokine and growth factor-like activities that contribute to inflammation and repair. However, TF itself has activities as a regulator of cellular processes via direct signaling, as well as by facilitating activation of proteolytically activated receptors by activated factors VII and X. The importance of hemostasis in the host response to injury makes it very difficult to separate the hemostatic from nonhemostatic effects of TF on wound healing. The literature in this area remains sparse but suggests that TF influences the course and tempo of healing by cell signaling events that impact inflammation, epithelialization, and angiogenesis.


Subject(s)
Hemostatics/immunology , Thromboplastin/immunology , Wound Healing/immunology , Humans
5.
Thromb Res ; 141 Suppl 2: S34-5, 2016 May.
Article in English | MEDLINE | ID: mdl-27207420

ABSTRACT

There are some issues in the current factor (F)VIII replacement therapy for severe hemophilia A. One is mental and physical burden for the multiple intravenous infusions, and the other is difficulty in the hemostatic treatment for the patients with FVIII inhibitor. The development of novel drug with fully hemostatic effect, simply procedure, and long-acting reaction has been expected. Recently, FVIIIa-mimicking humanized recombinant bispecific antibody (ACE910) against FIXa and FX was developed. In the non-human clinical study, primate model of acquired hemophilia A demonstrated that the ACE910 was effective on both on-going and spontaneous bleedings. A phase I clinical study was conducted in healthy adults by single subcutaneous infusion of ACE910, followed by the patients' part study, Japanese patients with severe hemophilia A without or with inhibitor were treated with once-weekly subcutaneous injection of ACE910 at three dose levels for 12 successive weeks. There was no significant adverse event related to ACE910 in the clinical and laboratorial findings, and t1/2 of ACE910 was ∼30 days. The median annual bleeding rates were reduced very markedly dose-dependently, independently of inhibitor. Furthermore, among the patients with dose escalation, bleeding rate was decreased as ACE910 dose was increased. In conclusion, ACE910 would have a number of promising features: its high subcutaneous bioavailability and long half-life make the patients possible to be injected subcutaneously with a once-a-week or less frequency. In addition, ACE910 would provide the bleeding prophylactic efficacy, independently of inhibitor.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Factor VIII/metabolism , Hemophilia A/drug therapy , Hemophilia A/metabolism , Hemostatics/therapeutic use , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Clinical Trials as Topic , Factor IXa/immunology , Factor IXa/metabolism , Factor X/immunology , Factor X/metabolism , Hemophilia A/blood , Hemophilia A/immunology , Hemorrhage/blood , Hemorrhage/immunology , Hemorrhage/metabolism , Hemorrhage/prevention & control , Hemostatics/immunology , Hemostatics/pharmacology , Humans
6.
J Thromb Haemost ; 14(2): 346-55, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26588198

ABSTRACT

UNLABELLED: ESSENTIALS: Anti-factor VIII (FVIII) inhibitory antibody formation is a severe complication in hemophilia A therapy. We genetically engineered and characterized a mouse model with complete deletion of the F8 coding region. F8(TKO) mice exhibit severe hemophilia, express no detectable F8 mRNA, and produce FVIII inhibitors. The defined background and lack of FVIII in F8(TKO) mice will aid in studying FVIII inhibitor formation. BACKGROUND: The most important complication in hemophilia A treatment is the development of inhibitory anti-Factor VIII (FVIII) antibodies in patients after FVIII therapy. Patients with severe hemophilia who express no endogenous FVIII (i.e. cross-reacting material, CRM) have the greatest incidence of inhibitor formation. However, current mouse models of severe hemophilia A produce low levels of truncated FVIII. The lack of a corresponding mouse model hampers the study of inhibitor formation in the complete absence of FVIII protein. OBJECTIVES: We aimed to generate and characterize a novel mouse model of severe hemophilia A (designated the F8(TKO) strain) lacking the complete coding sequence of F8 and any FVIII CRM. METHODS: Mice were created on a C57BL/6 background using Cre-Lox recombination and characterized using in vivo bleeding assays, measurement of FVIII activity by coagulation and chromogenic assays, and anti-FVIII antibody production using ELISA. RESULTS: All F8 exonic coding regions were deleted from the genome and no F8 mRNA was detected in F8(TKO) mice. The bleeding phenotype of F8(TKO) mice was comparable to E16 mice by measurements of factor activity and tail snip assay. Similar levels of anti-FVIII antibody titers after recombinant FVIII injections were observed between F8(TKO) and E16 mice. CONCLUSIONS: We describe a new C57BL/6 mouse model for severe hemophilia A patients lacking CRM. These mice can be directly bred to the many C57BL/6 strains of genetically engineered mice, which is valuable for studying the impact of a wide variety of genes on FVIII inhibitor formation on a defined genetic background.


Subject(s)
Factor VIII/genetics , Gene Deletion , Hemophilia A/genetics , Hemostasis , Animals , Antibodies/blood , Blood Coagulation Tests , Chromogenic Compounds , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Factor VIII/immunology , Factor VIII/metabolism , Factor VIII/pharmacology , Genetic Predisposition to Disease , Hemophilia A/blood , Hemophilia A/drug therapy , Hemophilia A/immunology , Hemostasis/drug effects , Hemostasis/genetics , Hemostatics/immunology , Hemostatics/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Severity of Illness Index
7.
Thromb Haemost ; 114(1): 56-64, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26017627

ABSTRACT

There is no prospective evidence on inhibitor recurrence among haemophilia A patients with low titre inhibitors or history of inhibitors, and whether or how therapeutic choices affect the risk of recurrence. The aims of this study were to synthesise safety data in patients with moderate-severe haemophilia A and with low titre inhibitors or inhibitor history enrolled in the rAHF PFM (ADVATE) - Post-Authorization Safety Studies (ADVATE-PASS) international programme. The study was conducted in clinics participating to the ADVATE PASS programme. The patient population consisted of patients entering the studies with low titre (≤ 5 BU) inhibitors or a positive personal history of inhibitors. Patients on Immune Tolerance Induction at study entry were excluded. Primary outcome was new or recurrent inhibitor titre > 5 BU. Secondary outcomes were any increase of inhibitor titre not reaching 5 BU; any unexplained change in treatment regimen. Primary analysis was done by two-stage random effects meta-analysis. Secondary analysis was done by a hierarchical Bayesian random effects logistic model. A total of 219 patients from seven studies were included. Of these 214 (97.7 %) patients had been previously treated for more than 50 exposure days. Two hundred ten patients had positive history for inhibitors, nine a baseline measurable titre. No patient presented a primary outcome event (95 % confidence interval [CI] 0-1.6 %). Six patients with previous history developed a low titre recurrence (overall rate 2.2, 95 %CI 0-4.8 %). When any increase of inhibitor titre or any treatment change was accounted for, overall 3.7 % (95 % CI 0 %-8.0 %) of patients experienced the outcome. In conclusion, the observed rate of events does not support the definition of this population as at high risk for inhibitor development.


Subject(s)
Antibodies/blood , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemorrhage/therapy , Hemostatics/therapeutic use , Bayes Theorem , Biomarkers/blood , Databases, Factual , Factor VIII/adverse effects , Factor VIII/immunology , Hemophilia A/blood , Hemophilia A/diagnosis , Hemophilia A/immunology , Hemorrhage/blood , Hemorrhage/diagnosis , Hemorrhage/immunology , Hemostatics/adverse effects , Hemostatics/immunology , Humans , Logistic Models , Monte Carlo Method , Observational Studies as Topic , Odds Ratio , Population Surveillance , Product Surveillance, Postmarketing , Risk Factors , Severity of Illness Index , Time Factors , Treatment Outcome
8.
Thromb Haemost ; 114(1): 46-55, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25879247

ABSTRACT

In nonsevere haemophilia A (HA) patients the presence of an inhibitor may exacerbate the bleeding phenotype dramatically. There are very limited data on the optimal therapeutic approach to eradicate inhibitors in these patients. We aimed to describe inhibitor eradication treatment in a large cohort of unselected nonsevere HA patients with inhibitors. We included 101 inhibitor patients from a source population of 2,709 nonsevere HA patients (factor VIII 2-40 IU/dl), treated in Europe and Australia (median age 37 years, interquartile range (IQR) 15-60; median peak titre 7 BU/ml, IQR 2-30). In the majority of the patients (71 %; 72/101) the inhibitor disappeared; either spontaneously (70 %, 51/73) or after eradication treatment (75 %, 21/28). Eradication treatment strategies varied widely, including both immune tolerance induction and immunosuppression. Sustained success (no inhibitor after rechallenge with factor VIII concentrate after inhibitor disappearance) was achieved in 64 % (30/47) of those patients rechallenged with FVIII concentrate. In high-titre inhibitor patients sustained success was associated with eradication treatment (unadjusted relative risk 2.3, 95 % confidence interval 1.3-4.3), compared to no eradication treatment. In conclusion, in nonsevere HA patients most inhibitors disappear spontaneously. However, in 35 % (25/72) of these patients an anamnestic response still can occur when rechallenged, thus disappearance in these patients does not always equal sustained response. Treatment for those requiring eradication has to be decided case by case, as one single approach is unlikely to be appropriate for all.


Subject(s)
Antibodies/blood , Desensitization, Immunologic/methods , Factor VIII/therapeutic use , Hemophilia A/therapy , Hemorrhage/therapy , Hemostatics/therapeutic use , Immunosuppressive Agents/therapeutic use , Adolescent , Adult , Biomarkers/blood , Child , Child, Preschool , Europe , Factor VIII/adverse effects , Factor VIII/immunology , Hemophilia A/blood , Hemophilia A/diagnosis , Hemophilia A/immunology , Hemorrhage/blood , Hemorrhage/diagnosis , Hemorrhage/immunology , Hemostatics/adverse effects , Hemostatics/immunology , Humans , Immune Tolerance , Middle Aged , South Australia , Time Factors , Treatment Outcome , Young Adult
9.
J Surg Res ; 194(2): 679-687, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25586331

ABSTRACT

BACKGROUND: Fibrocaps, a ready-to-use, dry-powder fibrin sealant containing human plasma-derived thrombin and fibrinogen, is being developed as an adjunct for surgical hemostasis. MATERIALS AND METHODS: Safety and efficacy of Fibrocaps applied directly or by spray device, in combination with gelatin sponge, was compared with that of gelatin sponge-alone in two randomized, single-blind controlled trials: FC-002 US (United States) and FC-002 NL (the Netherlands). A total of 126 adult patients were randomized (Fibrocaps: n = 47 [FC-002 US], n = 39 [FC-002 NL]; gelatin sponge alone: n = 23 [FC-002 US], n = 17 [FC-002 NL). One bleeding site was treated during a surgical procedure (n = 125). Time to hemostasis (primary end point) was measured, with a 28-d safety follow-up. Four surgical indications included hepatic resection (n = 58), spinal procedures (n = 37), peripheral vascular procedures (n = 30), and soft tissue dissection (n = 1). RESULTS: Mean (standard deviation) time to hemostasis was significantly shorter after Fibrocaps treatment than after gelatin sponge alone (FC-002 US: 1.9 [1.3] versus 4.8 min [3.1], P < 0.001; FC-002 NL: 2.2 [1.3] versus 4.4 min [3.1], P = 0.004). The incidence of hemostasis was greater after Fibrocaps compared with that of gelatin sponge alone within 3 min (FC-002 US: 83% versus 35%, P < 0.001; FC-002 NL: 77% versus 53%, P = 0.11), 5 min (94% versus 61%, P = 0.001; 95% versus 71%, P = 0.022), and 10 min (100% versus 78%, P = 0.003; 100% versus 82%, P = 0.025). Adverse events were consistent with surgical procedures performed and patients' underlying diseases and generally similar between treatment arms; most were mild or moderate in severity. Non-neutralizing antithrombin antibodies were detected in 5% of Fibrocaps-treated patients on day 29. CONCLUSIONS: Fibrocaps had good safety and efficacy profiles, supporting continuing clinical development as a novel fibrin sealant.


Subject(s)
Blood Loss, Surgical/prevention & control , Fibrin Tissue Adhesive/therapeutic use , Hemostasis, Surgical/instrumentation , Hemostatics/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Fibrin Tissue Adhesive/immunology , Gelatin Sponge, Absorbable , Hemostatics/immunology , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
10.
AAPS J ; 16(5): 1038-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24934295

ABSTRACT

Proteolytic cleavage of factor VIII (FVIII) to activated FVIIIa is required for participation in the coagulation cascade. The A2 domain is no longer covalently bound in the resulting activated heterotrimer and is highly unstable. Aspartic acid (D) 519 and glutamic acid (E) 665 at the A1-A2 and A2-A3 domain interfaces were identified as acidic residues in local hydrophobic pockets. Replacement with hydrophobic valine (V; D519V/E665V) improved the stability and activity of the mutant FVIII over the wild-type (WT) protein in several in vitro assays. In the current study, we examined the impact of mutations on secondary and tertiary structure as well as in vivo stability, pharmacokinetics (PK), efficacy, and immunogenicity in a murine model of Hemophilia A (HA). Biophysical characterization was performed with far-UV circular dichroism (CD) and fluorescence emission studies. PK and efficacy of FVIII was studied following i.v. bolus doses of 4, 10 and 40 IU/kg with chromogenic and tail clip assays. Immunogenicity was measured with the Bethesda assay and ELISA after a series of i.v. injections. Native secondary and tertiary structure was unaltered between variants. PK profiles were similar at higher doses, but at 4 IU/kg plasma survival of D519V/E665V was improved. Hemostasis at low concentrations was improved for the mutant. Immune response was similar between variants. Overall, these results demonstrate that stabilizing mutations in the A2 domain of FVIII can improve HA therapy in vivo.


Subject(s)
Factor VIII/pharmacology , Hemophilia A/drug therapy , Hemostatics/pharmacology , Amino Acid Substitution , Animals , Disease Models, Animal , Drug Stability , Factor VIII/administration & dosage , Factor VIII/chemistry , Factor VIII/genetics , Factor VIII/immunology , Factor VIII/pharmacokinetics , Hemophilia A/blood , Hemophilia A/genetics , Hemostasis/drug effects , Hemostatics/administration & dosage , Hemostatics/chemistry , Hemostatics/immunology , Hemostatics/pharmacokinetics , Hydrophobic and Hydrophilic Interactions , Injections, Intravenous , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Biological , Models, Molecular , Mutation , Protein Engineering , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
11.
Thromb Haemost ; 107(6): 1072-82, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22476554

ABSTRACT

We report a prospective trial of 55 previously untreated patients (PUPs) and minimally treated patients (MTPs) with severe/moderately severe haemophilia A (baseline factor VIII [FVIII] ≤2%) treated with a single FVIII replacement product. It was the objective of this study to evaluate the immunogenicity, efficacy, and safety of rAHF-PFM (Advate®). On-demand or prophylactic treatment regimens were determined at the discretion of the investigator. rAHF-PFM was also permitted for perioperative management. There were 633 bleeding episodes (BEs), including 517 treated, and 466 rated for efficacy. Haemostatic efficacy was considered excellent/good in 93% of 466 rated treatments. Of 517 treated BEs, 463/517 (90%) were managed with one (356/517 [69%]) or two infusions (107/517 [21%]). There were 27 surgeries. Intraoperative (n=22) and postoperative (n=25) haemostatic efficacies were considered excellent or good in 100% of rated surgeries. Related serious adverse events (SAEs) were inhibitor development in 16/55 (29.1%) subjects who received at least one infusion of rAHF-PFM. Non-serious, related adverse events (AEs) were few in number (14 in eight subjects). The odds ratio (OR [95% Confidence Interval, CI]) of developing inhibitors was significantly higher in subjects with a family history of inhibitor (4.95[1.29-19.06]), non-Caucasian ethnicity (4.18, [1.18-14.82]), and intensive treatment at high dose (4.5 [1.05-19.25]) within ≤20 exposure days (EDs). In conclusion, rAHF-PFM was safe and effective for the management and perioperative coverage of PUPs/MTPs with severe/moderately severe haemophilia A. This report supports previous findings from studies in which family history of inhibitor, non-Caucasian ethnicity, and high intensity treatment were associated with high risk of inhibitor development.


Subject(s)
Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemorrhage/drug therapy , Hemostasis/drug effects , Hemostatics/therapeutic use , Antibodies/blood , Drug Administration Schedule , Ethnicity , Europe/epidemiology , Factor VIII/administration & dosage , Factor VIII/adverse effects , Factor VIII/immunology , Female , Hemophilia A/blood , Hemophilia A/complications , Hemophilia A/ethnology , Hemorrhage/blood , Hemorrhage/ethnology , Hemorrhage/etiology , Hemostatics/administration & dosage , Hemostatics/adverse effects , Hemostatics/immunology , Humans , Infant , Infant, Newborn , Linear Models , Logistic Models , Male , North America/epidemiology , Odds Ratio , Prospective Studies , Risk Assessment , Risk Factors , Severity of Illness Index , Time Factors , Treatment Outcome
12.
Clin Appl Thromb Hemost ; 18(3): 243-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22345489

ABSTRACT

JMI-thrombin is used as topical hemostatic agent. While earlier clinically available JMI were reported to produce immunologic responses upon repeated exposure, the improved JMI, Recothrom?, and Evithrom? are claimed to be less immunogenic. Recothrom, despite its reduced immunogenic nature, upon repeated administration may result in the generation of antibodies (Abs) and that may cross react with bovine and human thrombin. Therefore, groups of rabbits were challenged repeatedly with Recothrom, Evithrom, and JMI over a 9-month period. Pre-immune blood and antiserum were collected from each rabbit on different time point. To determine their relative cross reactivity, JMI, Recothrom, and Evithrom were evaluated by western blotting using the rabbit IgG fractions. The results suggest that anti-Recothrom Abs cross-react with Evithrom and JMI in a time dependent fashion. Anti-JMI Abs did not cross-react with Recothrom, and Evithrom. Also, anti-Evithrom did not show any cross-reactivity with Recothrom and JMI at any time.


Subject(s)
Hemostatics/immunology , Immunoglobulin G/immunology , Thrombin/immunology , Animals , Cattle , Cross Reactions/immunology , Hemostatics/chemistry , Humans , Immunoglobulin G/chemistry , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Thrombin/chemistry
13.
J Am Coll Surg ; 213(6): 722-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21958507

ABSTRACT

BACKGROUND: This Phase 4, open-label study evaluated the immunogenicity and safety of a second exposure to recombinant human thrombin (rThrombin) in adult patients with previous exposure to rThrombin. STUDY DESIGN: Topical rThrombin was applied as a hemostatic aid during a surgical procedure (day 1). Adverse events and clinical laboratory abnormalities were monitored to day 29 (study end). Immunogenicity samples were collected on days 1 and 29. Thirty-one patients were treated at 9 study sites; 30 patients completed the study. RESULTS: Mean age was 59.5 years; 61.3% of patients were male. Study operations types included spinal (n = 23 of 31; 74.2%), arterial reconstruction or peripheral arterial bypass (n = 4; 12.9%), arteriovenous vascular access procedure (n = 3; 9.7%), and other (n = 1; 3.2%). A median of 10 mL rThrombin (1,000 IU/mL; range 5 to 60 mL) was prepared per patient. Median elapsed time since previous rThrombin exposure was 1.3 years (range 19 days to 3.3 years). Recombinant human thrombin was not observed to be immunogenic; no patients (n = 0 of 30, 0%; 95% CI 0.0%, 11.6%) became positive for anti-rThrombin product antibodies at day 29, approximately 1 month after the second exposure to rThrombin. The most commonly reported adverse events were procedural pain (n = 23 of 31, 74.2%), constipation (n = 8, 25.8%), and nausea (n = 8, 25.8%) All adverse events and clinical laboratory abnormalities were considered unrelated to treatment. For the majority of patients, maximal severity of any adverse event was mild or moderate. CONCLUSIONS: The immunogenicity and safety results of this Phase 4 rThrombin trial suggest that patients with known previous exposure may be safely re-exposed to topical rThrombin.


Subject(s)
Hemostasis, Surgical , Hemostatics/adverse effects , Hemostatics/immunology , Postoperative Complications , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Thrombin/adverse effects , Thrombin/immunology , Adult , Aged , Aged, 80 and over , Antibodies/blood , Cohort Studies , Drug Administration Schedule , Female , Hemostatics/administration & dosage , Humans , Male , Middle Aged , Recombinant Proteins/administration & dosage , Retreatment , Thrombin/administration & dosage , Time Factors , Young Adult
14.
J Pediatr Surg ; 46(10): 1992-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22008340

ABSTRACT

BACKGROUND: Previous studies of recombinant human thrombin (rThrombin) enrolled adult and adolescent patients. This phase 4, open-label, single-group study was conducted in pediatric patients undergoing synchronous burn wound excision and skin grafting to provide information regarding the safety and immunogenicity of rThrombin (primary and secondary endpoints) in this population. METHODS: Topical rThrombin was applied as a hemostatic aid during a surgical procedure (day 1). Adverse events and clinical laboratory abnormalities were recorded during the study. Immunogenicity samples were collected on days 1 and 29 (study end). Study results were summarized with descriptive statistics. RESULTS: Thirty patients enrolled and 28 completed the study. Mean age was 6.9 years (range, 0.9-17.8 years); 40.0% of patients were girls. Flame and scald were the most common burn types (33.3% each, n = 10/30). Mean graft size was 3.6% total body surface area. Procedural pain (50.0% patients), pruritus (43.3%), and anemia (30.0%) were the most commonly reported adverse events. All adverse events and clinical laboratory abnormalities were considered unrelated to treatment. No patients developed anti-rThrombin product antibodies at day 29. CONCLUSIONS: In pediatric patients undergoing burn wound excision and skin grafting, rThrombin was well tolerated and did not lead to the formation of anti-rThrombin product antibodies.


Subject(s)
Blood Loss, Surgical/prevention & control , Burns/surgery , Hemostatics/therapeutic use , Isoantibodies/blood , Recombinant Proteins/therapeutic use , Thrombin/therapeutic use , Administration, Topical , Adolescent , Anemia/epidemiology , Child , Child, Preschool , Combined Modality Therapy , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Epinephrine/therapeutic use , Female , Hemostatic Techniques , Hemostatics/adverse effects , Hemostatics/immunology , Humans , Infant , Isoantibodies/biosynthesis , Male , Pain/epidemiology , Postoperative Complications/epidemiology , Postoperative Hemorrhage/prevention & control , Pruritus/epidemiology , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Skin Transplantation , Thrombin/adverse effects , Thrombin/immunology
15.
Perfusion ; 26(6): 529-35, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21752926

ABSTRACT

OBJECTIVE: To determine how the anti-inflammatory properties of aprotinin impact on postoperative complications in children undergoing the Fontan procedure. METHODS: We included all patients between 14 months and 18 years (n=56) undergoing a Fontan operation at our institution between January 2005 and June 2009. The study group (n=29) included patients from January 2005 through December 2007 all of whom received aprotinin. The control group (n=27) included all patients from January 2008 through June 2009 who did not receive aprotinin. We reviewed all medical records and collected preoperative, intraoperative and postoperative data. Duration and volume of chest tube drainage were the primary outcome measures. RESULTS: Of the 20% of patients who had postoperative arrhythmias, multivariate logistic regression analysis demonstrated only aprotinin was associated with significantly decreased postoperative arrhythmias (P=0.01). Renal function and fenestration or Fontan thrombosis did not differ significantly; there was no statistically significant difference in volume or duration of chest tube drainage. Median duration of chest tube drainage was 7 days in the aprotinin group and 8 days for patients who did not receive aprotinin (P=0.36). CONCLUSION: The anti-inflammatory properties of aprotinin may be protective against postoperative arrhythmias. Aprotinin does not confer increased risks of prolonged chest tube drainage, renal dysfunction or thrombosis in patients undergoing the Fontan procedure.


Subject(s)
Aprotinin/therapeutic use , Arrhythmias, Cardiac/etiology , Fontan Procedure/adverse effects , Hemostatics/therapeutic use , Pleural Effusion/etiology , Postoperative Complications/etiology , Adolescent , Aprotinin/immunology , Child , Child, Preschool , Drainage , Female , Hemostatics/immunology , Humans , Infant , Male
16.
Clin Appl Thromb Hemost ; 17(6): 620-32, 2011.
Article in English | MEDLINE | ID: mdl-21596694

ABSTRACT

Topical bovine thrombin has been associated with immune responses and anecdotal reports of coagulopathy. This open-label study assessed the impact on clinical hemostasis of human antibodies to bovine thrombin (aBT) or factor V/Va (aBV/Va) in response to topical bovine thrombin (THROMBIN-JMI) in patients both with and without preexisting anti-bovine antibodies. Noninferiority analysis assessed primary endpoint for mean shift from baseline activated partial thromboplastin time (aPTT) at 48 hours postsurgery; secondary endpoints included changes from baseline antibodies/titers and coagulation parameters through 8 weeks postsurgery. A total of 550 patients underwent surgery with THROMBIN-JMI utilized at investigator's discretion. Adjusted mean aPTT change in (+)aBT/(+)THROMBIN-JMI cohort was greater than (-)aBT/(-)THROMBIN-JMI cohort; 4.67-second upper confidence bound exceeded 4.5-second margin (based on assumed mean aPTT of 30 seconds) and noninferiority was not met. Post hoc analysis indicated noninferiority would have been met had noninferiority margin been set prior at relative 15% of actual baseline aPTT. Antibodies/titers were unchanged by THROMBIN-JMI exposure 48 hours postsurgery and unrelated to postsurgical changes in coagulation. Thus, THROMBIN-JMI exposure in patients with/without preexisting aBT or aBV/Va does not alter hemostasis.


Subject(s)
Hemostatics/therapeutic use , Postoperative Complications/prevention & control , Thrombin/therapeutic use , Adult , Aged , Aged, 80 and over , Animals , Cattle , Cohort Studies , Hemostatics/adverse effects , Hemostatics/immunology , Humans , Middle Aged , Postoperative Complications/blood , Postoperative Complications/immunology , Prospective Studies , Thrombin/administration & dosage , Thrombin/adverse effects , Thrombin/immunology , Young Adult
17.
J Pediatr Hematol Oncol ; 33(2): 86-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21228719

ABSTRACT

Postoperative bleeding may occur from a number of etiologies. An uncommon cause of postoperative bleeding is immune-mediated coagulopathy resulting from reexposure to a topical hemostat containing bovine thrombin. Some patients may develop antibodies to bovine thrombin (and other bovine coagulation proteins present in the product) which cross-react with human coagulation proteins, resulting in a coagulopathy, and occasionally, in serious bleeding and death. Most of the clinical information on this coagulopathy is in the adult medical literature. This article reviews the literature on pediatric cases with this coagulopathy and summarizes clinical outcomes and effective therapies.


Subject(s)
Blood Coagulation Disorders/chemically induced , Hemostatics/adverse effects , Postoperative Hemorrhage/chemically induced , Thrombin/adverse effects , Administration, Topical , Adolescent , Animals , Autoantibodies/immunology , Blood Coagulation Disorders/immunology , Cattle , Child , Child, Preschool , Cross Reactions , Hemostasis, Surgical/adverse effects , Hemostasis, Surgical/methods , Hemostatics/administration & dosage , Hemostatics/immunology , Humans , Infant , Postoperative Hemorrhage/immunology , Thrombin/administration & dosage , Thrombin/immunology
18.
J Am Coll Surg ; 211(6): 798-803, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20980172

ABSTRACT

BACKGROUND: Immunoassays that detect antibovine thrombin product antibodies are not widely available. However, knowing whether these antibodies are present preoperatively would be useful because re-exposure to bovine thrombin-containing products is contraindicated in patients with pre-existing antiproduct antibodies due to the risk of developing immune-mediated coagulopathies. In these exploratory analyses, we characterized one aspect of immune sensitization, the persistence of circulating antibodies after exposure to bovine thrombin product. STUDY DESIGN: Elapsed time since a historical surgical procedure with documented or highly likely use of bovine thrombin product was determined for 204 patients enrolled in a recently completed trial. After study completion, baseline samples were assayed for antibovine thrombin product antibodies using validated immunoassays. Antibody data were sorted by time elapsed since the historical procedure. The proportion of patients with antibovine thrombin product antibodies and 95% confidence interval (CI) were determined for each 1-year period, providing an estimate for antibody persistence. RESULTS: Antibovine thrombin product antibodies were detected in 20.7% of patients (23 of 111; 95% CI 14.2%, 29.2%) with ≤1 year since the historical surgical procedure; 6.8% of patients (3 of 44; 95% CI 1.68%, 18.9%) with 1 to <2 years; 16.1% of patients (5 of 31; 95% CI 6.62%, 33.1%) with 2 to <3 years; and 5.6% of patients (1 of 18; 95% CI 0.00%, 27.6%) with ≥3 years since the historical procedure. CONCLUSIONS: The proportion of patients with antibovine thrombin product antibodies ranged from 5.6% to 20.7% across the multiyear postoperative window. Clinicians should be aware that antibodies to bovine thrombin products may persist for years after exposure.


Subject(s)
Antibodies/blood , Hemostasis, Surgical/methods , Hemostatics/administration & dosage , Hemostatics/immunology , Thrombin/administration & dosage , Thrombin/immunology , Administration, Topical , Adult , Aged , Animals , Cattle , Clinical Trials as Topic , Confidence Intervals , Female , Humans , Immunization , Immunoassay , Male , Middle Aged , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Time Factors
19.
Clin Appl Thromb Hemost ; 16(3): 273-80, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20547555

ABSTRACT

It has been reported that patients exposed to topical bovine thrombin preparations may develop antibodies against bovine thrombin, factr V or various other proteins found in these preparations. Such antibodies can cross-react with human endogenous coagulation proteins and may lead to alterations in the coagulation laboratory parameters, hypersensitivity reactions, and severe bleeding or thrombosis. The underlying mechanisms for the coagulopathy are not fully understood yet. To better understand the cross-reactivity of anti-bovine thrombin antibodies with human corresponding coagulation proteins, bovine crude thrombin, and its purified versions, thrombin 4A (the old version of Thrombin-JMI before year 2008) and thrombin 4B (the current version of Thrombin-JMI on market), were used to generate relevant anti-bovine thrombin immunoglobulin G (IgGs) in rabbits. Using Western blotting, the cross-reactivity of each IgG with human alpha-thrombin and a recombinant version of human thrombin (Recothrom) was investigated. The results indicated that no cross-reactivity with either human alpha-thrombin or Recothrom was observed with both anti-bovine crude thrombin IgGs and thrombin 4B IgGs. However, anti-bovine thrombin 4A IgGs showed apparent cross-reactivity with human alpha-thrombin and Recothrom in a protein amount-dependent manner. Furthermore, the results revealed that the cross-reactivity of anti-bovine thrombin 4A IgGs with human alpha-thrombin and Recothrom was immunization time-dependent. The minimum concentration of 4A IgG required to exhibit cross-reactivity with human alpha-thrombin and Recothrom varied considerably among individual rabbits. These results indicate that rabbit anti-bovine thrombin IgGs can cross-react with human alpha-thrombin and Recothrom, suggesting that human antibodies against bovine thrombin may also cross-react with human recombinant thrombin. Thus, the patients who were previously exposed to bovine thrombin may also develop antibodies which can cross-react with human recombinant thrombin.


Subject(s)
Hemostatics/immunology , Immunoglobulin G/immunology , Recombinant Proteins/immunology , Thrombin/immunology , Animals , Antigen-Antibody Reactions , Blotting, Western , Cattle , Cross Reactions , Humans , Immunization , Rabbits , Species Specificity
20.
J Am Coll Surg ; 210(2): 199-204, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20113940

ABSTRACT

BACKGROUND: We evaluated safety and immunogenicity observations pooled from 8 clinical trials of recombinant human thrombin (rThrombin), an active topical hemostatic agent. STUDY DESIGN: Recombinant thrombin was applied with an absorbable gelatin sponge or spray applicator during a surgical procedure (day 1). Adverse events and laboratory parameters were monitored until study end (day 29). Immunogenicity was evaluated after study completion on plasma samples collected at baseline and on day 29. RESULTS: Studies included 583 rThrombin-treated patients (median age, 59 years; 54% men). Surgical procedures included: spinal, 33% of patients; hepatic resection, 14%; peripheral arterial bypass, 23%; arteriovenous graft formation for hemodialysis access, 18%; and skin graft after burn wound excision, 12%. Adverse events reported for >or= 10% patients included incision site pain, procedural pain, nausea, constipation, pyrexia, anemia, insomnia, vomiting, and pruritus. Five of 552 patients developed antibodies to rThrombin (0.9%; 95% CI, 0.3 to 2.1; day 29); antibodies did not neutralize the biologic activity of native human thrombin. At baseline, 12 patients had pre-existing, antibodies recognizing rThrombin (12 of 552; 2.2%; 95% CI, 1.1 to 3.8); these patients had no previous exposure to rThrombin and their antibody titer did not increase >or= 1.0 unit (>or= 10-fold) at day 29. CONCLUSIONS: Results from 8 clinical trials collectively demonstrated that rThrombin is well tolerated in numerous surgical settings when used as a topical adjunct to hemostasis. Adverse events and changes in laboratory parameters were consistent with commonly reported postoperative events. Less than 1% of patients developed antibodies to rThrombin; the antibodies did not neutralize native human thrombin.


Subject(s)
Hemostasis, Surgical , Hemostatics/adverse effects , Hemostatics/immunology , Postoperative Complications , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Thrombin/adverse effects , Thrombin/immunology , Administration, Topical , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies/blood , Child , Clinical Trials as Topic , Female , Hemostatics/administration & dosage , Humans , Male , Middle Aged , Recombinant Proteins/administration & dosage , Thrombin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...