scholarly journals Real-World Early Treatment with Room Temperature–Stable Recombinant Factor VIIa in Hemophilia A/B and Inhibitors: SMART-7™ Post Hoc Analyses

TH Open ◽  
2017 ◽  
Vol 01 (02) ◽  
pp. e130-e138 ◽  
Author(s):  
Francesco Demartis ◽  
Angelika Batorova ◽  
Hervé Chambost ◽  
Peyman Eshghi ◽  
Mehran Karimi ◽  
...  

AbstractTreating hemophilia A or B patients with inhibitors is particularly challenging, as they do not respond to replacement therapy with factor VIII or factor IX concentrates. A room temperature–stable formulation of recombinant activated factor VII (rFVIIa; NovoSeven®), which provides improved convenience and treatment access to patients compared with the earlier formulation of rFVIIa, was shown to be safe and effective in a post-authorization, multinational, observational study (Study Monitoring Antibodies against Room Temperature–stable factor 7 [SMART-7™]). In post hoc, subgroup analyses of SMART-7™ data, the hemostatic response following rFVIIa monotherapy in patients with hemophilia A or B with inhibitors by time to first treatment and in different age cohorts was assessed. A total of 482/618 bleeding episodes treated with rFVIIa monotherapy and with (1) valid efficacy assessment, (2) no missing time for bleed start, (3) no missing time for any dose administration, and (4) valid time to first treatment were included in the analyses. Data on the type and location of bleeding episodes treated with rFVIIa monotherapy were also collected. The majority of bleeding episodes treated with rFVIIa monotherapy were treated within 1 hour after bleeding onset (318/482 [66%]) and, among them, 96.5% (307/318) were effectively treated (i.e., bleeding stopped). Hemostatic efficacy remained high for bleeding episodes treated >1 to ≤4 hours after the onset, with 94/101 (93.1%) treated effectively. Cause and location of bleeding varied across the different age groups assessed. Real-world evidence from post hoc, subgroup analyses of SMART-7™ data confirmed that patients were able to treat themselves quickly and that early treatment with rFVIIa was associated with high efficacy.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1439-1439
Author(s):  
Gary Benson ◽  
Hervé Chambost ◽  
Francesco Demartis ◽  
Soraya Benchikh El Fegoun ◽  
Katarina Cepo ◽  
...  

Abstract Introduction: SMART-7™ (NCT01220141) was a prospective, post-authorization, single-arm, multinational, multi-center, non-interventional study investigating the safety and effectiveness of room temperature stable rFVIIa (NovoSeven®) in patients with hemophilia A or B with inhibitors in a real-world setting. Here, we present a subgroup analysis of the hemostatic response of bleeding episodes to treatment with rFVIIa. Methods: Study medication was not provided; use was at the discretion of the treating physician in accordance with the local label. Bleeding history was collected at the initiation visit. Information on bleeding episodes, including home treatment, was recorded in patient diaries during the study. Patients evaluated the status of bleeding episodes after treatment as 'bleed stopped', 'bleed slowed', or 'no change/worsened'. Overall bleed outcome was defined as the last given patient evaluation of the bleed. Patients were observed until they had achieved ≥25 exposure days, defined as any exposure to rFVIIa during one 24 h period. Results: From November 2010 to March 2015, 51 patients were enrolled at 24 sites in 14 countries; 31 (60.8%) patients completed the study and 20 discontinued (11 of these due to global study discontinuation, a decision endorsed by the European Medicines Agency). Patients were aged 1.6-69.5 years (median 22.0 years) with an historical median bleeding rate of 1.0 episode per month. During the study, 48 patients experienced a total of 618 bleeding episodes: median number of bleeding episodes per patient was 10.5; median study duration per patient was 13.9 months. 63.4% of bleeding episodes occurred spontaneously and 31.2% were categorized as traumatic. Effectiveness evaluation at end of treatment was available for 609 bleeding episodes: 569 (93.4%) resolved, 35 (5.7%) slowed and 5 (0.8%) were unchanged/worsened. Nine bleeds had no reported outcome. A total of 538 bleeds were treated with rFVIIa monotherapy: 507 (94.2%) resolved, 27 (5.0%) slowed and 4 (0.7%) were unchanged/worsened. In a post-hoc analysis in which the data were divided by time to first treatment into 3 groups, the best hemostatic response (96.5%) was observed when rFVIIa treatment was initiated ≤1 h after onset of bleed; effectiveness was also high (93.1%) for bleeds treated >1-≤4 h of onset, decreasing for those treated >4 h after onset (87.3%; representing 13.1% of bleeds). Early treatment (≤1 h) with rFVIIa monotherapy was effective for both joint and muscle bleeds (96.2% and 97.3%, respectively). The initial rFVIIa dose administered was comparable for bleeds treated ≤1 h and those treated >1-≤4 h or >4 h after onset (Table). The median number of rFVIIa doses was higher for bleeds treated >4 h after onset than for those treated within 4 h, however these data should be interpreted with caution due to the low number of bleeds represented (13.1% of total) (Table). Conclusion: A subanalysis of the SMART-7™ study demonstrates higher effectiveness of early treatment with rFVIIa, with 96.5% of bleeds resolved when treatment was initiated ≤1 h after bleed onset, and remaining high (93.1%) for bleeding treatment >1-≤4 h. Total effectiveness was >87% in bleeds treated >4 hours after bleed onset. Disclosures Benson: Novo Nordisk: Consultancy. Benchikh El Fegoun:Novo Nordisk: Employment. Cepo:Novo Nordisk: Employment. Sommer:Novo Nordisk: Employment. Kavakli:Novo Nordisk: Other: Steering committee meetings.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4980-4980 ◽  
Author(s):  
Ekaterina Shiller ◽  
Victor Petrov ◽  
Pavel Svirin ◽  
Vladimir Vdovin ◽  
Igor Koltunov ◽  
...  

Abstract Background: Recent studies have shown that addition of bypassing agents to immuno-tolerance induction (ITI) protocol for patients with hemophilia A and inhibitor results in better control of bleeding episodes and improves quality of life. Few publications have addressed prophylactic usage of recombinant factors VIIa in these settings. Due to relatively low infusion volume, convenience of administration and high efficacy rFVIIa - Coagil-VII seems to be especially reasonable for ITI protocol. Aim: To assess the efficacy and safety of rFVIIa - Coagil-VII (SJC "GENERIUM", Russia) for prophylactic use during ITI protocol in patients with hemophilia A and inhibitor. Methods: Seven patients aged between 2 to 7 years with severe hemophilia A and inhibitor have been treated with ITI protocol using plasma derived factor VIII with von Willebrand factor. Seven of them simultaneously received treatment with Coagil-VII in individual doses (100-250 mkg/kg) and regimens (every 12 - 48 hours). When inhibitor reached level of 3 BU (Bethesda Unit) either dose or frequency of Coagil-VII administration were gradually reduced. After 1 BU the patients were given factor VIII only. Number and severity of bleeding events were assessed. Results: Five patients with high responding inhibitors and poor prognosis (history of high titer of factor VIII inhibitor, prolonged time between first inhibitor appearance and the beginning of ITI) received Coagil-VII in high doses 150 - 250 mkg/kg every 12-24 hours in 1-4 years. At time of booster effect titer of inhibitors reached 92 -16 000 BU. One of patients had ITI failure because of interruption of protocol, while 4 patients continue treatment. Level of 3 BU was reached by 4 patients at 40, 12, 35, and 3 months of treatment. Level of 1 BU was reached by 2 patients at 6 and 42 months of treatment. Significant clinical effect was achieved after 6 months of treatment. Time of bleeding episode was decreased from 7 (±2) to 2 (±1) days. Total number of hemorrhagic events, including hemarthrosis, hematomas and bleedings decreased by 3,7 fold (3,7 events per patient-month during first 6 months versus 1,0 events per patient-month). Only 1 hospital admission with bone fracture was recorded. All children have an active lifestyle and attend school. Two patients with low responding inhibitor and good prognosis received Coagil-VII in low doses 90 - 170 mkg/kg every 24-48 hours. Maximal titer of inhibitor was 1.3 - 1.9 BU. Both patients completed treatment with Coagil-VII in 2 and 4 months and continue ITI protocol and both achieved undetectable level of inhibitor. No bleeding episodes were recorded in these patients since the beginning of treatment. There was no clinical or laboratory evidence of thrombosis, thrombocytopenia, or disseminated intravascular coagulation. Conclusion: We report our experience of prolonged (2 months - 4 years) prophylactic treatment with recombinant activated factor VII (rFVIIa) - Coagil-VII in patients with hemophilia A and inhibitor. This prophylaxis is efficacious when doses and treatment regimens are individually determined. This approach results in reduction of bleeding episodes in all patients, as well as increase of quality of life. No any adverse events (AE and SAE) with prolonged use of Coagil-VII have been registered so far. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2397-2397
Author(s):  
Assaf Arie Barg ◽  
Tami Livnat ◽  
Tami Brutman-Barazani ◽  
Nurit Rosenberg ◽  
Ivan Budnik ◽  
...  

Background: Hemlibra is a bispecific antibody that bridges factor IXa and factor X to restore hemostasis in patients with Hemophilia A (HA). It has proven efficacy and safety in multicenter trials. However, real world data is currently lacking. Ancillary tests' results for monitoring Hemlibra's hemostatic effect are scarce. Aim: To evaluate laboratory monitoring and any clinical correlations to hemostasis in patients with HA who initiate prophylactic treatment with Hemlibra, per standard protocol. Methods: Any severe HA patient with or without inhibitors treated by Hemlibra and followed by our National Hemophilia Center was eligible for the study, as approved by our institutional review board. The first two subcutaneous loading injections (3 mg/kg body weight given once weekly for 4 weeks) were administered in the clinic, as was the first post-loading maintenance dose (1.5 mg/kg given once weekly) injection. The patients were instructed to contact and consult the center about any trauma, bleeding or other adverse events. Bleeding episodes as well as any surgical intervention were documented. Blood samples were obtained before initiation of therapy, during the loading period (week 2), and following the initiation of maintenance therapy (week 5). Platelet-poor plasma (PPP) was obtained and activated partial thromboplastin time (aPTT), FVIII activity and inhibitor Bethesda units (BU) assay were performed. Hemlibra levels were evaluated as previously described.1 Thrombin generation (TG) was measured in PPP and thrombin peak height and endogenous thrombin potential (ETP) were calculated.2 Results: Forty patients with HA, median age 10 years (range 6 month- 76 years) were enrolled. The group consisted of 25 children and 15 adults, of whom 18 patients had FVIII inhibitors (median 16, range 1-900BU) and 22 were without inhibitors, including 9 patients with previous history of inhibitor. Patients were clinically followed for a median of 18 weeks (range 9-76 weeks). During follow-up only one patient experienced spontaneous bleeding episodes. Hemarthroses (mainly target joints) occurred in 4/40 patients (1 child only) and post traumatic bleeds were documented in 8 patients. However, 17/40 experienced trauma that did not cause any bleeding. For 32/40 patients, Hemlibra prophylaxis was sufficient to maintain hemostasis without additional supplemental therapy. Five minor surgeries were safely performed in 4 children (2/5 without supplemental therapy), yet another procedure (circumcision of a 3-months-old baby) was complicated by major bleeding. Laboratory analyses, presented as median (interquartile range), disclosed statistically significant increase of Hemlibra plasma levels from 19 (15-22) µg/ml to 50 (42-57) µg/ml between week 2 and week 5, respectively (Fig 1A). The extended aPTT values measured before treatment of 85(61-127) sec were normalized at week 2 [28 (26-30) sec] with additional significant shortening at week 5 [23 (22-25) sec; Fig 1B]. Both ETP and peak height significantly increased from baseline 43 (0-374) nM×min and 14 (6-22) nM to 700 (202-1043) nM×min and 47 (12-76) nM after 2 weeks and further to 981 (476-1396) nM×min and 72 (35-100) nM after 5 weeks; however, TG did not reach the levels observed in normal controls [ETP 1594 (1505-1722) nM×min and peak height 221 (199-273) nM]; Fig 1C,D. No differences were found between adults and children or between inhibitor and non- inhibitor patients yet notably, initial aPTT was significantly prolonged among patients with inhibitors as compared to non- inhibitor patients and the same difference persisted at week 2 and disappeared at week 5. No differences were noted between patients experiencing any bleeding or non- bleeders, probably as most bleeding episodes were trauma related. Positive correlations were found between aPTT, Hemlibra levels and TG parameters. Notably lower TG was observed in very young infants, thus interpretation of laboratory results in this age required caution. Conclusion: This study confirms the safety and efficacy of Hemlibra prophylaxis in patients with HA, including young infants. Laboratory analyses prove that Hemlibra loading, results in higher drug levels and correlates with aPTT shortening and improved TG parameters. While aPTT normalizes during Hemlibra loading, TG is still lower than the normal range observed in controls and may be a more sensitive ancillary test to predict patients' hemostasis. Disclosures Kenet: Roche: Consultancy, Honoraria; Bayer: Consultancy, Honoraria, Research Funding; Opko Biologics: Consultancy, Honoraria, Research Funding; CSL: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria, Research Funding; Shire: Consultancy, Honoraria, Research Funding; Alnylam: Consultancy, Honoraria, Research Funding; BPL: Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 39-40
Author(s):  
Linda Neuman ◽  
Shraddha Desai ◽  
Tom Knudsen ◽  
Frank Del Greco ◽  
Howard Levy

Background Hemophilia A (HA) and HB are X-linked bleeding disorders caused by a deficiency of Factor VIII (HA) or Factor IX (HB). A significant number of individuals with HA and HB develop inhibitors against the wild-type FVIII or FIX, respectively, and thereby become refractory to factor replacement treatment. Treatment of episodic bleeding in these individuals requires technical expertise to gain the requisite intravenous (IV) access and is often associated with pain. This causes delays in administration, which results in prolonged bleeding and accumulation of blood in the joint or other bleeding site. Currently approved FVIIa containing products take 6-24 hours to achieve hemostasis and maintain efficacy. Marzeptacog alfa (activated) (MarzAA), a novel variant activated recombinant Factor VII (rFVIIa), is being developed for subcutaneous (SQ) administration to address this important unmet need. Data from the Phase 1 study, MAA-102, demonstrated that SQ MarzAA is quickly absorbed and has the potential to achieve and maintain plasma levels in the desired range with an acceptable safety profile. This open-label, global, multi-center, randomized, cross-over Phase 3 study will evaluate the efficacy and safety of MarzAA for on demand treatment of spontaneous or traumatic bleeding episodes in adolescents and adults with congenital HA or HB with inhibitors. MarzAA will be compared to the Standard of Care (SOC). Approximately 60 subjects will be enrolled at 54 sites in 19 countries. Study Design and Methods Key inclusion criteria: Eligible male or female subjects ≥ 12 years of age must have: confirmed diagnosis of HA or HB unresponsive to or intolerant of standard replacement therapy; history of bleeding with annualized bleeding rate of ≥ 8; investigator-confirmed subject's ability to identify bleeding episodes and administer MarzAA SQ and infuse SOC IV at home. Key exclusion criteria: Subjects should not have had previous exposure to SQ administration of rFVIIa or exposure to any other variant rFVIIa; known positive antibody or hypersensitivity to MarzAA, FVIIa, or variants thereof; history of other coagulation disorder(s); platelet count <50,000/µL; CD4 T cell count <200 cells/mm3. Treatment: Subjects >17 years of age will be randomized in blocks to target treatment of approximately 5 bleeds with MarzAA and an additional 5 bleeds with SOC in either order, Sequence A or Sequence B, with a washout period to assess anti-drug antibodies (ADA) status between treatments. A total of 488 eligible bleeds will be treated, ~244 bleeds with each treatment, until ≥80% of patients have ≥3 bleeds treated with MarzAA and SOC. Pharmacokinetic samples will be collected during the MarzAA treatment period. Subjects 12-17 years of age will be enrolled after a positive DSMB assessment of 30 eligible bleeds treated with MarzAA. Subjects will administer 60 µg/kg SQ dose of MarzAA, at 3-hour intervals, for up to 3 doses as required to achieve hemostasis. Endpoints: Primary endpoint includes percentage of treated bleeds resulting in effective hemostasis (excellent or good) at 24 hours after the initial dose. Key secondary endpoints include time to cessation of bleeding after the initial dose; percentage of treated bleeds resulting in effective hemostasis at fixed timepoints; percentage of those that maintain hemostasis at 48 hours after the initial dose; use and amount of rescue therapy. Key safety endpoints include the incidence of adverse events; occurrence of thrombotic events and binding and/or neutralizing anti-drug antibodies. Key exploratory endpoints include assess patient satisfaction with the Treatment Satisfaction Questionnaire for Medicine- 9; assessment of pain using the Visual Analogue Scale; time required to administer treatment. Statistics: The goal is to demonstrate the true proportion of bleeds effectively treated with MarzAA is not inferior to SOC. Using a baseline efficacy of 85% for SOC, with a -12% margin of non-inferiority, treatment of 244 bleeds in each group provides 90% power with a one-sided 2.5% significance level to substantiate this result. Figure Disclosures Neuman: Catalyst Biosciences: Current Employment, Current equity holder in publicly-traded company. Desai:Catalyst Biosciences: Current Employment, Current equity holder in publicly-traded company. Knudsen:Catalyst Biosciences: Current Employment, Current equity holder in publicly-traded company. Del Greco:Catalyst Biosciences: Current Employment, Current equity holder in publicly-traded company. Levy:Catalyst Biosciences: Current Employment, Current equity holder in publicly-traded company.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 573-573 ◽  
Author(s):  
Johnny N. Mahlangu ◽  
Pei Lin Koh ◽  
Heng Joo Ng ◽  
Toshko Lissitchkov ◽  
Marion Hardtke ◽  
...  

Abstract Introduction Up to 30% of patients with hemophilia A and 5% of patients with hemophilia B develop neutralizing antibodies (inhibitors) against replacement factor VIII or factor IX, respectively. Acute bleeding episodes in these patients with inhibitors are treated with bypassing agents, which include activated recombinant factor VII (rFVIIa). BAY 86-6150 is a modified rFVIIa which in preclinical studies was shown to have prolonged half-life and improved potency compared with currently available rFVIIa. In a phase 1, randomized, double-blind trial, BAY 86-6150 was not associated with any clinically significant adverse events (AEs). We report the immunologic response to BAY 86-6150 in a phase 2/3 clinical trial in patients with hemophilia with inhibitors. Methods TRUST (Treatment with Unique Recombinant FVII Study) was a multicenter, open-label, 2-part study (part A and part B) which included males aged 12−62 years with moderate or severe hemophilia A or B, with a history of high-titer inhibitors (≥5 Bethesda units), and ≥4 bleeding episodes in the 6 months prior to enrollment. Part A was a sequential dose-escalation study of 4 BAY 86-6150 dose levels (6.5, 20, 50, and 90 μg/kg body weight; n≥10/cohort). Dose escalation was dependent on both efficacy and an Independent Data Monitoring Committee (IDMC) approval of safety in 10 patients per cohort who had ≥1 bleeding episode treated with BAY 86-6150. Part B was designed as a single-arm investigation of the efficacy and safety of the recommended dose of BAY 86-6150 determined in all patients from Part A. Safety endpoints were AEs and immunogenicity. Anti-drug antibody testing was performed at screening (prior to exposure), after the second exposure, then every fifth exposure, and at the end of study visit in both part A and part B. Anti-BAY 86-6150 binding antibodies were measured using a validated enzyme-linked immunosorbent assay (ELISA). Samples that revealed a specific immunoreactivity in this assay were further characterized for neutralizing activity using a validated platelet-activated clotting assay. Additional functional assays were performed to determine the cross-reactive neutralizing effect on rFVIIa (NovoSeven®) of any detected anti-BAY 86-6150 antibodies. The presence of neutralizing antibodies was considered a serious adverse event (SAE) requiring prompt IDMC review. Results In cohort 1, 10 patients (mean age, 27.4 years) were treated with 6.5 mg/kg BAY 86-6150. These patients had a total of 73 bleeding events and received a total of 84 study drug injections. No anti-drug antibodies or anti-FVIIa was detected in the patients at screening prior to exposure to the study drug. BAY 86-6150 was well tolerated in all patients with no clinical or laboratory symptoms or signs of venous thromboembolism. Binding antibodies to BAY 86-6150 were detected on a scheduled screening visit in 1 patient after 3 exposures to BAY 86-6150; these anti-BAY 86-6150 antibodies displayed neutralizing activity against BAY 86-6150 and were also cross-reactive and neutralizing for rFVIIa. The affected patient had received rFVIIa before entry into the study. At the time of diagnosis of binding and neutralizing antibodies, the affected patient was not bleeding and did not require emergency treatment. Exposure to BAY 86-6150 was stopped and the trial was terminated at the first cohort. Subsequent bleeding episodes in this patient were successfully managed with FEIBATM (Factor Eight Inhibitor Bypass Activity). No other treatment-related AEs or SAEs were reported in this study. Additionally, the IDMC has recommended safety follow-up assessments for all the patients who actively participated in the trial. Conclusions The TRUST trial has been discontinued as a precautionary measure because of potential safety concerns related to the detection of the antidrug antibodies in 1 patient. Development of neutralizing antibodies against BAY 86-6150 that had a cross-reactive neutralizing effect on rFVIIa was considered a serious risk because of the limited treatment options in patients with inhibitors. These results underline the fact that it is currently not possible to predict immunologic response based on preclinical and phase 1 studies. Disclosures: Hardtke: Bayer Pharma AG: Employment. Schroeder:Bayer Pharma AG: Employment.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4299-4299 ◽  
Author(s):  
Mohammad Faranoush ◽  
Hassan Abolghassemi ◽  
Gh Toogeh ◽  
Mehran Karimi ◽  
Peyman Eshghi ◽  
...  

Abstract Introduction: Concentrated factor VIII replacement therapy in severe hemophilia prevents disabling arthropathy, life-threatening bleeding, improves health related quality of life and increase life expectancy. Patients with neutralizing antibodies (inhibitors) did not respond to usual doses of FVIII and they need higher doses of FVIII or bypassing agents (eg. rFVIIa, FEIBA) to control bleeding. The aim of this study was to compare the efficacy of a new biosimilar recombinant factor VIIa (Aryoseven ™) with Novoseven® in controlling bleeding episodes in hemophilia A patients with inhibitors. Methods: A randomized double blind clinical trial study was conducted in eight comprehensive hemophilia care centers in Iran. We randomized 66 male patients in two groups, with 4 consecutive block randomization when bleeding occurred. All patients entered the study with only one bleed. Informed consent signed by all patients or their parents. Group A (31 patients, 47%) received Aryoseven ™ and group B (35 patients, 53%) received Novoseven®. rFVIIa dosage was 90 -120 μg/kg intravenously every 2 hours. Results: During the study 66 patients (All male) were enrolled in two arms. The mean age was 20.5±10.9 years. Comparison of baseline data between the two showed no significant difference. The distributions of these variables were similar between the two groups. Other factors, such as vital signs, coagulation tests, liver, renal function tests, and blood count were not significantly different between the two groups. Median plasma level of FVII clotting activity (FVII: C) in group A and B was 103.8±38.4 IU/ dl and 98.6±26.7 IU/dl before injection respectively. Median plasma level of FVIII inhibitor in group A and B was 15.0 BU (IQR: 8.8-58.0) and 19.0 BU (IQR: 11.0-31.0). The average time from onset of bleeding to start treatment were 1246± 1104 and 2301 ± 1693 minutes (p = 0.311) in group A and B respectively. This study results showed that increased levels of factor VII were comparable between two groups after rFVIIa injection. A comparison of the Kavakli global response scores after injection and the treatment success rate in terms of achieving to score 6 or higher showed that both groups were comparable in treatment success rates. The global treatment response rate was 96.8% in group A and 91.4% in group B. Administration of either Aryoseven™ or Novoseven® had comparable effect on controlling pain and joint mobility. Reported side effects were minor (headache, nausea, and rash) and occurred in similar frequency. Discussion: The present study showed increased FVII levels and clinical efficacy in the control of the bleeding episodes in Hemophilia A patients with inhibitor for both drugs. Some similarities exist between our findings and the previous clinical trials designed for Novoseven®. In other studies, the treatment success rate by using the global response scoring system has been 70% -86% by different rFVIIa dosage regimen, in our study was above 90% in both treatment groups. In most studies, it was reported more than 90% achieving to cessation of bleeding at 9 hours after first injection of rFVIIa as response time which was also similar to our study. Conclusion: This study showed non-inferiority of Aryoseven ™ compared to Novoseven® in cessation of bleeding episodes in hemophilia A patients with inhibitor. There was no evidence of significant difference between the efficacy and safety of two drugs. There were no clinically meaningful differences between the biosimilar product and the original product in terms of the safety, purity, and efficacy. Biosimilar rFVIIa is effective in treatment of acute joint bleeding in patients with inhibitor in comparison to Novoseven®. Disclosures Mehrvar: Aryogen Zist Darou: Employment. Khoein:Aryogen Zist Darou: Employment. Kaymar:Aryogen Zist Darou: Employment. Mahbodi:Aryogen Zist Darou: Employment. Vaziri:Aryogen Zist Darou: Employment.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3028-3028
Author(s):  
Katharine Batt ◽  
Bob G Schultz ◽  
Jorge Caicedo ◽  
Christopher S Hollenbeak ◽  
Neha Agrawal ◽  
...  

Abstract Background Hemophilia A (HA) is a rare genetic disease characterized by a deficiency in clotting factor VIII (FVIII). Persons with HA suffer from spontaneous and traumatic bleeds which significantly impact short- and long-term quality of life. Prophylaxis treatment with FVIII replacement or non-factor replacement (e.g. emicizumab) intends to prevent bleeding episodes. To date, clinical comparisons between FVIII and emicizumab are limited to non-interventional studies and indirect comparisons. Comparisons of costs are limited to cost-effectiveness models or observational studies that include patients with and without inhibitors. An increase in availability of real-world data since emicizumab's approval in 2018 has created opportunity for comparative outcomes research in the non-inhibitor HA population. Objective To compare billed annualized bleed rates (ABR b) and all-cause costs (ACC) among non-inhibitor HA patients switching from prophylaxis with FVIII replacement to emicizumab. Methods This retrospective, observational, pre-post study used the IQVIA PharMetrics® Plus database (2015-2020)-a large longitudinal US commercial health plan database with over 190 million lives. International Classification of Diseases codes (ICD-10), National Drug Codes, and Healthcare Common Procedure Coding System were used to identify diagnoses, therapies, and procedures. Males with ≥1 claim for emicizumab who were on prophylaxis treatment with FVIII prior to initiating emicizumab were included in the analysis. Patients who received bypassing agents, immune tolerance induction, or rituximab were assumed to have inhibitors and were excluded. Patients with ≥2 occurrences of any of the following diagnoses were excluded: von Willebrand disease, hemophilia B, acquired HA, or other coagulation disorders. Annualized bleed rate was defined as billed ABR and represents bleeding episodes that required evaluation, treatment, or procedure resulting in an ICD-10 claim. Therefore, bleeds treated at home and untreated bleeds were not captured. A clinical review of ICD-10 codes resulted in a list of 535 codes used to identify HA-related bleeding episodes (e.g. hemarthrosis). The ACC were calculated as the mean cost per patient per year in 2020 US dollars actually paid by the insurer. Descriptive statistics were used to summarize, and Bayesian models were developed to compare, ABR b and ACC in the pre- and post-switch periods. Bayesian inferences estimated the population mean difference in ABR b and ACC after switching from FVIII prophylaxis to emicizumab. Inferences were conducted by computing posterior probabilities for hypotheses and summarized with 95% credible intervals (CrI). Results A total of 121 patients were included with mean age [range] of 25.9 [2-63] years. The majority of patients were over the age of 18 (60.3%), 33.1% were ≥7-18, and 6.6% were <7. The mean (SD) years on FVIII replacement (pre-switch) and emicizumab (post-switch) were 2.5 (1.5) and 1.1 (0.4), respectively (Table 1). Descriptive In the majority of patients, ABR b remained unchanged from pre-switch to post-switch (42%) while 38% had some magnitude of improvement, and 20% experienced a worsening of ABR b. The mean observed ABR b and ACC were 0.68 and $518,151, respectively, in the pre-switch period, and 0.55 and $652,679, respectively, in the post-switch period. Bayesian Model The Bayesian model demonstrated a mean change in ABR b of -0.128 [95% CrI: -0.441 to 0.184] after switch (Table 2). The mean change in ACC was +$159,680 [95% CrI: $74,842 to $247,841] after switch. The model determined there is a 21.0% probability ABR b will worsen after switch and a 99.9% probability ACC will increase after switch. Conclusions Prophylaxis with FVIII replacement and emicizumab result in similar prevention of billed bleeds in a real-world switch population. Although the population mean ABR b is more likely to fall after switching from FVIII replacement to emicizumab, there is only a 1.02% posterior probability the population mean ABR b will fall by ≥0.5 after switching to emicizumab and a 21.0% probability the ABR b will worsen after switch. Additionaly, ACC are almost certain to substantially increase after switching to emicizumab (99.9%). As additional real-world data becomes available in the non-inhibitor HA population, further research should help to strengthen clinical and economic outcomes for different prophylaxis treatment options. Figure 1 Figure 1. Disclosures Batt: Sanofi: Current equity holder in publicly-traded company; Bayer Therapeutics: Consultancy; Sprouts Consulting: Other: CEO, Principal Consultant; Merck: Current equity holder in publicly-traded company; Forma: Consultancy, Current equity holder in publicly-traded company; Precision Health: Consultancy; Takeda Pharmaceuticals U.S.A.: Consultancy. Schultz: Takeda Pharmaceuticals U.S.A., Inc.: Current Employment, Current holder of individual stocks in a privately-held company. Caicedo: Takeda Pharmaceuticals U.S.A., Inc.: Current Employment, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Hollenbeak: Takeda Pharmaceuticals U.S.A., Inc.: Consultancy. Agrawal: Takeda Pharmaceuticals U.S.A., Inc.: Consultancy. Chatterjee: Takeda Pharmaceuticals U.S.A., Inc.: Consultancy. Dayma: Takeda Pharmaceuticals U.S.A., Inc.: Consultancy. Bullano: Takeda Pharmaceuticals U.S.A., Inc.: Current Employment, Current holder of individual stocks in a privately-held company.


2021 ◽  
Vol 10 (19) ◽  
pp. 4303
Author(s):  
Sarina Levy-Mendelovich ◽  
Tami Brutman-Barazani ◽  
Ivan Budnik ◽  
Einat Avishai ◽  
Assaf A. Barg ◽  
...  

Emicizumab (Hemlibra™) is approved for prophylaxis of hemophilia A (HA) patients. The HAVEN studies addressed bleeding reduction in emicizumab-treated patients, but real-world data on bleeding patterns during emicizumab therapy are lacking. We aimed to compare the occurrence of breakthrough bleeding at different time points, starting from emicizumab initiation. This longitudinal prospective observational cohort study included HA patients (n = 70, aged 1 month to 74.9 years) that completed at least 18 months of follow-up in our center. We analyzed the number of spontaneous and traumatic bleeds during selected time points of the study (“bleeding periods”). The percentage of traumatic and spontaneous bleeding episodes was not significantly different among “bleeding periods” (P = 0.053 and P = 0.092, respectively). Most trauma-related treated bleeds resulted from either hemarthrosis (53%) or head trauma (33%). Spontaneous bleeding episodes were mostly hemarthroses (80%). Potential associations of the patients’ age, annualized bleeding rate before emicizumab treatment, and the presence of inhibitors with spontaneous bleed occurrence were analyzed with binomial logistic regression. The odds of bleeding while on emicizumab increased by a factor of 1.029 (P = 0.034) for every one year of age. Conclusions: Our real-world data revealed that the risk of bleeding persists, especially in older patients, despite therapy with emicizumab. These data may help clinicians in counselling their patients and in planning their management.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. lba-4-lba-4
Author(s):  
Paris Margaritis ◽  
Elise Roy ◽  
Majed N Aljamali ◽  
Harre Downey ◽  
Shangzhen Zhou ◽  
...  

Abstract Recombinant human activated Factor VII (rhFVIIa) is extensively used in the management of hemophilic inhibitor patients. However, the short plasma half-life and need for repeated infusions are amongst the limiting factors for its use in prophylaxis. In an attempt to overcome these, we have designed a gene transfer approach using continuous expression of a modified FVII transgene that is secreted in the active form, FVIIa. In proof-of-concept experiments, we previously demonstrated that Adeno-associated virus (AAV)–mediated gene transfer of the modified Factor VII gene can result in long-term phenotypic correction in hemophilia B mice. However, a prerequisite to a human application is the demonstration of efficacy as well as safety in a large animal model of hemophilia. Hence, we utilized the canine hemophilia model that closely resembles the human disease and has been shown to be an excellent predictor of efficacy of treatments for hemophilia in humans. As we previously reported (Blood 110 (11), p65a. 2007), we generated a canine FVII transgene that is expressed and secreted in a two-chain, activated form (cFVIIa). An AAV serotype 8 vector directing expression of this transgene was delivered via the portal vein into 4 hemophilia dogs, at vector doses of 2.06 E13 (one hemophilia B dog) − 1.25 E14 (three hemophilia A dogs) vector genomes (vg)/kg. The current abstract presents a rigorous characterization of the clotting parameters in the treated dogs, clinical safety and efficacy results of a substantially longer follow-up, and a laboratory safety profile of continuous expression of canine FVIIa. Gene expression in the treated dogs was followed by antigen and clotting assays (prothrombin time [PT], whole blood clotting time [WBCT]) as well as rotational thromboelastography (TEG). The initial dose of 2.06 E13 vg/kg resulted in a modest and transient reduction in the PT, WBCT and TEG parameters over a period of >31 months, accompanied by marginal changes in cFVIIa antigen levels. However, a considerable and sustained reduction in PT (supra-physiologic), WBCT (partial correction) and TEG parameters (near-normalization) was observed following administration of 6.25 E13 – 1.25 E14 vg/kg in three hemophilia A dogs in a cumulative total of 42 months of observation. Antigen levels in the treated hemophilia A dogs were in the range of 1–2.5 μg/ml. As untreated hemophilic dogs exhibit 5–6 spontaneous bleeds per year, we utilized this as an important efficacy endpoint. None of the treated dogs (including the lowest dose hemophilia B dog) exhibited any spontaneous bleeding episodes (34 expected within the observation period) in contrast to concurrently observed controls, which exhibited clinically detectable bleeding episodes at the predicted rate. Given the procoagulant nature of cFVIIa, the safety profile of this approach was thoroughly investigated. We did not observe any abnormal changes in serum chemistries, platelet counts, levels of D-dimer, fibrinogen or thrombin-antithrombin, arguing against a procoagulant state following cFVIIa-gene transfer. In summary, our results demonstrate for the first time that AAV8 vector doses of up to 1.25 E14 vg/kg (2.6 E15 vg total) are well-tolerated in hemophilic dogs; continuous expression of FVIIa at levels of 1–2.5 μg/ml is safe in hemophilic dogs, and can result in a sustained, measurable improvement in the clinical bleeding course of these animals. These data suggest that this model approach of a gene-based Factor VIII/Factor IX bypassing agent has therapeutic potential for hemophilia as has been shown for bolus infusion of rhFVIIa


Sign in / Sign up

Export Citation Format

Share Document