Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Blood ; 143(22): 2256-2269, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38452197

RESUMO

ABSTRACT: Fitusiran, a subcutaneous investigational small interfering RNA therapeutic, targets antithrombin to rebalance hemostasis in people with hemophilia A or B (PwHA/B), irrespective of inhibitor status. This phase 3, open-label study evaluated the efficacy and safety of fitusiran prophylaxis in males aged ≥12 years with hemophilia A or B, with or without inhibitors, who received prior bypassing agent (BPA)/clotting factor concentrate (CFC) prophylaxis. Participants continued their prior BPA/CFC prophylaxis for 6 months before switching to once-monthly 80 mg fitusiran prophylaxis for 7 months (onset and efficacy periods). Primary end point was annualized bleeding rate (ABR) in the BPA/CFC prophylaxis and fitusiran efficacy period. Secondary end points included spontaneous ABR (AsBR) and joint ABR (AjBR). Safety and tolerability were assessed. Of 80 enrolled participants, 65 (inhibitor, n = 19; noninhibitor, n = 46) were eligible for ABR analyses. Observed median ABRs were 6.5 (interquartile range [IQR], 2.2-19.6)/4.4 (IQR, 2.2-8.7) with BPA/CFC prophylaxis vs 0.0 (IQR, 0.0-0.0)/0.0 (IQR, 0.0-2.7) in the corresponding fitusiran efficacy period. Estimated mean ABRs were substantially reduced with fitusiran by 79.7% (P = .0021) and 46.4% (P = .0598) vs BPA/CFC prophylaxis, respectively. Forty-one participants (63.1%) experienced 0 treated bleeds with fitusiran vs 11 (16.9%) with BPAs/CFCs. Median AsBR and AjBR were both 2.2 with BPA/CFC prophylaxis and 0.0 in the fitusiran efficacy period. Two participants (3.0%) experienced suspected or confirmed thromboembolic events with fitusiran. Once-monthly fitusiran prophylaxis significantly reduced bleeding events vs BPA/CFC prophylaxis in PwHA/B, with or without inhibitors, and reported adverse events were generally consistent with previously identified risks of fitusiran. This trial was registered at www.ClinicalTrials.gov as #NCT03549871.


Assuntos
Hemofilia A , Hemofilia B , Hemorragia , Humanos , Masculino , Hemofilia B/tratamento farmacológico , Hemofilia B/complicações , Adulto , Hemofilia A/tratamento farmacológico , Hemofilia A/complicações , Pessoa de Meia-Idade , Adolescente , Adulto Jovem , Hemorragia/induzido quimicamente , Hemorragia/prevenção & controle , Criança , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Fatores de Coagulação Sanguínea/uso terapêutico , Fatores de Coagulação Sanguínea/administração & dosagem , Idoso
2.
Lancet Haematol ; 10(5): e322-e332, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37003278

RESUMO

BACKGROUND: Fitusiran, a subcutaneous investigational siRNA therapeutic, targets antithrombin with the goal of rebalancing haemostasis in people with haemophilia A or haemophilia B, regardless of inhibitor status. We aimed to evaluate the efficacy and safety of fitusiran prophylaxis in people with severe haemophilia without inhibitors. METHODS: This multicentre, open-label, randomised phase 3 study was conducted at 45 sites in 17 countries. Male participants aged at least 12 years with severe haemophilia A or B without inhibitors, who had previously been treated on-demand with clotting factor concentrates, were randomly assigned in a 2:1 ratio to receive 80 mg subcutaneous fitusiran prophylaxis once per month or to continue on-demand clotting factor concentrates for a total of 9 months. Randomisation was stratified by the number of bleeding events in the 6 months before screening (≤10 bleeds and >10 bleeds) and by haemophilia type (haemophilia A or B). The primary endpoint was annualised bleeding rate, analysed in the intention-to-treat analysis set. Safety and tolerability were assessed in the safety analysis set. This trial is registered with ClinicalTrials.gov, NCT03417245, and is complete. FINDINGS: Between March 1, 2018, and July 14, 2021, 177 male participants were screened for eligibility and 120 were randomly assigned to receive fitusiran prophylaxis (n=80) or on-demand clotting factor concentrates (n=40). Median follow-up was 7·8 months (IQR 7·8-7·8) in the fitusiran group and 7·8 months (7·8-7·8) in the on-demand clotting factor concentrates group. The median annualised bleeding rate was 0·0 (0·0-3·4) in the fitusiran group and 21·8 (8·4-41·0) in the on-demand clotting factor concentrates group. The estimated mean annualised bleeding rate was significantly lower in the fitusiran prophylaxis group (3·1 [95% CI 2·3-4·3]) than in the on-demand clotting factor concentrates group (31·0 [21·1-45·5]; rate ratio 0·101 [95% CI 0·064-0·159]; p<0·0001). In the fitusiran group, 40 (51%) of 79 treated participants had no treated bleeds compared with two (5%) of 40 participants in the on-demand clotting factor concentrates group. Increased alanine aminotransferase concentration (18 [23%] of 79 participants in the safety analysis set) was the most common treatment-emergent adverse event in the fitusiran group and hypertension (four (10%) of 40 participants) was the most common in the on-demand clotting factor concentrates group. Treatment-emergent serious adverse events were reported in five (6%) participants in the fitusiran group (cholelithiasis [n=2, 3%], cholecystitis [n=1, 1%], lower respiratory tract infection [n=1, 1%], and asthma [n=1, 1%]) and five (13%) participants in the on-demand clotting factor concentrates group (gastroenteritis, pneumonia, suicidal ideation, diplopia, osteoarthritis, epidural haemorrhage, humerus fracture, subdural haemorrhage, and tibia fracture [all n=1, 3%]). No treatment-related thrombosis or deaths were reported. INTERPRETATION: In participants with haemophilia A or B without inhibitors, fitusiran prophylaxis resulted in significant reductions in annualised bleeding rate compared with on-demand clotting factor concentrates and no bleeding events in approximately half of participants. Fitusiran prophylaxis shows haemostatic efficacy in both haemophilia A and haemophilia B, and therefore has the potential to be transformative in the management of all people with haemophilia. FUNDING: Sanofi.


Assuntos
Hemofilia A , Hemofilia B , Masculino , Humanos , Hemofilia A/complicações , Hemofilia A/tratamento farmacológico , Hemofilia B/complicações , Hemofilia B/tratamento farmacológico , Hemorragia/etiologia , Hemorragia/prevenção & controle , Hemorragia/tratamento farmacológico , Fatores de Coagulação Sanguínea/uso terapêutico , RNA Interferente Pequeno/uso terapêutico
3.
Lancet ; 401(10386): 1427-1437, 2023 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-37003287

RESUMO

BACKGROUND: Fitusiran, a subcutaneous investigational small interfering RNA therapeutic, targets antithrombin to rebalance haemostasis in people with haemophilia A or haemophilia B, irrespective of inhibitor status. We evaluated the efficacy and safety of fitusiran prophylaxis in people with haemophilia A or haemophilia B with inhibitors. METHODS: This multicentre, randomised, open-label phase 3 study was done at 26 sites (primarily secondary or tertiary centres) in 12 countries. Men, boys, and young adults aged 12 years or older with severe haemophilia A or haemophilia B with inhibitors previously treated with on-demand bypassing agents were randomly assigned (2:1) to receive once-a-month 80 mg subcutaneous fitusiran prophylaxis (fitusiran prophylaxis group) or to continue with bypassing agents on-demand (bypassing agents on-demand group) for 9 months. The primary endpoint was mean annualised bleeding rate during the efficacy period in the intention-to-treat population estimated by negative binomial model. Safety was assessed as a secondary endpoint in the safety population. This trial is complete and is registered with ClinicalTrials.gov, NCT03417102. FINDINGS: Between Feb 14, 2018, and June 23, 2021, 85 participants were screened for inclusion, of whom 57 (67%; 57 [100%] men; median age 27·0 years [IQR 19·5-33·5]) were randomly assigned: 19 (33%) participants to the bypassing agent on-demand group and 38 (67%) participants to the fitusiran prophylaxis. Negative binomial model-based mean annualised bleeding rate was significantly lower in the fitusiran prophylaxis group (1·7 [95% CI 1·0-2·7]) than in the bypassing agents on-demand group (18·1 [10·6-30·8]), corresponding to a 90·8% (95% CI 80·8-95·6) reduction in annualised bleeding rate in favour of fitusiran prophylaxis (p<0·0001). 25 (66%) participants had zero treated bleeds in the fitusiran prophylaxis group versus one (5%) in the bypassing agents on-demand group. The most frequent treatment-emergent adverse event in the fitusiran prophylaxis group was increased alanine aminotransferase in 13 (32%) of 41 participants in the safety population; there were no increased alanine aminotransferase treatment-emergent adverse events in the bypassing agents on-demand group. Suspected or confirmed thromboembolic events were reported in two (5%) participants in the fitusiran prophylaxis group. No deaths were reported. INTERPRETATION: Subcutaneous fitusiran prophylaxis resulted in statistically significant reductions in annualised bleeding rate in participants with haemophilia A or haemophilia B with inhibitors, with two-thirds of participants having zero bleeds. Fitusiran prophylaxis might show haemostatic efficacy in participants with haemophilia A or haemophilia B with inhibitors; therefore, the therapeutic might have the potential to improve the management of people with haemophilia. FUNDING: Sanofi.


Assuntos
Hemofilia A , Hemofilia B , Masculino , Adulto Jovem , Humanos , Adulto , Feminino , Hemofilia A/complicações , Hemofilia A/tratamento farmacológico , Hemofilia B/complicações , Hemofilia B/tratamento farmacológico , Alanina Transaminase , Hemorragia/epidemiologia , RNA Interferente Pequeno/uso terapêutico
4.
J Thromb Haemost ; 19(6): 1436-1446, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33587824

RESUMO

BACKGROUND: Fitusiran, an investigational small interfering RNA therapy, reduces antithrombin production to rebalance hemostasis in people with hemophilia A or B, with or without inhibitors. OBJECTIVES: To evaluate the safety and efficacy of fitusiran treatment for people with moderate/severe hemophilia A or B with inhibitors. PATIENTS/METHODS: In this open-label phase 1, part D study, 17 males with hemophilia A or B with inhibitors received three once-monthly subcutaneous injections of fitusiran 50 mg (n = 6) or 80 mg (n = 11); followed for up to 112 days. Endpoints included safety (primary), pharmacokinetics/pharmacodynamics (secondary), annualized bleeding rate, and patient-reported outcomes (exploratory). RESULTS: The most common adverse event was injection site erythema (n = 8). No thrombotic events were reported. At nadir, mean (standard error of the mean [SEM]) antithrombin activity decreased from baseline by 82.0% (2.2) and 87.4% (0.7) in the 50 mg and 80 mg groups, respectively. Antithrombin reduction was associated with increased thrombin generation. 11/17 (64.7%) participants had no bleeds during the observation period (mean [standard deviation] 69.4 [16.3] days). Mean (SEM) changes from baseline in Haemophilia Quality of Life Questionnaire for Adults total (-9.2 [2.9]) and physical health (-12.3 [3.9]) domain scores suggested clinically meaningful improvement. CONCLUSIONS: Monthly fitusiran was generally well tolerated, lowered antithrombin levels from baseline, and resulted in improved thrombin generation. These preliminary results suggest that monthly fitusiran treatment may reduce bleeding episodes and improve quality of life in participants with hemophilia A or B with inhibitors.


Assuntos
Hemofilia A , Hemofilia B , Acetilgalactosamina , Adulto , Antitrombinas/efeitos adversos , Hemofilia A/diagnóstico , Hemofilia A/tratamento farmacológico , Hemofilia B/diagnóstico , Hemofilia B/tratamento farmacológico , Hemofilia B/genética , Humanos , Masculino , Qualidade de Vida , RNA Interferente Pequeno
5.
Blood ; 135(17): 1484-1496, 2020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32078672

RESUMO

Factor VIII (FVIII) replacement products enable comprehensive care in hemophilia A. Treatment goals in severe hemophilia A are expanding beyond low annualized bleed rates to include long-term outcomes associated with high sustained FVIII levels. Endogenous von Willebrand factor (VWF) stabilizes and protects FVIII from degradation and clearance, but it also subjects FVIII to a half-life ceiling of ∼15 to 19 hours. Increasing recombinant FVIII (rFVIII) half-life further is ultimately dependent upon uncoupling rFVIII from endogenous VWF. We have developed a new class of FVIII replacement, rFVIIIFc-VWF-XTEN (BIVV001), that is physically decoupled from endogenous VWF and has enhanced pharmacokinetic properties compared with all previous FVIII products. BIVV001 was bioengineered as a unique fusion protein consisting of a VWF-D'D3 domain fused to rFVIII via immunoglobulin-G1 Fc domains and 2 XTEN polypeptides (Amunix Pharmaceuticals, Inc, Mountain View, CA). Plasma FVIII half-life after BIVV001 administration in mice and monkeys was 25 to 31 hours and 33 to 34 hours, respectively, representing a three- to fourfold increase in FVIII half-life. Our results showed that multifaceted protein engineering, far beyond a few amino acid substitutions, could significantly improve rFVIII pharmacokinetic properties while maintaining hemostatic function. BIVV001 is the first rFVIII with the potential to significantly change the treatment paradigm for severe hemophilia A by providing optimal protection against all bleed types, with less frequent doses. The protein engineering methods described herein can also be applied to other complex proteins.


Assuntos
Fator VIII/metabolismo , Hemofilia A/terapia , Hemorragia/prevenção & controle , Proteínas Recombinantes de Fusão/administração & dosagem , Fator de von Willebrand/metabolismo , Animais , Fator VIII/genética , Hemofilia A/metabolismo , Hemofilia A/patologia , Hemostasia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Primatas , Fator de von Willebrand/genética
6.
Thromb Haemost ; 117(3): 508-518, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28004057

RESUMO

The safety, efficacy, and prolonged half-life of recombinant factor IX Fc fusion protein (rFIXFc) were demonstrated in the Phase 3 B-LONG (adults/adolescents ≥12 years) and Kids B-LONG (children <12 years) studies of subjects with haemophilia B (≤2 IU/dl). Here, we report interim, long-term safety and efficacy data from B-YOND, the rFIXFc extension study. Eligible subjects who completed B-LONG or Kids B-LONG could enrol in B-YOND. There were four treatment groups: weekly prophylaxis (20-100 IU/kg every 7 days), individualised prophylaxis (100 IU/kg every 8-16 days), modified prophylaxis (further dosing personalisation to optimise prophylaxis), and episodic (on-demand) treatment. Subjects could change treatment groups at any point. Primary endpoint was inhibitor development. One hundred sixteen subjects enrolled in B-YOND. From the start of the parent studies to the B-YOND interim data cut, median duration of rFIXFc treatment was 39.5 months and 21.9 months among adults/adolescents and children, respectively; 68/93 (73.1 %) adults/adolescents and 9/23 (39.1 %) children had ≥100 cumulative rFIXFc exposure days. No inhibitors were observed. Median annualised bleeding rates (ABRs) were low in all prophylaxis regimens: weekly (≥12 years: 2.3; <6 years: 0.0; 6 to <12 years: 2.7), individualised (≥12 years: 2.3; 6 to <12 years: 2.4), and modified (≥12 years: 2.4). One or two infusions were sufficient to control 97 % (adults/adolescents) and 95 % (children) of bleeding episodes. Interim data from B-YOND are consistent with data from B-LONG and Kids B-LONG, and confirm the long-term safety of rFIXFc, absence of inhibitors, and maintenance of low ABRs with prophylactic dosing every 1 to 2 weeks.


Assuntos
Coagulantes/administração & dosagem , Fator IX/administração & dosagem , Hemofilia B/tratamento farmacológico , Hemorragia/prevenção & controle , Hemostasia/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Adolescente , Adulto , Anticorpos Neutralizantes/sangue , Criança , Coagulantes/efeitos adversos , Esquema de Medicação , Fator IX/efeitos adversos , Fator IX/imunologia , Hemofilia B/sangue , Hemofilia B/diagnóstico , Hemorragia/sangue , Humanos , Fragmentos Fc das Imunoglobulinas/efeitos adversos , Fragmentos Fc das Imunoglobulinas/imunologia , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/imunologia , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
7.
Crit Rev Biotechnol ; 36(6): 1110-1122, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26383226

RESUMO

Biotherapeutic proteins represent a mainstay of treatment for a multitude of conditions, for example, autoimmune disorders, hematologic disorders, hormonal dysregulation, cancers, infectious diseases and genetic disorders. The technologies behind their production have changed substantially since biotherapeutic proteins were first approved in the 1980s. Although most biotherapeutic proteins developed to date have been produced using the mammalian Chinese hamster ovary and murine myeloma (NS0, Sp2/0) cell lines, there has been a recent shift toward the use of human cell lines. One of the most important advantages of using human cell lines for protein production is the greater likelihood that the resulting recombinant protein will bear post-translational modifications (PTMs) that are consistent with those seen on endogenous human proteins. Although other mammalian cell lines can produce PTMs similar to human cells, they also produce non-human PTMs, such as galactose-α1,3-galactose and N-glycolylneuraminic acid, which are potentially immunogenic. In addition, human cell lines are grown easily in a serum-free suspension culture, reproduce rapidly and have efficient protein production. A possible disadvantage of using human cell lines is the potential for human-specific viral contamination, although this risk can be mitigated with multiple viral inactivation or clearance steps. In addition, while human cell lines are currently widely used for biopharmaceutical research, vaccine production and production of some licensed protein therapeutics, there is a relative paucity of clinical experience with human cell lines because they have only recently begun to be used for the manufacture of proteins (compared with other types of cell lines). With additional research investment, human cell lines may be further optimized for routine commercial production of a broader range of biotherapeutic proteins.


Assuntos
Produtos Biológicos/metabolismo , Linhagem Celular/metabolismo , Animais , Humanos , Engenharia Metabólica , Proteínas/metabolismo
8.
Biologicals ; 43(4): 213-9, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26094124

RESUMO

Recombinant factor VIII Fc fusion protein (rFVIIIFc) is a long-acting coagulation factor approved for the treatment of hemophilia A. Here, the rFVIIIFc manufacturing process and results of studies evaluating product quality and the capacity of the process to remove potential impurities and viruses are described. This manufacturing process utilized readily transferable and scalable unit operations and employed multi-step purification and viral clearance processing, including a novel affinity chromatography adsorbent and a 15 nm pore size virus removal nanofilter. A cell line derived from human embryonic kidney (HEK) 293H cells was used to produce rFVIIIFc. Validation studies evaluated identity, purity, activity, and safety. Process-related impurity clearance and viral clearance spiking studies demonstrate robust and reproducible removal of impurities and viruses, with total viral clearance >8-15 log10 for four model viruses (xenotropic murine leukemia virus, mice minute virus, reovirus type 3, and suid herpes virus 1). Terminal galactose-α-1,3-galactose and N-glycolylneuraminic acid, two non-human glycans, were undetectable in rFVIIIFc. Biochemical and in vitro biological analyses confirmed the purity, activity, and consistency of rFVIIIFc. In conclusion, this manufacturing process produces a highly pure product free of viruses, impurities, and non-human glycan structures, with scale capabilities to ensure a consistent and adequate supply of rFVIIIFc.


Assuntos
Fator VIII/biossíntese , Preparações de Ação Retardada , Fator VIII/isolamento & purificação , Fator VIII/uso terapêutico , Células HEK293 , Humanos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/uso terapêutico
9.
Br J Haematol ; 168(1): 113-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25209873

RESUMO

In the phase 3 B-LONG [Recombinant Factor IX Fc Fusion Protein (rFIXFc) in Subjects with Haemophilia B] study, rFIXFc dosed every 1-2 weeks was safe and efficacious in previously treated subjects with haemophilia B. To date, there are no evaluations of transitioning from conventional to long-acting factor IX (FIX) prophylaxis. This post-hoc analysis of B-LONG subjects compared prophylaxis with other FIX products and rFIXFc. Pre- and on-study data were analysed to assess dosing regimen, weekly FIX consumption and annualized bleeding rates (ABRs). Population pharmacokinetics models were used to generate FIX activity profiles with rFIXFc and recombinant FIX prophylaxis. Thirty-nine subjects, previously treated prophylactically, were evaluated. Prior to study, most subjects (69·2%) received twice-weekly FIX infusions; on study, subjects infused rFIXFc once every 1-2 weeks with c. 30-50% reductions in weekly consumption. On-study estimated mean ABRs were lower than pre-study estimated mean ABRs. Models predicted that rFIXFc administered 50 iu/kg weekly and 100 iu/kg every 10 d would maintain steady-state FIX trough levels ≥1 iu/dl in 95·4% and 89·2% of subjects, respectively. These results indicate that patients receiving rFIXFc prophylaxis can markedly reduce infusion frequency and FIX consumption, have a greater likelihood of maintaining FIX activity >1 iu/dl and experience fewer bleeding episodes compared with prior FIX prophylaxis.


Assuntos
Quimioprevenção , Fator IX/uso terapêutico , Hemofilia B/tratamento farmacológico , Hemorragia/prevenção & controle , Proteínas Recombinantes de Fusão/uso terapêutico , Adolescente , Adulto , Idoso , Criança , Substituição de Medicamentos , Fator IX/farmacologia , Hemofilia B/complicações , Hemorragia/etiologia , Humanos , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/farmacologia , Resultado do Tratamento , Adulto Jovem
10.
PLoS One ; 9(11): e113600, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25415306

RESUMO

Recombinant factor VIII Fc (rFVIIIFc) is a fusion protein consisting of a single B-domain-deleted (BDD) FVIII linked recombinantly to the Fc domain of human IgG1 to extend half-life. To determine if rFVIIIFc could be further improved by maintaining the heavy and light chains within a contiguous single chain (SC), we evaluated the activity and function of SC rFVIIIFc, an isoform that is not processed at residue R1648. SC rFVIIIFc showed equivalent activity in a chromogenic assay compared to rFVIIIFc, but approximately 40% activity by the one-stage clotting assay in the presence of von Willebrand Factor (VWF), with full activity in the absence of VWF. Moreover, SC rFVIIIFc demonstrated markedly delayed thrombin-mediated release from VWF, but an activity similar to that of rFVIIIFc upon activation in FXa generation assays. Therefore, the apparent reduction in specific activity in the aPTT assay appears to be primarily due to delayed release of FVIII from VWF. To assess whether stability and activity of SC rFVIIIFc were affected in vivo, a tail vein transection model in Hemophilia A mice was utilized. The results demonstrated similar pharmacokinetic profiles and comparable efficacy for SC rFVIIIFc and rFVIIIFc. Thus, while the single chain configuration did not promote enhanced half-life, it reduced the rate of release of FVIII from VWF required for activation. This impaired release may underlie the observed reduction in the one-stage clotting assay, but does not appear to affect the physiological activity of SC rFVIIIFc.


Assuntos
Fator VIII/genética , Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacocinética , Animais , Clonagem Molecular/métodos , Modelos Animais de Doenças , Fator VIII/química , Fator VIII/uso terapêutico , Meia-Vida , Hemofilia A/sangue , Hemorragia , Humanos , Técnicas In Vitro , Masculino , Camundongos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/uso terapêutico , Trombina/metabolismo , Fator de von Willebrand/metabolismo
11.
Mol Biotechnol ; 45(3): 248-56, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20379804

RESUMO

Many therapeutic proteins require appropriate glycosylation for their biological activities and plasma half life. Coagulation factor VIII (FVIII) is a glycoprotein which has extensive post-translational modification by N-linked glycosylation. The terminal sialic acid in the N-linked glycans of FVIII is required for maximal circulatory half life. The extent of FVIII sialylation can be determined by high pH anion-exchange chromatography coupled with a pulse electrochemical detector (HPAEC-PED), but this requires a large amount of purified protein. Using FVIII as a model, the objective of the present study was to develop assays that enable detection and prediction of sialylation deficiency at an early stage in the process and thus prevent downstream product quality excursions. Lectin ECA (Erythrina Cristagalli) binds to unsialylated Galbeta1-4 GlcNAc and the ECA-binding level (i.e., terminal Gal(beta1-4) exposure) is inversely proportional to the level of sialylation. By using ECA, a cell-based assay was developed to measure the global sialylation profile in FVIII producing cells. To examine the Galbeta1-4 exposure on the FVIII molecule in bioreactor tissue culture fluid (TCF), an ELISA-based ECA-FVIII binding assay was developed. The ECA-binding specificity in both assays was assessed by ECA-specific sugar inhibitors and neuraminidase digestion. The ECA-binding specificity was also independently confirmed by a ST3GAL4 siRNA knockdown experiment. To establish the correlation between Galbeta1-4 exposure and the HPAEC-PED determined FVIII sialylation value, the FVIII containing bioreactor TCF and the purified FVIII samples were tested with ECA ELISA binding assay. The results indicated an inverse correlation between ECA binding and the corresponding HPAEC-PED sialylation value. The ECA-binding assays are cost effective and can be rapidly performed, thereby making them effective for in-process monitoring of protein sialylation.


Assuntos
Biotecnologia/métodos , Fator VIII/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Lectinas de Plantas/metabolismo , Amino Açúcares/farmacologia , Animais , Reatores Biológicos , Meios de Cultura , Ensaio de Imunoadsorção Enzimática , Fator VIII/química , Citometria de Fluxo , Fluoresceína-5-Isotiocianato , Galactose/metabolismo , Glicosilação , Humanos , Camundongos , Neuraminidase/farmacologia , Reação em Cadeia da Polimerase , Polissacarídeos/química , Ligação Proteica/efeitos dos fármacos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Sialiltransferases/genética , Sialiltransferases/metabolismo , beta-Galactosídeo alfa-2,3-Sialiltransferase
12.
Blood ; 116(2): 270-9, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20194895

RESUMO

A long-acting factor VIII (FVIII) as a replacement therapy for hemophilia A would significantly improve treatment options for patients with hemophilia A. To develop a FVIII with an extended circulating half-life, but without a reduction in activity, we have engineered 23 FVIII variants with introduced surface-exposed cysteines to which a polyethylene glycol (PEG) polymer was specifically conjugated. Screening of variant expression level, PEGylation yield, and functional assay identified several conjugates retaining full in vitro coagulation activity and von Willebrand factor (VWF) binding.PEGylated FVIII variants exhibited improved pharmacokinetics in hemophilic mice and rabbits. In addition, pharmacokinetic studies in VWF knockout mice indicated that larger molecular weight PEG may substitute for VWF in protecting PEGylated FVIII from clearance in vivo. In bleeding models of hemophilic mice, PEGylated FVIII not only exhibited prolonged efficacy that is consistent with the improved pharmacokinetics but also showed efficacy in stopping acute bleeds comparable with that of unmodified rFVIII. In summary site-specifically PEGylated FVIII has the potential to be a long-acting prophylactic treatment while being fully efficacious for on-demand treatment for patients with hemophilia A.


Assuntos
Coagulantes/farmacocinética , Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Polietilenoglicóis/farmacocinética , Animais , Western Blotting , Coagulantes/administração & dosagem , Coagulantes/química , Preparações de Ação Retardada , Eletroforese em Gel de Poliacrilamida , Fator VIII/administração & dosagem , Fator VIII/química , Meia-Vida , Camundongos , Camundongos Knockout , Mutagênese Sítio-Dirigida , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Coelhos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
13.
Mol Biotechnol ; 34(2): 165-78, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17172662

RESUMO

Loss of coagulation factor VIII (FVIII) function results in a bleeding disorder, hemophilia A, which requires FVIII replacement therapy. Owing to its large size and complexity, the expression level of recombinant FVIII is two to three orders of magnitude lower than other recombinant proteins produced in mammalian cell lines. To understand cellular factors limiting FVIII expression, we studied the expression of FVIII in a human cell line, HKB11 (a hybrid cell line of HEK293 and a human B cell line). In comparison with other cell lines, such as HEK293 and BHK-21, HKB11 showed increased FVIII expression levels. With unamplified, pooled stable cells, FVIII expression in HKB11 cells was 8- to 30-fold higher than the other cell lines tested. In this study, HKB11 clones expressing varying levels of FVIII were analyzed and FVIII secreted from these clones had similar specific activity. Characterization of these clones by immunofluorescence staining, Western blotting analysis, and flow cytometry showed that high-producing cells not only secreted more active FVIII but also accumulated more FVIII protein intracellularly. FVIII expression appears to be controlled by the rates of transcription, translation, and secretion, but transcription and translation may play more important roles than secretion in determining expression level in HKB11 cells. FACS analysis of live cells showed that the high-producing clones also had more FVIII on the HKB11 cell surface than low-producing cells, thus opening the possibility of using FACS to select high-producing cell lines. Expression levels of the chaperone protein Hsp70 and antiapoptotic proteins such as Bcl-2 and Bcl-xL were similar among HKB11 clones with different FVIII productivity. In conclusion, HKB11 is an efficient host cell line for expression of FVIII and possibly other recombinant proteins. Systematic approaches, such as gene expression profiling by DNA microarray, will be necessary to understand the global changes in the cells producing recombinant proteins.


Assuntos
Biotecnologia/métodos , Fator VIII/biossíntese , Proteínas Recombinantes/biossíntese , Western Blotting , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Fator VIII/análise , Fator VIII/genética , Citometria de Fluxo , Proteínas de Choque Térmico HSP70/análise , Proteínas de Choque Térmico HSP70/biossíntese , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/análise , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Proteína bcl-X/análise , Proteína bcl-X/biossíntese
14.
Hepatology ; 40(5): 1106-15, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15389776

RESUMO

Liver fibrosis is characterized by increased synthesis, and decreased degradation, of extracellular matrix (ECM) within the injured tissue. Decreased ECM degradation results, in part, from increased expression of tissue inhibitor of metalloproteinase-1 (TIMP-1), which blocks matrix metalloproteinase (MMP) activity. TIMP-1 is also involved in promoting survival of activated hepatic stellate cells (HSCs), a major source of ECM. This study examined the effects of blocking TIMP-1 activity in a clinically relevant model of established liver fibrosis. Rats were treated with carbon tetrachloride (CCl(4)), or olive oil control, for 6 weeks; 24 days into the treatment, the rats were administered a neutralizing anti-TIMP-1 antibody derived from a fully human combinatorial antibody library (HuCAL), PBS, or an isotype control antibody. Livers from CCl(4)-treated rats exhibited substantial damage, including bridging fibrosis, inflammation, and extensive expression of smooth muscle alpha-actin (alpha-SMA). Compared to controls, rats administered anti-TIMP-1 showed a reduction in collagen accumulation by histological examination and hydroxyproline content. Administration of anti-TIMP-1 resulted in a marked decrease in alpha-SMA staining. Zymography analysis showed antibody treatment decreased the activity of MMP-2. In conclusion, administration of a TIMP-1 antibody attenuated CCl(4)-induced liver fibrosis and decreased HSC activation and MMP-2 activity.


Assuntos
Cirrose Hepática/patologia , Cirrose Hepática/prevenção & controle , Inibidor Tecidual de Metaloproteinase-1/fisiologia , Actinas/antagonistas & inibidores , Actinas/metabolismo , Animais , Anticorpos/farmacologia , Tetracloreto de Carbono , Colágeno/metabolismo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Masculino , Inibidores de Metaloproteinases de Matriz , Músculo Liso/metabolismo , Ratos , Ratos Wistar , Índice de Gravidade de Doença , Inibidor Tecidual de Metaloproteinase-1/imunologia
15.
Biochem J ; 364(Pt 1): 137-44, 2002 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-11988086

RESUMO

We originally identified preadipocyte factor-1 (Pref-1) as an inhibitor of adipogenesis by the fact that constitutive expression of full-length Pref-1A inhibits differentiation of 3T3-L1 cells into adipocytes. Subsequently, we found that the membrane form of Pref-1 is proteolytically processed at two sites in the extracellular domain, resulting in the larger (50 kDa) and smaller (25 kDa) soluble forms. A specific form(s) of Pref-1, which is active in inhibiting adipocyte differentiation, has not been elucidated. Here, various artificial constructs and alternative-splicing variants of Pref-1 were stably transfected into 3T3-L1 cells, or conditioned media from COS cells transfected with the various forms were added into differentiating 3T3-L1 cells. Judging by Oil Red O staining for lipid accumulation and expression of adipocyte markers, we determined that, unlike the full-length Pref-1A and the constructed large soluble form, the artificial membrane form of Pref-1 lacking the processing site proximal to the membrane was not effective in inhibiting adipogenesis. Furthermore, conditioned media from COS cells transfected with the construct containing only the first three epidermal growth factor repeats, corresponding to the small soluble form, was not effective in inhibiting adipocyte differentiation. Of the four alternative-splicing products, Pref-1A and Pref-1B, which generate both large and small soluble forms, inhibited adipogenesis, whereas Pref-1C and Pref-1D, which lack the processing site proximal to the membrane and therefore generate only the smaller soluble form, did not show any effect. We conclude that only the large soluble form, and not the transmembrane or the small soluble form, of Pref-1 is biologically active and that alternative splicing therefore determines Pref-1 function in adipocyte differentiation.


Assuntos
Adipócitos/citologia , Processamento Alternativo , Membrana Celular/metabolismo , Proteínas de Membrana/química , Proteínas Repressoras/química , Células 3T3 , Adipócitos/metabolismo , Animais , Northern Blotting , Western Blotting , Células COS , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Meios de Cultivo Condicionados/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA