Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Baisong Mei is active.

Publication


Featured researches published by Baisong Mei.


Critical Reviews in Biotechnology | 2016

Human cell lines for biopharmaceutical manufacturing: history, status, and future perspectives

Jennifer A. Dumont; Don Euwart; Baisong Mei; Scott Estes; Rashmi Kshirsagar

Abstract 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.


Biologicals | 2015

Manufacturing process used to produce long-acting recombinant factor VIII Fc fusion protein

Justin McCue; Rashmi Kshirsagar; Keith Selvitelli; Qi Lu; Mingxuan Zhang; Baisong Mei; Robert T. Peters; Glenn F. Pierce; Jennifer A. Dumont; Stephen Raso; Heidi Reichert

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.


British Journal of Haematology | 2015

Switching to recombinant factor IX Fc fusion protein prophylaxis results in fewer infusions, decreased factor IX consumption and lower bleeding rates.

Jerry S. Powell; Amy D. Shapiro; Margaret V. Ragni; Claude Negrier; Jerzy Windyga; Margareth Castro Ozelo; John Pasi; Ross Baker; James Potts; Shuanglian Li; Baisong Mei; Glenn F. Pierce; Brian Robinson

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.


Haemophilia | 2014

Validation of the manufacturing process used to produce long‐acting recombinant factor IX Fc fusion protein

Justin McCue; D. Osborne; Jennifer A. Dumont; Robert T. Peters; Baisong Mei; Glenn F. Pierce; K. Kobayashi; D. Euwart

Recombinant factor IX Fc (rFIXFc) fusion protein is the first of a new class of bioengineered long‐acting factors approved for the treatment and prevention of bleeding episodes in haemophilia B. The aim of this work was to describe the manufacturing process for rFIXFc, to assess product quality and to evaluate the capacity of the process to remove impurities and viruses. This manufacturing process utilized a transferable and scalable platform approach established for therapeutic antibody manufacturing and adapted for production of the rFIXFc molecule. rFIXFc was produced using a process free of human‐ and animal‐derived raw materials and a host cell line derived from human embryonic kidney (HEK) 293H cells. The process employed multi‐step purification and viral clearance processing, including use of a protein A affinity capture chromatography step, which binds to the Fc portion of the rFIXFc molecule with high affinity and specificity, and a 15 nm pore size virus removal nanofilter. Process validation studies were performed to evaluate identity, purity, activity and safety. The manufacturing process produced rFIXFc with consistent product quality and high purity. Impurity clearance validation studies demonstrated robust and reproducible removal of process‐related impurities and adventitious viruses. The rFIXFc manufacturing process produces a highly pure product, free of non‐human glycan structures. Validation studies demonstrate that this product is produced with consistent quality and purity. In addition, the scalability and transferability of this process are key attributes to ensure consistent and continuous supply of rFIXFc.


Haemophilia | 2016

Changes in health‐related quality of life with treatment of longer‐acting clotting factors: results in the A‐LONG and B‐LONG clinical studies

K. W. Wyrwich; S. Krishnan; P. Auguste; J.-L. Poon; R. von Maltzahn; R. Yu; Glenn F. Pierce; Baisong Mei; Johnny Mahlangu; S. Von Mackensen

In haemophilia, prophylactic infusion of replacement factor can result in improvements in health‐related quality of life (HRQoL) when compared with episodic treatment. The Haemophilia‐specific Quality of Life (Haem‐A‐QoL) questionnaire assessed HRQoL in adults with severe haemophilia A or B who received prophylactic or episodic treatment with recombinant factor VIII or IX Fc fusion protein (rFVIIIFc or rFIXFc) in the A‐LONG or B‐LONG clinical studies.


Thrombosis and Haemostasis | 2016

Long-term safety and efficacy of extended-interval prophylaxis with recombinant factor IX Fc fusion protein (rFIXFc) in subjects with haemophilia B

K.J. Pasi; K. Fischer; Margaret V. Ragni; Beatrice Nolan; David J. Perry; Roshni Kulkarni; Margareth Castro Ozelo; Johnny Mahlangu; A. Shapiro; Ross Baker; Carolyn M. Bennett; C. Barnes; Johannes Oldenburg; Tadashi Matsushita; Huixing Yuan; Alejandra Ramirez-Santiago; Glenn F. Pierce; Geoffrey Allen; Baisong Mei

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.


PLOS ONE | 2014

A Single Chain Variant of Factor VIII Fc Fusion Protein Retains Normal In Vivo Efficacy but Exhibits Altered In Vitro Activity

Yang Buyue; Tongyao Liu; John Kulman; Garabet G. Toby; George D. Kamphaus; Susannah Patarroyo-White; Qi Lu; Thomas Reidy; Baisong Mei; Haiyan Jiang; Glenn F. Pierce; Jurg M. Sommer; Robert T. Peters

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.


Archive | 2012

FACTOR VIII COMPOSITIONS AND METHODS OF MAKING AND USING SAME

Volker Schellenberger; Pei-Yun Chang; Fatbardha Varfaj; John Kulman; Tongyao Liu; Garabet G. Toby; Haiyan Jiang; Robert T. Peters; Deping Wang; Baisong Mei; Joshua Silverman; Chia-Wei Wang; Benjamin Spink; Nathan Geething


Fusion Protein Technologies for Biopharmaceuticals: Applications and Challenges | 2013

Monomeric Fc‐Fusion Proteins

Baisong Mei; Susan C. Low; Snejana Krassova; Robert T. Peters; Glenn F. Pierce; Jennifer A. Dumont


Japanese Journal of Thrombosis and Hemostasis | 2015

Japanese subject subpopulation analysis of B-LONG: a Phase 3 study of long-acting recombinant factor IX Fc fusion protein

Midori Shima; Katsuyuki Fukutake; Hideji Hanabusa; Tadashi Matsushita; Masashi Taki; Michio Sakai; Toshiyuki Hirakata; Yingwen Dong; Shuanglian Li; Lynda M. Cristiano; Glenn F. Pierce; Baisong Mei

Collaboration


Dive into the Baisong Mei's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge