T-cell activation and B-cell depletion in chimpanzees treated with a bispecific anti-CD19/anti-CD3 single-chain antibody construct

2005 ◽  
Vol 55 (5) ◽  
pp. 503-514 ◽  
Author(s):  
Bernd Schlereth ◽  
Cornelia Quadt ◽  
Torsten Dreier ◽  
Peter Kufer ◽  
Grit Lorenczewski ◽  
...  
2010 ◽  
Vol 207 (8) ◽  
pp. 1579-1587 ◽  
Author(s):  
Hafid Ait-Oufella ◽  
Olivier Herbin ◽  
Jean-David Bouaziz ◽  
Christoph J. Binder ◽  
Catherine Uyttenhove ◽  
...  

B cell depletion significantly reduces the burden of several immune-mediated diseases. However, B cell activation has been until now associated with a protection against atherosclerosis, suggesting that B cell–depleting therapies would enhance cardiovascular risk. We unexpectedly show that mature B cell depletion using a CD20-specific monoclonal antibody induces a significant reduction of atherosclerosis in various mouse models of the disease. This treatment preserves the production of natural and potentially protective anti–oxidized low-density lipoprotein (oxLDL) IgM autoantibodies over IgG type anti-oxLDL antibodies, and markedly reduces pathogenic T cell activation. B cell depletion diminished T cell–derived IFN-γ secretion and enhanced production of IL-17; neutralization of the latter abrogated CD20 antibody–mediated atheroprotection. These results challenge the current paradigm that B cell activation plays an overall protective role in atherogenesis and identify new antiatherogenic strategies based on B cell modulation.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1841-1841
Author(s):  
Gordon Ng ◽  
Thomas Spreter ◽  
Rupert Davies ◽  
Grant Wickman

Abstract Although blinatumomab is an approved treatment for Philadelphia chromosome negative relapsed or refractory (r/r) precursor-B cell ALL and is under development for r/r B cell NHL, blinatumomab has several limitations impacting fuller therapeutic utility. For instance, blinatumomab therapy requires continuous infusion due to its rapid clearance owing to its size and lack of an antibody Fc and has been associated with potentially life-threatening CNS toxicities and cytokine release syndrome (Viardot A et al, Blood 2016; Goebeler ME et al, J Clin Oncol 2016; Topp et al, Lancet Oncol 2015). In addition, blinatumomab treatment is associated with higher incidence of relapse in patients with high disease burden, and its T cell redirected killing is limited by T cell immunosuppression (e.g. PD-1/PD-L1 up-regulation [Köhnke et al, J Hematol & Oncol 2015]; Treg suppression [Duell et al, ASH abstract 2014]). ZW38 is designed to address each of these limitations and represents a best in class CD19-directed CD3 T cell engager and a novel class of bispecific antibody drug conjugate (ADC). ZW38 contains an Azymetric IgG1-like Fc that carries mutations in the CH2 domain preventing FcgR dependent ADCC and ADCP and exhibits typical IgG1-like PK in rodent studies. Transient expression in mammalian CHO cells demonstrate ZW38 can be expressed at a titre of hundreds mg/L and can be purified using conventional IgG antibody methods and resins with typical IgG step purification yields and high heterodimer purity. Additionally, ZW38 has been conjugated to a microtubule inhibitor that lacks bystander killing. ZW38 antibody paratopes have been engineered to favor T cell-B cell functional engagement and selective target B cell cytotoxicity. In vitro studies demonstrated that ZW38 binds to human CD19+ B cells with >30-fold higher affinity than to human CD3+ T cells. Similar to blinatumomab, ZW38 can redirect the killing of target cancer B cells via T cell subtypes from human PBMC and its cytotoxicity is target B cell dependent. At concentrations that result in efficacious B cell depletion, ZW38 does not overly activate T cells. ZW38 is specifically engineered to induce more 'controlled' T cell activation than blinatumomab while still mediating sufficient T cell redirected target B cell depletion. ZW38 mediates T cell activation, cytokine release, and proliferation at nanomolar potency. By design, the sufficient, lower cytokine levels necessary for B cell killing may reduce the risk of cytokine release syndrome and T cell anergy. ZW38 exhibited potent growth inhibition in a panel of different B cell ALL and NHL cancer lines including but not limited to: G2, Nalm-6, RS4-11, Daudi, SUDHL-4 and SUDHL-6. In comparison to blinatumomab, ZW38 exhibits superior target B cell depletion in in vitro co-cultures of Raji lymphoma cells and human PBMC (ZW38 depletes > 90% B cells; blinatumomab depletes 20-90% B cells depending on the PBMC donor). In addition, ZW38 was effective in killing target B cells in PBMC cultures in which PD-1 has been up-regulated, which were resistant to blinatumomab killing in this assay. The dual mechanisms of action of ZW38, redirected T cell cytotoxicity and ADC cytotoxicity, may prolong and/or boost response rates, lower the incidence of relapse, and reduce the likelihood of acquired resistance while its 'controlled' T cell activation profile may reduce the risk of life-threatening and potentially fatal neurotoxicity and CRS. Disclosures Ng: Zymeworks: Employment, Patents & Royalties. Spreter:Zymeworks: Employment, Patents & Royalties. Davies:Zymeworks Biopharmaceuticals: Employment. Wickman:Zymeworks: Employment.


2007 ◽  
Vol 104 (52) ◽  
pp. 20878-20883 ◽  
Author(s):  
J.-D. Bouaziz ◽  
K. Yanaba ◽  
G. M. Venturi ◽  
Y. Wang ◽  
R. M. Tisch ◽  
...  

Diabetologia ◽  
2018 ◽  
Vol 61 (6) ◽  
pp. 1397-1410 ◽  
Author(s):  
Larissa C. Da Rosa ◽  
Joanne Boldison ◽  
Evy De Leenheer ◽  
Joanne Davies ◽  
Li Wen ◽  
...  

2007 ◽  
Vol 56 (10) ◽  
pp. 1551-1563 ◽  
Author(s):  
Christian Brandl ◽  
Cornelia Haas ◽  
Sandrine d’Argouges ◽  
Tanja Fisch ◽  
Peter Kufer ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4638-4638 ◽  
Author(s):  
Ursula B. Jeffry ◽  
Mohammad Luqman ◽  
Kay Huh ◽  
Julie Klinger ◽  
Werner Frings ◽  
...  

Abstract CD40 is highly expressed in B-cell malignancies and its activation is a growth and survival signal for these cells. We have generated a novel, highly potent, fully human anti-CD40 IgG1 monoclonal antibody, CHIR-12.12 using XenoMouse® mice (Abgenix, Inc), a strain of transgenic mice expressing fully human IgG antibodies. CHIR-12.12 has at least two mechanisms of cytotoxicity in vitro: it blocks CD40-ligand mediated CD40 activation and mediates B-cell killing by ADCC. In-vitro crossreactivity studies were performed and the antibody showed cross reactivity to non-human primate but not to rodent tissue. To investigate the potential long-term toxicity of the antibody and to observe the recovery of the induced B-cell depletion, a single-dose toxicity study was conducted in cynomolgus monkeys. Three animals per sex per group (two animals per sex in control group) were given a single intravenous dose of 0, 10, or 100 mg/kg CHIR-12.12 and were monitored for 23 weeks post dosing. In addition to a detailed physical examination, samples for clinical chemistry, hematology and coagulation parameters as well as urinalysis were taken every 2 weeks. FACS analyses were performed every two weeks to monitor changes in B-cell, T-cell subpopulation and NK-cell numbers. In addition, CD40 expression and antigen saturation were measured on the remaining B-cell populations over 23 weeks. To investigate T-cell activation and T-cell function after CHIR-12.12 dosing, CD25 and CD40 expression on T-cells were monitored and at Week 23 a Mixed Lymphocyte Reaction (MLR) test was performed. The results of the study showed that a single dose of CHIR-12.12 was well tolerated and did not elicit any adverse clinical signs or effect on body weights, clinical pathology and hematology including differential blood counts. Immunophenotyping analysis showed a dramatic reduction of B-cells after dosing. The remaining B-cells mostly consist of the CD20high CD40low population, which is a population uniquely found in cynomolgus monkey blood but not in human blood (Vugmeyster et al.; Cytometry. 2003 Apr;52A). Both treatment groups showed a recovery of B-cells to normal pre-dose values in the majority of animals 19 weeks after dosing. In contrast, the average CHIR-12.12 antigen saturation level returned to pre-dose values in the low dose group after 11 weeks and in the high dose group after 19 weeks. T-cell counts (CD3, CD4 and CD8 subpopulation) showed no treatment related changes in absolute cell numbers throughout the study. No changes in T-cell activation measured by CD25 and CD40 expression were detected. T-cell responsiveness was not altered as measured by MLR. The pharmacokinetic analysis showed that, consistent with other human IgG antibodies, CHIR-12.12 had a small volume of distribution, limited to the blood volume, and was slowly eliminated from the circulation. Dose-dependency was observed with the area under serum concentration-time curve (AUC) increasing in a greater than dose-proportional fashion. Likewise, the half-life also increased with dose, ranging from 10.3 days at 10 mg/kg to 16.8 days at 100 mg/kg. In conclusion, all animals remained healthy and no evidence of immune impairment other then the expected reversible B-cell depletion was detected with a single dose of CHIR-12.12 up to 100 mg/kg. These results support the clinical development of CHIR-12.12 antibody for treatment of B-cell malignancies.


Sign in / Sign up

Export Citation Format

Share Document