Network


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

Hotspot


Dive into the research topics where Donmienne Doen Mun Leung is active.

Publication


Featured researches published by Donmienne Doen Mun Leung.


mAbs | 2015

Balancing charge in the complementarity-determining regions of humanized mAbs without affecting pI reduces non-specific binding and improves the pharmacokinetics

Amita Datta-Mannan; Arunkumar Thangaraju; Donmienne Doen Mun Leung; Ying Tang; Derrick Ryan Witcher; Jirong Lu; Victor J. Wroblewski

Lowering the isoelectric point (pI) through engineering the variable region or framework of an IgG can improve its exposure and half-life via a reduction in clearance mediated through non-specific interactions. As such, net charge is a potentially important property to consider in developing therapeutic IgG molecules having favorable pharmaceutical characteristics. Frequently, it may not be possible to shift the pI of monoclonal antibodies (mAbs) dramatically without the introduction of other liabilities such as increased off-target interactions or reduced on-target binding properties. In this report, we explored the influence of more subtle modifications of molecular charge on the in vivo properties of an IgG1 and IgG4 monoclonal antibody. Molecular surface modeling was used to direct residue substitutions in the complementarity-determining regions (CDRs) to disrupt positive charge patch regions, resulting in a reduction in net positive charge without affecting the overall pI of the mAbs. The effect of balancing the net positive charge on non-specific binding was more significant for the IgG4 versus the IgG1 molecule that we examined. This differential effect was connected to the degree of influence on cellular degradation in vitro and in vivo clearance, distribution and metabolism in mice. In the more extreme case of the IgG4, balancing the charge yielded an ∼7-fold improvement in peripheral exposure, as well as significantly reduced tissue catabolism and subsequent excretion of proteolyzed products in urine. Balancing charge on the IgG1 molecule had a more subtle influence on non-specific binding and yielded only a modest alteration in clearance, distribution and elimination. These results suggest that balancing CDR charge without affecting the pI can lead to improved mAb pharmacokinetics, the magnitude of which is likely dependent on the relative influence of charge imbalance and other factors affecting the molecules disposition.


mAbs | 2015

The interplay of non-specific binding, target-mediated clearance and FcRn interactions on the pharmacokinetics of humanized antibodies.

Amita Datta-Mannan; Jirong Lu; Derrick Ryan Witcher; Donmienne Doen Mun Leung; Ying Tang; Victor J. Wroblewski

The application of protein engineering technologies toward successfully improving antibody pharmacokinetics has been challenging due to the multiplicity of biochemical factors that influence monoclonal antibody (mAb) disposition in vivo. Physiological factors including interactions with the neonatal Fc receptor (FcRn) and specific antigen binding properties of mAbs, along with biophysical properties of the mAbs themselves play a critical role. It has become evident that applying an integrated approach to understand the relative contribution of these factors is critical to rationally guide and apply engineering strategies to optimize mAb pharmacokinetics. The study presented here evaluated the influence of unintended non-specific interactions on the disposition of mAbs whose clearance rates are governed predominantly by either non-specific (FcRn) or target-mediated processes. The pharmacokinetics of 8 mAbs representing a diverse range of these properties was evaluated in cynomolgus monkeys. Results revealed complementarity-determining region (CDR) charge patch engineering to decrease charge-related non-specific binding can have a significant impact on improving the clearance. In contrast, the influence of enhanced in vitro FcRn binding was mixed, and related to both the strength of charge interaction and the general mechanism predominant in governing the clearance of the particular mAb. Overall, improved pharmacokinetics through enhanced FcRn interactions were apparent for a CDR charge-patch normalized mAb which was affected by non-specific clearance. The findings in this report are an important demonstration that mAb pharmacokinetics requires optimization on a case-by-case basis to improve the design of molecules with increased therapeutic application.


Experimental Neurology | 2015

Increased brain bio-distribution and chemical stability and decreased immunogenicity of an engineered variant of GDNF.

Rosamund C. Smith; Linda Maureen O'Bryan; Pamela Jean Mitchell; Donmienne Doen Mun Leung; Mahmoud Ghanem; Jonathan M. Wilson; Jeffrey C. Hanson; Sandra Sossick; Jane Cooper; Lihua Huang; Kalpana M. Merchant; Jirong Lu; Michael J. O'Neill

Several lines of evidence indicate that Glial cell line-derived neurotrophic factor (GDNF) is a trophic factor for dopaminergic neurons. Direct parenchymal administration of GDNF is robustly neuroprotective and neurorestorative in multiple neurotoxin-based animal models (rat and non-human primate (NHP)) of Parkinsons Disease (PD), suggesting its potential as a therapeutic agent. Although small, open-label clinical trials of intra-putamenal administration of bacteria-derived, full length, wild type GDNF (GDNFwt) were efficacious in improving standardized behavioral scores, a double-blinded, randomized controlled trial failed to do so. We hypothesize that the lack of clinical efficacy of GDNFwt in the larger randomized trial was due to poor bio-distribution in the putamen and/or poor chemical stability while in the delivery device for prolonged time periods at 37°C. The development of neutralizing antibodies in some patients may also have been a contributing factor. GDNFv is an engineered form of GDNFwt, expressed and purified from mammalian cells, designed to overcome these limitations, including removal of the N-terminal heparin-binding domain to improve its diffusivity in brain parenchyma by reducing its binding to extracellular matrix (ECM), and key amino acid substitutions to improve chemical stability. Intra-striatal administration of a single injection of GDNFv in the rat produced significantly greater brain distribution than GDNFwt, consistent with reduced binding to ECM. Using liquid chromatography/mass spectrometry (LS/MS) methods GDNFv was shown to have improved chemical stability compared to GDNFwt when stored at 37°C for 4weeks. In addition, GDNFv resulted in lower predicted clinical immunogenicity compared to GDNFwt, as demonstrated by reduced CD4+ T cell proliferation and reduced IL-2-induced secretion in peripheral blood mononucleated cells collected from volunteers representing the worlds major histocompatibility complex (MHC) haplotypes. GDNFv was demonstrated to be pharmacologically equivalent to GDNFwt in the key parameters in vitro of GFRα1 receptor binding, c-Ret phosphorylation, neurite outgrowth, and in vivo in its ability to increase dopamine turnover (DA). GDNFv protected dopamine nerve terminals and neurons in a 6-hydroxy-dopamine (6-OHDA) rat model. In summary, we empirically demonstrate the superior properties of GDNFv compared to GDNFwt through enhanced bio-distribution and chemical stability concurrently with decreased predicted clinical immunogenicity while maintaining pharmacological and neurotrophic activity. These data indicate that GDNFv is an improved version of GDNF suitable for clinical assessment as a targeted regenerative therapy for PD.


Cancer Research | 2017

Abstract 3090: LY3207447, a tetravalent bispecific antibody targeting VEGFR2 and angiopoietin-2, provides a more efficient anti-angiogenic therapy and an alternative for combination

Jonathan Tetreault; Sudhakar Chintharlapalli; Donmienne Doen Mun Leung; Damien Gerald; Linda Lee; Rowena Almonte-Baldonado; Lysiane Huber; Jianghuai Xu; Bharathi Ramamurthy; Jennifer Pereira; Johnny E. Croy; Jirong Lu; Ling Liu

Angiopoietin-2 (Ang-2) is released from endothelial cells only in response to stimulus (e.g. wound healing, tumor growth) and facilitates blood vessel sprouting and inhibits pericyte-endothelial cell interaction via Tie2 signaling. In tumors, Ang-2 is up-regulated and acts together with the VEGF/VEGFR2 pathway to stimulate tumor angiogenesis and metastasis. While therapeutic intervention using antagonists to the VEGF/VEGFR2 pathway has proven to be successful in limiting disease progression in a number of different clinical settings, there is an obvious need for an improved response. In various preclinical mouse angiogenesis or xenograft models, the combination treatment with anti-Ang2 antibody and the VEGF/VEGFR blocker provided additional benefit over inhibiting the individual pathway. Here as an alternative to combo therapy, we have engineered a tetravalent IgG-scFv bispecific antibody, LY3207447. LY3207447 is an immunoglobulin G4 (IgG4) antibody, comprising of VEGFR2 antibody derived from ramucirumab and a C-terminally fused single-chain variable fragment (scFv) targeting Ang2. LY3207447 binds to both the extracellular domain of VEGFR2 and soluble Ang2 with high affinity and blocks binding of Ang2 to Tie2 and VEGF to VEGFR2, and therefore inhibits signaling. We have shown that LY3207447 blocks binding of human Ang-2 to human Tie2-Fc by an ELISA assay and neutralizes Ang-2 induced phospho-Tie-2, but not Ang-1 induced phospho-Tie-2 in CHO cells overexpressing Tie-2 receptor. Moreover, LY3207447 neutralizes human VEGF165-induced phospho-VEGFR2 stimulation, cord formation and cell proliferation in human endothelial colony forming cells (ECFCs) and human dermal microvascular endothelial cells (HMVEC-d). LY3207447 blocks human and cyno VEGFR2, but not rodent VEGFR2. For anti-Ang2 arm, LY3207447 blocks human, cyno and mouse Ang2. Pre-clinical evaluation of LY3207447 in mouse retinal angiogenesis model resulted in an abrogation of angiogenesis. Combination studies using a rodent-specific surrogate VEGFR2 blocking antibody, DC101, with parental Ang2 antibody from which the scFv was derived from, inhibited both tumor growth and metastasis, resulting in increased survival compared to monotherapies in mouse xenograft model. These data establish VEGFR2/Ang2 bispecific antibodies as a promising anti-angiogenic, anti-metastatic and anti-tumor agent for the treatment of cancer in combination with other therapies. Citation Format: Jonathan Tetreault, Sudhakar Chintharlapalli, Donmienne Leung, Damien Gerald, Linda Lee, Rowena Almonte-Baldonado, Lysiane Huber, Jianghuai Xu, Bharathi Ramamurthy, Jennifer Pereira, Johnny Croy, Jirong Lu, Ling Liu. LY3207447, a tetravalent bispecific antibody targeting VEGFR2 and angiopoietin-2, provides a more efficient anti-angiogenic therapy and an alternative for combination [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3090. doi:10.1158/1538-7445.AM2017-3090


Cancer Research | 2016

Abstract 3259: LY3127804, a novel anti-Angiopoietin-2 antibody in combination with an anti-VEGFR2 antibody potently inhibits angiogenesis, tumor growth and metastasis

Sudhakar Chintharlapalli; Johnny E. Croy; Donmienne Doen Mun Leung; Damien Gerald; Jirong Lu; Philip W. Iversen; Linda N. Lee; Lysiane Huber; Jonathan Tetreault; Rowena Almonte-Baldonado; Jianghuai Xu; Bharathi Ramamurthy; Jennifer Pereira; Chi-Kin Chow; Axel-Rainer Hanauske; Volker Wacheck; Laura E. Benjamin; Ling Liu

Angiopoeitin-2 (Ang2) is released from endothelial cells only in response to stimulus (e.g. wound healing, tumor growth) and facilitates blood vessel sprouting and inhibits pericyte-endothelial cell interaction via Tie2 signaling. Combination of an anti-Ang2 antibody and aflibercept, a VEGF trap, has been shown to inhibit tumor growth and decrease tumor vascularity in mouse xenograft tumor models (Daly et al., Cancer Res (2013) 73(1):108). Multiple investigational anti-Ang2 antibody therapies are currently in clinical trials. LY3127804 is a humanized and engineered IgG4 isotype antibody that selectively binds to Ang2 with high affinity and neutralizes Ang2 induced phospho-Tie2. LY3127804 inhibits sprouting angiogenesis and increases pericyte coverage in a mouse developmental retinal angiogenesis model and in mice bearing PC3 xenograft tumors. Combination of LY3127804 and DC101, a potent anti-VEGFR2 antibody, exhibits enhanced efficacy when compared to monotherapy in multiple patient derived xenograft models including NSCLC and ovarian cancers. Anti-Ang2 antibody monotherapy alone resulted in marginal reduction of tumor growth and improved overall survival, while DC101monotherapy had greater reduction in tumor volume with no survival benefit in MDA-MB-231 breast orthotopic model. Combination of anti-Ang2 antibody with anti-VEGFR2 antibody shows reduction in tumor volume and improved overall survival. This robust pre-clinical evidence supports testing the combination of anti-Ang2 and anti-VEGFR2 antibodies in the clinic. LY3127804 is currently in Phase 1 clinical trials (NCT02597036) Citation Format: Sudhakar R. Chintharlapalli, Johnny E. Croy, Donmienne Leung, Damien Gerald, Jirong Lu, Philip W. Iversen, Linda N. Lee, Lysiane Huber, Jonathan Tetreault, Rowena Almonte-Baldonado, Jianghuai Xu, Bharathi Ramamurthy, Jennifer A. Pereira, Chi-Kin Chow, Axel-Rainer Hanauske, Volker Wacheck, Laura Benjamin, Ling Liu. LY3127804, a novel anti-Angiopoietin-2 antibody in combination with an anti-VEGFR2 antibody potently inhibits angiogenesis, tumor growth and metastasis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3259.


Blood | 2013

LY2928057, An Antibody Targeting Ferroportin, Is a Potent Inhibitor Of Hepcidin Activity and Increases Iron Mobilization In Normal Cynomolgus Monkeys

Donmienne Doen Mun Leung; Karen A Hill; David C. De Rosa; Jianghuai Xu; Joseph Manetta; Victor J. Wroblewski; Robert J. Benschop


Archive | 2009

Anti-ferroportin 1 monoclonal antibodies and uses thereof

Donmienne Doen Mun Leung; Peng Luan; Joseph Manetta; Ying Tang; Derrick Ryan Witcher


Archive | 2009

Anti-hepcidin-25 selective antibodies and uses thereof

Donmienne Doen Mun Leung; Peng Luan; Ying Tang; Derrick Ryan Witcher; Pia Pauliina Yachi


Archive | 2014

Anti-TNF-anti-IL-17 bispecific antibodies

Barrett Allan; Andrew Lawrence Glasebrook; Jirong Lu; Ying Tang; Derrick Ryan Witcher; Donmienne Doen Mun Leung; Pia Pauliina Yachi; Andrew Charles Vendel


Archive | 2016

VEGFA/Ang2 Compounds

Donmienne Doen Mun Leung; Ying Tang; Peter Edward Vaillancourt; Jianghuai Xu

Collaboration


Dive into the Donmienne Doen Mun Leung's collaboration.

Top Co-Authors

Avatar

Ying Tang

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jirong Lu

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peng Luan

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ling Liu

Eli Lilly and Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge