Network


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

Hotspot


Dive into the research topics where Diego Ellerman is active.

Publication


Featured researches published by Diego Ellerman.


Nature Biotechnology | 2013

Bispecific antibodies with natural architecture produced by co-culture of bacteria expressing two distinct half-antibodies

Christoph Spiess; Mark Merchant; Arthur Jyh-Yen Huang; Zhong Zheng; Nai-Ying Yang; Jing Peng; Diego Ellerman; Whitney Shatz; Dorothea Reilly; Daniel G. Yansura; Justin Scheer

By enabling the simultaneous engagement of two distinct targets, bispecific antibodies broaden the potential utility of antibody-based therapies. However, bispecific-antibody design and production remain challenging, owing to the need to incorporate two distinct heavy and light chain pairs while maintaining natural nonimmunogenic antibody architecture. Here we present a bispecific-antibody production strategy that relies on co-culture of two bacterial strains, each expressing a half-antibody. Using this approach, we produce 28 unique bispecific antibodies. A bispecific antibody against the receptor tyrosine kinases MET and EGFR binds both targets monovalently, inhibits their signaling, and suppresses MET and EGFR-driven cell and tumor growth. Our strategy allows rapid generation of bispecific antibodies from any two existing antibodies and yields milligram to gram quantities of bispecific antibodies sufficient for a wide range of discovery and preclinical applications.


Cancer Research | 2014

Antitumor Efficacy of a Bispecific Antibody That Targets HER2 and Activates T Cells

Teemu T. Junttila; Ji Li; Jennifer Johnston; Maria Hristopoulos; Robyn Clark; Diego Ellerman; Bu-Er Wang; Yijin Li; Mary Mathieu; Guangmin Li; Judy Young; Elizabeth Luis; Gail Lewis Phillips; Eric Stefanich; Christoph Spiess; Andrew G. Polson; Bryan Irving; Justin Scheer; Melissa R. Junttila; Mark S. Dennis; Robert F. Kelley; Klara Totpal; Allen Ebens

Clinical results from the latest strategies for T-cell activation in cancer have fired interest in combination immunotherapies that can fully engage T-cell immunity. In this study, we describe a trastuzumab-based bispecific antibody, HER2-TDB, which targets HER2 and conditionally activates T cells. HER2-TDB specifically killed HER2-expressing cancer cells at low picomolar concentrations. Because of its unique mechanism of action, which is independent of HER2 signaling or chemotherapeutic sensitivity, HER2-TDB eliminated cells refractory to currently approved HER2 therapies. HER2-TDB exhibited potent antitumor activity in four preclinical model systems, including MMTV-huHER2 and huCD3 transgenic mice. PD-L1 expression in tumors limited HER2-TDB activity, but this resistance could be reversed by anti-PD-L1 treatment. Thus, combining HER2-TDB with anti-PD-L1 yielded a combination immunotherapy that enhanced tumor growth inhibition, increasing the rates and durability of therapeutic response.


Science Translational Medicine | 2015

Anti-CD20/CD3 T cell–dependent bispecific antibody for the treatment of B cell malignancies

Liping L. Sun; Diego Ellerman; Mary Mathieu; Maria Hristopoulos; Xiaocheng Chen; Yijin Li; Xiao-Jie Yan; Robyn Clark; Arthur E. Reyes; Eric Stefanich; Elaine Mai; Judy Young; Clarissa Johnson; Mahrukh Huseni; Xinhua Wang; Yvonne Chen; Peiyin Wang; Hong Wang; Noel Dybdal; Yu-Waye Chu; Nicholas Chiorazzi; Justin Scheer; Teemu T. Junttila; Klara Totpal; Mark S. Dennis; Allen Ebens

Anti-CD20/CD3 T cell–dependent bispecific antibodies may be useful for the treatment of B cell malignancies. Two-headed cancer therapy Immunotherapeutic approaches harness either humoral (antibody-mediated) or cellular (T cell–mediated) immunity to fight cancer. Sun et al. combine these approaches by designing a CD3/CD20 TDB (T cell–dependent bispecific), a dual-targeted antibody that recruits T cells to CD20-expressing cells. Their humanized TDB induces T cells to kill primary patient leukemia and lymphoma cells both in vitro and in a mouse model and can deplete CD20-expressing B cells in a macaque model with similar properties as conventional antibodies. If these data hold true in clinical studies, this CD20/CD3 TDB could add to our expanding arsenal of cancer immunotherapeutics. Bispecific antibodies and antibody fragments in various formats have been explored as a means to recruit cytolytic T cells to kill tumor cells. Encouraging clinical data have been reported with molecules such as the anti-CD19/CD3 bispecific T cell engager (BiTE) blinatumomab. However, the clinical use of many reported T cell–recruiting bispecific modalities is limited by liabilities including unfavorable pharmacokinetics, potential immunogenicity, and manufacturing challenges. We describe a B cell–targeting anti-CD20/CD3 T cell–dependent bispecific antibody (CD20-TDB), which is a full-length, humanized immunoglobulin G1 molecule with near-native antibody architecture constructed using “knobs-into-holes” technology. CD20-TDB is highly active in killing CD20-expressing B cells, including primary patient leukemia and lymphoma cells both in vitro and in vivo. In cynomolgus monkeys, CD20-TDB potently depletes B cells in peripheral blood and lymphoid tissues at a single dose of 1 mg/kg while demonstrating pharmacokinetic properties similar to those of conventional monoclonal antibodies. CD20-TDB also exhibits activity in vitro and in vivo in the presence of competing CD20-targeting antibodies. These data provide rationale for the clinical testing of CD20-TDB for the treatment of CD20-expressing B cell malignancies.


Cancer Cell | 2017

Membrane-Proximal Epitope Facilitates Efficient T Cell Synapse Formation by Anti-FcRH5/CD3 and Is a Requirement for Myeloma Cell Killing

Ji Li; Nicola J. Stagg; Jennifer Johnston; Michael J. Harris; Sam A. Menzies; Danielle Dicara; Vanessa Clark; Maria Hristopoulos; Ryan Cook; Dionysos Slaga; Rin Nakamura; Luke McCarty; Siddharth Sukumaran; Elizabeth Luis; Zhengmao Ye; Thomas D. Wu; Teiko Sumiyoshi; Dimitry M. Danilenko; Genee Y. Lee; Klara Totpal; Diego Ellerman; Isidro Hotzel; John Robert James; Teemu T. Junttila

Summary The anti-FcRH5/CD3 T cell-dependent bispecific antibody (TDB) targets the B cell lineage marker FcRH5 expressed in multiple myeloma (MM) tumor cells. We demonstrate that TDBs trigger T cell receptor activation by inducing target clustering and exclusion of CD45 phosphatase from the synapse. The dimensions of the target molecule play a key role in the efficiency of the synapse formation. The anti-FcRH5/CD3 TDB kills human plasma cells and patient-derived myeloma cells at picomolar concentrations and results in complete depletion of B cells and bone marrow plasma cells in cynomolgus monkeys. These data demonstrate the potential for the anti-FcRH5/CD3 TDB, alone or in combination with inhibition of PD-1/PD-L1 signaling, in the treatment of MM and other B cell malignancies.


Blood | 2017

An anti-CD3/anti–CLL-1 bispecific antibody for the treatment of acute myeloid leukemia

Steven R. Leong; Siddharth Sukumaran; Maria Hristopoulos; Klara Totpal; Shannon Stainton; Elizabeth Lu; Alfred Wong; Lucinda Tam; Robert J. Newman; Brian R. Vuillemenot; Diego Ellerman; Chen Gu; Mary Mathieu; Mark S. Dennis; Allen Nguyen; Bing Zheng; Crystal Zhang; Genee Lee; Yu-Waye Chu; Rodney A. Prell; Kedan Lin; Steven T. Laing; Andrew G. Polson

Acute myeloid leukemia (AML) is a major unmet medical need. Most patients have poor long-term survival, and treatment has not significantly changed in 40 years. Recently, bispecific antibodies that redirect the cytotoxic activity of effector T cells by binding to CD3, the signaling component of the T-cell receptor, and a tumor target have shown clinical activity. Notably, blinatumomab is approved to treat relapsed/refractory acute lymphoid leukemia. Here we describe the design, discovery, pharmacologic activity, pharmacokinetics, and safety of a CD3 T cell-dependent bispecific (TDB) full-length human IgG1 therapeutic antibody targeting CLL-1 that could potentially be used in humans to treat AML. CLL-1 is prevalent in AML and, unlike other targets such as CD33 and CD123, is not expressed on hematopoietic stem cells providing potential hematopoietic recovery. We selected a high-affinity monkey cross-reactive anti-CLL-1 arm and tested several anti-CD3 arms that varied in affinity, and determined that the high-affinity CD3 arms were up to 100-fold more potent in vitro. However, in mouse models, the efficacy differences were less pronounced, probably because of prolonged exposure to TDB found with lower-affinity CD3 TDBs. In monkeys, assessment of safety and target cell depletion by the high- and low-affinity TDBs revealed that only the low-affinity CD3/CLL1 TDB was well tolerated and able to deplete target cells. Our data suggest that an appropriately engineered CLL-1 TDB could be effective in the treatment of AML.


mAbs | 2017

Efficient production of bispecific IgG of different isotypes and species of origin in single mammalian cells

Michael Dillon; Yiyuan Yin; Jianhui Zhou; Luke McCarty; Diego Ellerman; Dionysos Slaga; Teemu T. Junttila; Guanghui Han; Wendy Sandoval; Meric Ovacik; Kedan Lin; Zhilan Hu; Amy Shen; Jacob E. Corn; Christoph Spiess; Paul Carter

ABSTRACT Bispecific IgG production in single host cells has been a much sought-after goal to support the clinical development of these complex molecules. Current routes to single cell production of bispecific IgG include engineering heavy chains for heterodimerization and redesign of Fab arms for selective pairing of cognate heavy and light chains. Here, we describe novel designs to facilitate selective Fab arm assembly in conjunction with previously described knobs-into-holes mutations for preferential heavy chain heterodimerization. The top Fab designs for selective pairing, namely variants v10 and v11, support near quantitative assembly of bispecific IgG in single cells for multiple different antibody pairs as judged by high-resolution mass spectrometry. Single-cell and in vitro-assembled bispecific IgG have comparable physical, in vitro biological and in vivo pharmacokinetics properties. Efficient single-cell production of bispecific IgG was demonstrated for human IgG1, IgG2 and IgG4 thereby allowing the heavy chain isotype to be tailored for specific therapeutic applications. Additionally, a reverse chimeric bispecific IgG2a with humanized variable domains and mouse constant domains was generated for preclinical proof-of-concept studies in mice. Efficient production of a bispecific IgG in stably transfected mammalian (CHO) cells was shown. Individual clones with stable titer and bispecific IgG composition for >120 days were readily identified. Such long-term cell line stability is needed for commercial manufacture of bispecific IgG. The single-cell bispecific IgG designs developed here may be broadly applicable to biotechnology research, including screening bispecific IgG panels, and to support clinical development.


mAbs | 2016

Precise quantification of mixtures of bispecific IgG produced in single host cells by liquid chromatography-Orbitrap high-resolution mass spectrometry

Yiyuan Yin; Guanghui Han; Jianhui Zhou; Michael Dillon; Luke McCarty; Lou Gavino; Diego Ellerman; Christoph Spiess; Wendy Sandoval; Paul Carter

ABSTRACT Bispecific IgG are heterotetramers comprising 2 pairs of heavy and light chains. Co-expression of the 4 component chains in a single host cell typically yields the desired bispecific IgG plus up to 9 additional incorrect chain pairings. Several protein engineering strategies have been reported to facilitate the heterodimerization of antibody heavy chains or cognate pairing of antibody heavy and light chains. These technologies have been used to direct the efficient assembly of bispecific IgG in single host cells and minimize unwanted chain pairings. When purifying bispecific IgGs, the identification and quantification of low levels of closely related IgG contaminants are substantial analytical challenges. Here we have developed a robust high-throughput method for quantitative analysis of bispecific IgG preparations using novel online liquid chromatography in conjunction with an extended mass range Orbitrap-based high-resolution mass spectrometer. A mathematical method was developed to estimate the yields of the 2 isobaric species, namely the desired bispecific IgG and the light chain-scrambled IgG. The analytical methods described herein are anticipated to be broadly applicable to the development of bispecific IgG as drugs and potentially to other complex next-generation biotherapeutics.


Analytical Chemistry | 2016

Characterization of Chain Pairing Variants of Bispecific IgG Expressed in a Single Host Cell by High-Resolution Native and Denaturing Mass Spectrometry

Luis Schachner; Guanghui Han; Michael Patrick Dillon; Jianhui Zhou; Luke McCarty; Diego Ellerman; Yiyuan Yin; Christoph Spiess; Jennie R. Lill; Paul Carter; Wendy Sandoval

Bispecific antibodies, including bispecific IgG, show some promise in clinical trials as a means to extend the therapeutic potential of antibodies. Bispecific IgG can be made by separate expression and purification of each parent half antibody followed by in vitro reconstitution. Generating bispecific IgG by coexpression of two different light and heavy chains in a single host cell is potentially more efficient because it obviates the need for two separate cell lines and purification processes. However, this workflow may produce unwanted mispaired IgG species in addition to the desired bispecific IgG. Development and identification of designs that facilitate cognate light chain pairing may benefit from more refined methods to identify and quantify low levels of mispaired IgG. Using an anti-IL-4/IL-13 bispecific IgG, a mass spectrometric characterization method was developed using native or denaturing conditions by direct infusion into an Exactive Plus Extended Mass Range Orbitrap instrument. The high mass resolving power of the instrument allows unambiguous identification and accurate quantification of all light and heavy chain pairing variants in a mixture of bispecific IgG assembled in vivo upon coexpression down to 1% impurity. Preferential pairing of the anti-IL-13 light chain to its cognate heavy chain was observed, which may be leveraged to guide the design of a single-cell solution for streamlined production of bispecific IgG. Additionally, the utility of native mass spectrometry in deconvoluting complex antibody mixtures and in antigen-binding experiments to understand the contribution of doubly light chain mispaired bispecific IgG was demonstrated.


Archive | 2011

Generation of Bispecific Antibodies by Chemical Conjugation

Diego Ellerman; Justin Scheer

Bispecifc antibodies (bsAbs) are emerging as a promising new class of biotherapeutics. Although Ig domain fusion by DNA engineering is the prevalent methodology for producing bsAbs, a significant number of studies are being performed with chemically crosslinked Abs. By using different starting material and various conjugation strategies, bispecific fragments, full-length Abs, or combinations thereof can be generated. Two types or crosslinkers are used for the generation of bsAbs: hetero- or homobifunctional. Here we compare the pros and cons of the two types and we describe the use of the crosslinkers succinimidyl -3(2-pyridylthiol)propionate (SPDP), Traut’s reagent,sulpho-[succinimidyl-4-(N-maleimidomethyl)-4-cyclohexane-1-carboxylate] (SMCC), o-phenylenedimaleimide (o-PDM). We also review the clinical studies carried out with some of the products generated by these methodologies. The main hurdle for the transfer of chemically conjugated bsAbs to clinical applications has been the low yield and heterogeneity of the products. In this chapter we describe the generation of bisFabs, crosslinked Fabs with engineered Cys residues, to improve the efficiency and quality of bsAbs. We also discuss the use of non-natural amino acids along with click chemistry reactions and other approaches for the addition of new functionalities into bsAbs. These new technologies may help to improve both the yield and the homogeneity of bsAbs as well as their pharmacokinetics properties, paving the way for future clinical applications.


mAbs | 2017

bisFabs: Tools for rapidly screening hybridoma IgGs for their activities as bispecific antibodies

Sanket Patke; Ji Li; Peiyin Wang; Dion Slaga; Jennifer Johnston; Sunil Bhakta; Siler Panowski; Liping L. Sun; Teemu T. Junttila; Justin Scheer; Diego Ellerman

ABSTRACT Bispecific antibodies are a growing class of therapeutic molecules. Many of the current bispecific formats require DNA engineering to convert the parental monoclonal antibodies into the final bispecific molecules. We describe here a method to generate bispecific molecules from hybridoma IgGs in 3–4 d using chemical conjugation of antigen-binding fragments (Fabs) (bisFabs). Proteolytic digestion conditions for each IgG isotype were analyzed to optimize the yield and quality of the final conjugates. The resulting bisFabs showed no significant amounts of homodimers or aggregates. The predictive value of murine bisFabs was tested by comparing the T-cell redirected cytotoxic activity of a panel of antibodies in either the bisFab or full-length IgG formats. A variety of antigens with different structures and expression levels was used to extend the comparison to a wide range of binding geometries and antigen densities. The activity observed for different murine bisFabs correlated with those observed for the full-length IgG format across multiple different antigen targets, supporting the use of bisFabs as a screening tool. Our method may also be used for the screening of bispecific antibodies with other mechanisms of action, allowing for a more rapid selection of lead therapeutic candidates.

Collaboration


Dive into the Diego Ellerman'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
Researchain Logo
Decentralizing Knowledge