Network


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

Hotspot


Dive into the research topics where Will Somers is active.

Publication


Featured researches published by Will Somers.


Neuron | 2004

Two N-terminal domains of Kv4 K(+) channels regulate binding to and modulation by KChIP1.

Robert H Scannevin; KeWei Wang; Flora Jow; Jennifer Megules; David Kopsco; Wade Edris; Karen C. Carroll; Qiang Lu; Weixin Xu; Zhangbao Xu; Alan H. Katz; Stephane Olland; Laura Lin; Meggin Taylor; Mark Stahl; Karl Malakian; Will Somers; Lydia Mosyak; Mark R. Bowlby; Pranab K. Chanda; Kenneth J. Rhodes

The family of calcium binding proteins called KChIPs associates with Kv4 family K(+) channels and modulates their biophysical properties. Here, using mutagenesis and X-ray crystallography, we explore the interaction between Kv4 subunits and KChIP1. Two regions in the Kv4.2 N terminus, residues 7-11 and 71-90, are necessary for KChIP1 modulation and interaction with Kv4.2. When inserted into the Kv1.2 N terminus, residues 71-90 of Kv4.2 are also sufficient to confer association with KChIP1. To provide a structural framework for these data, we solved the crystal structures of Kv4.3N and KChIP1 individually. Taken together with the mutagenesis data, the individual structures suggest that that the Kv4 N terminus is required for stable association with KChIP1, perhaps through a hydrophobic surface interaction, and that residues 71-90 in Kv4 subunits form a contact loop that mediates the specific association of KChIPs with Kv4 subunits.


Biotechnology and Bioengineering | 2012

Engineering novel Lec1 glycosylation mutants in CHO-DUKX cells: molecular insights and effector modulation of N-acetylglucosaminyltransferase I.

Xiaotian Zhong; Cecilia Cooley; Nilufer Seth; Z. Sean Juo; Ella Presman; Nicole Resendes; Ravi Kumar; Martin J Allen; Lidia Mosyak; Mark Stahl; Will Somers; Ronald Kriz

Many secreted or cell surface proteins are post‐translationally modified by carbohydrate chains which are a primary source of heterogeneity. The Lec1 mutant, which is defective in Golgi N‐acetylglucosaminyltransferase I (GnTI) activity, produces relatively homogeneous Man5GlcNAc2 glycan modifications, and is widely used for various applications. To facilitate the investigation of GnTI, its Man5 glycan endproduct, and the impact of Man5 on effector function, the present study has established several novel Lec1 mutants in dhfr− CHO–DUKX cells through chemical mutagenesis and lectin selection. A total of nine clonal lines exhibiting the Lec1‐phenotype are characterized, six of which harbor non‐sense mutations leading to a truncated GnTI, and three (R415K, D291N, and P138L) of which are novel loss‐of‐function sense mutations. Analysis of the rabbit GnTI structure (Unligil et al., 2000 ) indicates that D291 is the proposed catalytic base and R415 is a crucial residue in forming the substrate binding pocket, whereas P138 is key to maintaining two β strands in proximity to the substrate binding pocket. Computational modeling reveals that the oligomannose glycan backbone of a glycoprotein (the acceptor substrate) fits nicely into the unoccupied channel of the substrate binding pocket partly through hydrogen bonding with R415 and D291. This finding is consistent with the ordered sequential Bi Bi kinetic mechanism suggested for GnTI, in which binding of UDP–GlcNAc (the donor substrate)/Mn2+ induces conformational changes that promote acceptor binding. When an anti‐human CD20 antibody protein is stably expressed in one CHO–DUKX–Lec1 line, it is confirmed that N‐glycans are predominantly Man5GlcNAc2 and they do not contain an α1,6‐fucose linked to the innermost GlcNAc. Furthermore, this Man5GlcNAc2 modified antibody exhibits a significantly increased ADCC activity than the wild‐type protein, while displaying a lower CDC activity. The data support the hypothesis that modulating GnTI activity can influence antibody effector functions for proteins with an IgG1 immunoglobulin Fc domain. Biotechnol. Bioeng. 2012; 109:1723–1734.


Scientific Reports | 2016

Improved variants of SrtA for site-specific conjugation on antibodies and proteins with high efficiency

Long Chen; Justin M. Cohen; Xiaoda Song; Aishan Zhao; Zi Ye; Christine J. Feulner; Patrick Doonan; Will Somers; Laura Lin; Peng Chen

Sortase mediated ligation is a highly specific platform for conjugation that relies on the specificity of the transpeptidase Sortase A (SrtA) for short peptide sequences (LPXTG and GGG). SrtA retains its specificity while accepting a wide range of potential substrates, but its broad use is limited by the wild-type enzyme’s poor kinetics, which require large amounts of SrtA and extended reaction times for efficient conjugation. Prior explorations have aimed to improve the kinetics of SrtA with limited success. Herein we describe the discovery of further improved SrtA variants with increased efficiency for the conjugation reaction, and demonstrate their robustness in labelling proteins and antibodies in a site-specific manner. Our variants require significantly lower amounts of enzyme than WT SrtA and can be used to attach small molecules to the N or C-terminus of the heavy or light chain in antibodies with excellent yields. These improved variants can also be used for highly efficient site-specific PEGylation.


mAbs | 2013

CDR-restricted engineering of native human scFvs creates highly stable and soluble bifunctional antibodies for subcutaneous delivery

Brian J. Fennell; Barry McDonnell; Amy Tam; Lijun Chang; John Steven; Ian David Broadbent; Huilan Gao; Elizabeth Kieras; Jennifer Alley; Deborah Luxenberg; Jason Edmonds; Lori Fitz; Wenyan Miao; Matthew J. Whitters; Quintus G. Medley; Yongjing J Guo; Alfredo Darmanin-Sheehan; Bénédicte Autin; Deirdre Ní Shúilleabháin; Emma Cummins; Amy King; Mark Rh Krebs; Christopher Grace; Timothy P. Hickling; Angela Boisvert; Xiaotian Zhong; Matthew McKenna; Christopher Francis; Stephane Olland; Laird Bloom

While myriad molecular formats for bispecific antibodies have been examined to date, the simplest structures are often based on the scFv. Issues with stability and manufacturability in scFv-based bispecific molecules, however, have been a significant hindrance to their development, particularly for high-concentration, stable formulations that allow subcutaneous delivery. Our aim was to generate a tetravalent bispecific molecule targeting two inflammatory mediators for synergistic immune modulation. We focused on an scFv-Fc-scFv format, with a flexible (A4T)3 linker coupling an additional scFv to the C-terminus of an scFv-Fc. While one of the lead scFvs isolated directly from a naïve library was well-behaved and sufficiently potent, the parental anti-CXCL13 scFv 3B4 required optimization for affinity, stability, and cynomolgus ortholog cross-reactivity. To achieve this, we eschewed framework-based stabilizing mutations in favor of complementarity-determining region (CDR) mutagenesis and re-selection for simultaneous improvements in both affinity and thermal stability. Phage-displayed 3B4 CDR-mutant libraries were used in an aggressive “hammer-hug” selection strategy that incorporated thermal challenge, functional, and biophysical screening. This approach identified leads with improved stability and >18-fold, and 4,100-fold higher affinity for both human and cynomolgus CXCL13, respectively. Improvements were exclusively mediated through only 4 mutations in VL-CDR3. Lead scFvs were reformatted into scFv-Fc-scFvs and their biophysical properties ranked. Our final candidate could be formulated in a standard biopharmaceutical platform buffer at 100 mg/ml with <2% high molecular weight species present after 7 weeks at 4 °C and viscosity <15 cP. This workflow has facilitated the identification of a truly manufacturable scFv-based bispecific therapeutic suitable for subcutaneous administration.


Journal of Biological Chemistry | 2013

Pyroglutamate and O-Linked Glycan Determine Functional Production of Anti-IL17A and Anti-IL22 Peptide-Antibody Bispecific Genetic Fusions

Xiaotian Zhong; Elizabeth Kieras; Eric Sousa; Aaron M. D'Antona; J. Christian Baber; Tao He; Joel Desharnais; Lauren Diane Wood; Deborah Luxenberg; Mark Stahl; Ronald Kriz; Laura Lin; Will Somers; Lori Fitz; Jill F. Wright

Background: Protein biosynthesis and secretion are essential biological processes for therapeutic protein production. Results: Generating functional anti-IL17A and anti-IL22 peptide-antibody bispecific therapeutic proteins requires pyroglutamate addition and O-linked glycan removal. Conclusion: Post-translational modifications play critical roles in determining structure and function of therapeutic proteins. Significance: The peptide-antibody genetic fusion is promising for targeting multiple antigens in a single antibody-like molecule. Protein biosynthesis and extracellular secretion are essential biological processes for therapeutic protein production in mammalian cells, which offer the capacity for correct folding and proper post-translational modifications. In this study, we have generated bispecific therapeutic fusion proteins in mammalian cells by combining a peptide and an antibody into a single open reading frame. A neutralizing peptide directed against interleukin-17A (IL17A) was genetically fused to the N termini of an anti-IL22 antibody, through either the light chain, the heavy chain, or both chains. Although the resulting fusion proteins bound and inhibited IL22 with the same affinity and potency as the unmodified anti-IL22 antibody, the peptide modality in the fusion scaffold was not active in the cell-based assay due to the N-terminal degradation. When a glutamine residue was introduced at the N terminus, which can be cyclized to form pyroglutamate in mammalian cells, the IL17A neutralization activity of the fusion protein was restored. Interestingly, the mass spectroscopic analysis of the purified fusion protein revealed an unexpected O-linked glycosylation modification at threonine 5 of the anti-IL17A peptide. The subsequent removal of this post-translational modification by site-directed mutagenesis drastically enhanced the IL17A binding affinity and neutralization potency for the resulting fusion protein. These results provide direct experimental evidence that post-translational modifications during protein biosynthesis along secretory pathways play critical roles in determining the structure and function of therapeutic proteins produced by mammalian cells. The newly engineered peptide-antibody genetic fusion is promising for therapeutically targeting multiple antigens in a single antibody-like molecule.


FEBS Letters | 2009

Swift residue-screening identifies key N-glycosylated asparagines sufficient for surface expression of neuroglycoprotein Lingo-1.

Xiaotian Zhong; Jennifer Pocas; Yan Liu; Paul W. Wu; Lidia Mosyak; Will Somers; Ron Kriz

Advances in genomics and proteomics have generated the needs for the efficient identification of key residues for structure and function of target proteins. Here we report the utilization of a new residue‐screening approach, which combines a mammalian high‐throughput transient expression system with a PCR‐based expression cassette, for the study of the post‐translational modification. Applying this approach results in a quick identification of essential N‐glycosylation sites of a heavily glycosylated neuroglycoprotein Lingo‐1, which are sufficient for the support of its surface expression. These key N‐glycosylated sites uniquely locate on the concave surface of the elongated arc‐shape structure of the leucine‐rich repeat domain. The swift residue‐screening approach may provide a new strategy for structural and functional analysis.


Journal of Biotechnology | 2017

Mechanistic understanding of the cysteine capping modifications of antibodies enables selective chemical engineering in live mammalian cells

Xiaotian Zhong; Tao He; Amar S. Prashad; Wenge Wang; Justin Cohen; Darren Ferguson; Amy Tam; Eric Sousa; Laura Lin; Lioudmila Tchistiakova; Scott Gatto; Aaron M. D’Antona; Yen-Tung Luan; Weijun Ma; Richard Zollner; Jing Zhou; Bo Arve; Will Somers; Ronald Kriz

Protein modifications by intricate cellular machineries often redesign the structure and function of existing proteins to impact biological networks. Disulfide bond formation between cysteine (Cys) pairs is one of the most common modifications found in extracellularly-destined proteins, key to maintaining protein structure. Unpaired surface cysteines on secreted mammalian proteins are also frequently found disulfide-bonded with free Cys or glutathione (GSH) in circulation or culture, the mechanism for which remains unknown. Here we report that these so-called Cys-capping modifications take place outside mammalian cells, not in the endoplasmic reticulum (ER) where oxidoreductase-mediated protein disulfide formation occurs. Unpaired surface cysteines of extracellularly-arrived proteins such as antibodies are uncapped upon secretion before undergoing disulfide exchange with cystine or oxidized GSH in culture medium. This observation has led to a feasible way to selectively modify the nucleophilic thiol side-chain of cell-surface or extracellular proteins in live mammalian cells, by applying electrophiles with a chemical handle directly into culture medium. These findings provide potentially an effective approach for improving therapeutic conjugates and probing biological systems.


Journal of Biological Chemistry | 2016

A Combination of Structural and Empirical Analyses Delineates the Key Contacts Mediating Stability and Affinity Increases in an Optimized Biotherapeutic Single-chain Fv (scFv)

Chao Tu; Virginie Terraube; Amy Tam; Wayne Stochaj; Brian J. Fennell; Laura Lin; Mark Stahl; Edward R. LaVallie; Will Somers; William J. J. Finlay; Lydia Mosyak; Joel Bard; Orla Cunningham

Background: Antibody v-domains in scFv format often suffer from aggregation and stability issues that restrict formulation. Results: Structural and empirical analyses of an optimized scFv revealed that three VL-CDR3 mutations were sufficient to mediate significant stability and affinity improvements. Conclusion: scFv issues were resolved via removal of side-chain clashes at the VL/VH interface. Significance: CDR-restricted mutagenesis delivers stability-optimized molecules for high concentration dosing. Fully-human single-chain Fv (scFv) proteins are key potential building blocks of bispecific therapeutic antibodies, but they often suffer from manufacturability and clinical development limitations such as instability and aggregation. The causes of these scFv instability problems, in proteins that should be theoretically stable, remains poorly understood. To inform the future development of such molecules, we carried out a comprehensive structural analysis of the highly stabilized anti-CXCL13 scFv E10. E10 was derived from the parental 3B4 using complementarity-determining region (CDR)-restricted mutagenesis and tailored selection and screening strategies, and carries four mutations in VL-CDR3. High-resolution crystal structures of parental 3B4 and optimized E10 scFvs were solved in the presence and absence of human CXCL13. In parallel, a series of scFv mutants was generated to interrogate the individual contribution of each of the four mutations to stability and affinity improvements. In combination, these analyses demonstrated that the optimization of E10 was primarily mediated by removing clashes between both the VL and the VH, and between the VL and CXCL13. Importantly, a single, germline-encoded VL-CDR3 residue mediated the key difference between the stable and unstable forms of the scFv. This work demonstrates that, aside from being the critical mediators of specificity and affinity, CDRs may also be the primary drivers of biotherapeutic developability.


Acta Crystallographica Section F-structural Biology and Crystallization Communications | 2008

Expression, purification and crystallization of the ecto-enzymatic domain of rat E-NTPDase1 CD39

Xiaotian Zhong; Madhavan R. Buddha; Guido Guidotti; Ron Kriz; Will Somers; Lidia Mosyak

CD39 is a prototype member of the ecto-nucleoside triphosphate diphosphohydrolase family that hydrolyzes extracellular nucleoside diphosphates and triphosphates in the presence of divalent cations. Here, the expression, purification and crystallization of the ecto-enzymatic domain of rat CD39, sCD39, are described. The 67 kDa secreted soluble glycoprotein was recombinantly overexpressed in a glycosylation mutant CHO line, Lec.3.2.8.1, and purified from conditioned media. Diffraction-quality crystals of sCD39 were produced by the vapor-diffusion method using PEG 3350 and ammonium dihydrogen phosphate as precipitants. The enzyme crystallized in a primitive trigonal form in space group P3(2), with unit-cell parameters a = b = 118.1, c = 81.6 A and with two sCD39 copies in the asymmetric unit. Several low- to medium-resolution diffraction data sets were collected using an in-house X-ray source. Analysis of the intensity statistics showed that the crystals were invariably merohedrally twinned with a high twin fraction. For initial phasing, a molecular-replacement search was performed against the complete 3.2 A data set using a maximum-likelihood molecular-replacement method as implemented in Phaser. The initial model of the two sCD39 monomers was placed into the P3(2) lattice and rigid-body refined and position-minimized with PHENIX.


Scientific Reports | 2018

Glyco-engineered Long Acting FGF21 Variant with Optimal Pharmaceutical and Pharmacokinetic Properties to Enable Weekly to Twice Monthly Subcutaneous Dosing

Yan Weng; Tetsuya Ishino; Annette Sievers; Saswata Talukdar; Jeffrey R. Chabot; Amy Tam; Weili Duan; Kelvin M. Kerns; Eric Sousa; Tao He; Alison Logan; Darwin V. Lee; Dongmei Li; Yingjiang Zhou; Barbara L. Bernardo; Alison Joyce; Mania Kavosi; Denise M. O’Hara; Tracey Clark; Jie Guo; Craig Giragossian; Mark Stahl; Roberto A. Calle; Ron Kriz; Will Somers; Laura Lin

Pharmacological administration of FGF21 analogues has shown robust body weight reduction and lipid profile improvement in both dysmetabolic animal models and metabolic disease patients. Here we report the design, optimization, and characterization of a long acting glyco-variant of FGF21. Using a combination of N-glycan engineering for enhanced protease resistance and improved solubility, Fc fusion for further half-life extension, and a single point mutation for improving manufacturability in Chinese Hamster Ovary cells, we created a novel FGF21 analogue, Fc-FGF21[R19V][N171] or PF-06645849, with substantially improved solubility and stability profile that is compatible with subcutaneous (SC) administration. In particular, it showed a low systemic clearance (0.243 mL/hr/kg) and long terminal half-life (~200 hours for intact protein) in cynomolgus monkeys that approaches those of monoclonal antibodies. Furthermore, the superior PK properties translated into robust improvement in glucose tolerance and the effects lasted 14 days post single SC dose in ob/ob mice. PF-06645849 also caused greater body weight loss in DIO mice at lower and less frequent SC doses, compared to previous FGF21 analogue PF-05231023. In summary, the overall PK/PD and pharmaceutical profile of PF-06645849 offers great potential for development as weekly to twice-monthly SC administered therapeutic for chronic treatment of metabolic diseases.

Collaboration


Dive into the Will Somers's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge