Network


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

Hotspot


Dive into the research topics where Stephane Olland is active.

Publication


Featured researches published by Stephane Olland.


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.


mAbs | 2012

Improving the pharmacokinetic properties of biologics by fusion to an anti-HSA shark VNAR domain

Mischa R. Müller; Kenneth Saunders; Christopher Grace; Macy Jin; Nicole Piche-Nicholas; John Steven; Ronan O'Dwyer; Leeying Wu; Lam Khetemenee; Yulia Vugmeyster; Timothy P. Hickling; Lioudmila Tchistiakova; Stephane Olland; Davinder Gill; Allan Jensen; Caroline Barelle

Advances in recombinant antibody technology and protein engineering have provided the opportunity to reduce antibodies to their smallest binding domain components and have concomitantly driven the requirement for devising strategies to increase serum half-life to optimise drug exposure, thereby increasing therapeutic efficacy. In this study, we adopted an immunization route to raise picomolar affinity shark immunoglobulin new antigen receptors (IgNARs) to target human serum albumin (HSA). From our model shark species, Squalus acanthias, a phage display library encompassing the variable binding domain of IgNAR (VNAR) was constructed, screened against target, and positive clones were characterized for affinity and specificity. N-terminal and C-terminal molecular fusions of our lead hit in complex with a naïve VNAR domain were expressed, purified and exhibited the retention of high affinity binding to HSA, but also cross-selectivity to mouse, rat and monkey serum albumin both in vitro and in vivo. Furthermore, the naïve VNAR had enhanced pharmacokinetic (PK) characteristics in both N- and C-terminal orientations and when tested as a three domain construct with naïve VNAR flanking the HSA binding domain at both the N and C termini. Molecules derived from this platform technology also demonstrated the potential for clinical utility by being available via the subcutaneous route of delivery. This study thus demonstrates the first in vivo functional efficacy of a VNAR binding domain with the ability to enhance PK properties and support delivery of multifunctional therapies.


Bioconjugate Chemistry | 2012

Pharmacokinetic, biodistribution, and biophysical profiles of TNF nanobodies conjugated to linear or branched poly(ethylene glycol).

Yulia Vugmeyster; Clifford Entrican; Alison Joyce; Rosemary Lawrence-Henderson; Beth Leary; Christopher S. Mahoney; Himakshi K. Patel; Stephen W. Raso; Stephane Olland; Martin Hegen; Xin Xu

Covalent attachment of poly(ethylene glycol) (PEG) to therapeutic proteins has been used to prolong in vivo exposure of therapeutic proteins. We have examined pharmacokinetic, biodistribution, and biophysical profiles of three different tumor necrosis factor alpha (TNF) Nanobody-40 kDa PEG conjugates: linear 1 × 40 KDa, branched 2 × 20 kDa, and 4 × 10 kDa conjugates. In accord with earlier reports, the superior PK profile was observed for the branched versus linear PEG conjugates, while all three conjugates had similar potency in a cell-based assay. Our results also indicate that (i) a superior PK profile of branched versus linear PEGs is likely to hold across species, (ii) for a given PEG size, the extent of PEG branching affects the PK profile, and (iii) tissue penetration may differ between linear and branched PEG conjugates in a tissue-specific manner. Biophysical analysis (R(g)/R(h) ratio) demonstrated that among the three protein-PEG conjugates the linear PEG conjugate had the most extended time-average conformation and the most exposed surface charges. We hypothesized that these biophysical characteristics of the linear PEG conjugate accounts for relatively less optimal masking of sites involved in elimination of the PEGylated Nanobodies (e.g., intracellular uptake and proteolysis), leading to lower in vivo exposure compared to the branched PEG conjugates. However, additional studies are needed to test this hypothesis.


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.


Rheumatology | 2011

Distinct in vitro binding properties of the anti-CD20 small modular immunopharmaceutical 2LM20-4 result in profound and sustained in vivo potency in cynomolgus monkeys

Cheryl Nickerson-Nutter; Lioudmila Tchistiakova; Nilufer Seth; Marion Kasaian; Barbara Sibley; Stephane Olland; Richard Zollner; William A. Brady; Kendall M. Mohler; Peter Robert Baum; Alan Wahl; Deborah Herber; Yulia Vugmeyster; David Wensel; Neil M. Wolfman; Davinder Gill; Mary Collins; Kyri Dunussi-Joannopoulos

Objectives. To characterize the in vitro binding and effector function properties of CD20-directed small modular immunopharmaceutical (SMIP) 2LM20-4, and to compare its in vivo B-cell depletion activity with the mutated 2LM20-4 P331S [no in vitro complement-dependent cytotoxicity (CDC)] and rituximab in cynomolgus monkeys. Methods. Direct binding is examined in flow cytometry, confocal microscopy, scatchard and lipid raft assays. Effector function assays include CDC and Fc-mediated cellular toxicity. In the 6-month-long in vivo B-cell depletion study, single i.v. dosages of 1 or 10u2009mg/kg of anti-CD20 proteins were administered to monkeys and B-cell counts were monitored in peripheral blood, bone marrow and lymph nodes. Results. 2LM20-4 has lower saturation binding to human primary B cells and recruits fewer CD20 molecules into lipid rafts compared with rituximab; however, it induces higher in vitro CDC. In competitive binding, 2LM20-4 only partially displaces rituximab, suggesting that it binds to a fraction of CD20 molecules within certain locations of the plasma membrane as compared with rituximab. In monkeys, 2LM20-4 had more sustained B-cell depletion activity than rituximab in peripheral blood and had significantly more profound and sustained activity than 2LM20-4 P331S and rituximab in the lymph nodes. Conclusions. SMIP 2LM20-4, which binds to a fraction of CD20 molecules as compared with rituximab, has more potent in vitro CDC, and more potent and sustained B-cell depletion activity in cynomolgus monkeys. Our work has considerable clinical relevance since it provides novel insights related to the emerging B-cell depletion therapies in autoimmune diseases.


International Journal of Mass Spectrometry | 2012

Primary sequence determination of a monoclonal antibody against α-synuclein using a novel mass spectrometry-based approach

Eric Sousa; Stephane Olland; Heather H. Shih; Kim Marquette; Robert Martone; Zhijian Lu; Janet E. Paulsen; Davinder Gill; Tao He


Archive | 2011

Modified single domain antigen binding molecules and uses thereof

Martin Hegen; Stephane Olland; Yulia Vugmeyster; Xin Xu


Archive | 2011

Molécules de liaison à un antigène à domaine unique modifiées et leurs utilisations

Martin Hegen; Stephane Olland; Yulia Vugmeyster; Xin Xu


The FASEB Journal | 2008

Role of IL-21R in the pathogenesis of murine models of SLE

Tatyana Andreyeva; Heath M. Guay; Christie Damphousse; Laird Bloom; Stephane Olland; Tom Brown; Cheryl Nickerson-Nutter; Mary Collins; Debbie Young


Cytokine | 2008

110 Role of IL21R in the pathogenesis of murine models of SLE

Leslie Lowe; Heath M. Guay; Tatyana Andreyeva; Christie Damphousse; Nancy Stedman; Laird Bloom; Stephane Olland; Tom Brown; Cheryl Nickerson-Nutter; Mary Collins; Debbie Young

Collaboration


Dive into the Stephane Olland's collaboration.

Top Co-Authors

Avatar

Xin Xu

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Mary Collins

University College London

View shared research outputs
Top Co-Authors

Avatar

Heath M. Guay

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge