Network


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

Hotspot


Dive into the research topics where Sandrine Guillard is active.

Publication


Featured researches published by Sandrine Guillard.


Journal of Experimental Medicine | 2012

Identification of broadly protective human antibodies to Pseudomonas aeruginosa exopolysaccharide Psl by phenotypic screening

Antonio DiGiandomenico; Paul Warrener; Sandrine Guillard; Peter Ravn; Ralph Minter; Maria Margarita Camara; Vignesh Venkatraman; Randall S. MacGill; Jia Lin; Qun Wang; Ashley E. Keller; Mladen Tomich; Lutz Jermutus; Michael P. McCarthy; David A. Melnick; JoAnn Suzich; C. Kendall Stover

A human antibody facilitates opsonophagocytic killing, inhibits attachment of Pseudomonas aeruginosa, and exerts protective effects in several animal models of P. aeruginosa infection.


Molecular Cancer | 2013

Combining phenotypic and proteomic approaches to identify membrane targets in a ‘triple negative’ breast cancer cell type

Steven Rust; Sandrine Guillard; Kris Sachsenmeier; Carl Hay; Max Davidson; Anders Karlsson; Roger Karlsson; Erin Brand; David Lowne; John Elvin; Matt Flynn; Gene Kurosawa; Robert E. Hollingsworth; Lutz Jermutus; Ralph Minter

BackgroundThe continued discovery of therapeutic antibodies, which address unmet medical needs, requires the continued discovery of tractable antibody targets. Multiple protein-level target discovery approaches are available and these can be used in combination to extensively survey relevant cell membranomes. In this study, the MDA-MB-231 cell line was selected for membranome survey as it is a ‘triple negative’ breast cancer cell line, which represents a cancer subtype that is aggressive and has few treatment options.MethodsThe MDA-MB-231 breast carcinoma cell line was used to explore three membranome target discovery approaches, which were used in parallel to cross-validate the significance of identified antigens. A proteomic approach, which used membrane protein enrichment followed by protein identification by mass spectrometry, was used alongside two phenotypic antibody screening approaches. The first phenotypic screening approach was based on hybridoma technology and the second was based on phage display technology. Antibodies isolated by the phenotypic approaches were tested for cell specificity as well as internalisation and the targets identified were compared to each other as well as those identified by the proteomic approach. An anti-CD73 antibody derived from the phage display-based phenotypic approach was tested for binding to other ‘triple negative’ breast cancer cell lines and tested for tumour growth inhibitory activity in a MDA-MB-231 xenograft model.ResultsAll of the approaches identified multiple cell surface markers, including integrins, CD44, EGFR, CD71, galectin-3, CD73 and BCAM, some of which had been previously confirmed as being tractable to antibody therapy. In total, 40 cell surface markers were identified for further study. In addition to cell surface marker identification, the phenotypic antibody screening approaches provided reagent antibodies for target validation studies. This is illustrated using the anti-CD73 antibody, which bound other ‘triple negative’ breast cancer cell lines and produced significant tumour growth inhibitory activity in a MDA-MB-231 xenograft model.ConclusionsThis study has demonstrated that multiple methods are required to successfully analyse the membranome of a desired cell type. It has also successfully demonstrated that phenotypic antibody screening provides a mechanism for rapidly discovering and evaluating antibody tractable targets, which can significantly accelerate the therapeutic discovery process.


Molecular Cancer | 2015

Identification of anti-tumour biologics using primary tumour models, 3-D phenotypic screening and image-based multi-parametric profiling.

Alan Sandercock; Steven Rust; Sandrine Guillard; Kris Sachsenmeier; Nick Holoweckyj; Carl Hay; Matt Flynn; Qihui Huang; Kuan Yan; Bram Herpers; Leo Price; Jo Soden; Jim Freeth; Lutz Jermutus; Robert E. Hollingsworth; Ralph Minter

BackgroundMonolayer cultures of immortalised cell lines are a popular screening tool for novel anti-cancer therapeutics, but these methods can be a poor surrogate for disease states, and there is a need for drug screening platforms which are more predictive of clinical outcome. In this study, we describe a phenotypic antibody screen using three-dimensional cultures of primary cells, and image-based multi-parametric profiling in PC-3 cells, to identify anti-cancer biologics against new therapeutic targets.MethodsScFv Antibodies and designed ankyrin repeat proteins (DARPins) were isolated using phage display selections against primary non-small cell lung carcinoma cells. The selected molecules were screened for anti-proliferative and pro-apoptotic activity against primary cells grown in three-dimensional culture, and in an ultra-high content screen on a 3-D cultured cell line using multi-parametric profiling to detect treatment-induced phenotypic changes. The targets of molecules of interest were identified using a cell-surface membrane protein array. An anti-CUB domain containing protein 1 (CDCP1) antibody was tested for tumour growth inhibition in a patient-derived xenograft model, generated from a stage-IV non-small cell lung carcinoma, with and without cisplatin.ResultsTwo primary non-small cell lung carcinoma cell models were established for antibody isolation and primary screening in anti-proliferative and apoptosis assays. These assays identified multiple antibodies demonstrating activity in specific culture formats. A subset of the DARPins was profiled in an ultra-high content multi-parametric screen, where 300 morphological features were measured per sample. Machine learning was used to select features to classify treatment responses, then antibodies were characterised based on the phenotypes that they induced. This method co-classified several DARPins that targeted CDCP1 into two sets with different phenotypes. Finally, an anti-CDCP1 antibody significantly enhanced the efficacy of cisplatin in a patient-derived NSCLC xenograft model.ConclusionsPhenotypic profiling using complex 3-D cell cultures steers hit selection towards more relevant in vivo phenotypes, and may shed light on subtle mechanistic variations in drug candidates, enabling data-driven decisions for oncology target validation. CDCP1 was identified as a potential target for cisplatin combination therapy.


Trends in Biotechnology | 2015

Engineering therapeutic proteins for cell entry: the natural approach

Sandrine Guillard; Ralph Minter; Ronald Jackson

Owing to the challenges of cell entry, protein-based therapies have so far been restricted to extracellular targets, whereas intracellular targets have been almost exclusively addressed by small molecules. The specificity and potency of proteins would enable them to be effective intracellular drugs, provided that the proteins are delivered efficiently to appropriate intracellular compartments within specific cell types. By mimicking the natural mechanisms of toxins and other natural proteins, new intracellular delivery systems are being developed, the first of which are showing clinical efficacy. This review highlights a range of ingenious approaches designed to adapt natural entry mechanisms to facilitate delivery of proteins and open up a range of validated intracellular targets to modulation by potent and specific therapeutic drugs.


Journal of Biomolecular Screening | 2012

Development of a Novel Ectonucleotidase Assay Suitable for High-Throughput Screening

Kris Sachsenmeier; Carl Hay; Erin Brand; Lori Clarke; Kim Rosenthal; Sandrine Guillard; Steven Rust; Ralph Minter; Robert E. Hollingsworth

5′-Ectonucleotidase (NT5E) catalyzes the conversion of adenosine monophosphate to adenosine and free phosphate. The role of this ectonucleotidase and its production of adenosine are linked with immune function, angiogenesis, and cancer. NT5E activity is typically assayed either by chromatographic quantification of substrates and products using high-performance liquid chromatography (HPLC) or by quantification of free phosphate using malachite green. These methods are not suitable for robust screening assays of NT5E activity. HPLC is not readily suitable for the rapid and efficient assay of multiple samples and malachite green is highly sensitive to the phosphate-containing buffers common in various media and sample buffers. Here the development and validation of a novel high-throughput ectonucleotidase screening assay are described, which makes use of a luciferase-based assay reagent, the Promega CellTiter-Glo kit, to measure the catabolism of AMP by NT5E. This multiwell plate-based assay facilitates the screening of potential ectonucleotidase antagonists and is unaffected by the presence of contaminating phosphate molecules present in screening samples.


Nature Communications | 2017

Structural and functional characterization of a DARPin which inhibits Ras nucleotide exchange.

Sandrine Guillard; Paulina Kolasinska-Zwierz; Judit É. Debreczeni; Jason Breed; Jing Zhang; Nicolas Bery; Rose Marwood; Jon Tart; Ross Overman; Pawel Stocki; Bina Mistry; Christopher Phillips; Terence H. Rabbitts; Ronald Jackson; Ralph Minter

Ras mutations are the oncogenic drivers of many human cancers and yet there are still no approved Ras-targeted cancer therapies. Inhibition of Ras nucleotide exchange is a promising new approach but better understanding of this mechanism of action is needed. Here we describe an antibody mimetic, DARPin K27, which inhibits nucleotide exchange of Ras. K27 binds preferentially to the inactive Ras GDP form with a Kd of 4 nM and structural studies support its selectivity for inactive Ras. Intracellular expression of K27 significantly reduces the amount of active Ras, inhibits downstream signalling, in particular the levels of phosphorylated ERK, and slows the growth in soft agar of HCT116 cells. K27 is a potent, non-covalent inhibitor of nucleotide exchange, showing consistent effects across different isoforms of Ras, including wild-type and oncogenic mutant forms.


Oncotarget | 2016

Phenotypic screening reveals TNFR2 as a promising target for cancer immunotherapy

Geoffrey S. Williams; Bina Mistry; Sandrine Guillard; Jane Coates Ulrichsen; Alan Sandercock; Jun Wang; Andrea González-Muñoz; Julie Parmentier; Chelsea Black; Jo Soden; Jim Freeth; Jelena Jovanović; Rebecca Leyland; Rafia S. Al-Lamki; Andrew J. Leishman; Steven J. Rust; Ross Stewart; Lutz Jermutus; John R. Bradley; Vahe Bedian; Viia Valge-Archer; Ralph Minter; Robert W. Wilkinson

Antibodies that target cell-surface molecules on T cells can enhance anti-tumor immune responses, resulting in sustained immune-mediated control of cancer. We set out to find new cancer immunotherapy targets by phenotypic screening on human regulatory T (Treg) cells and report the discovery of novel activators of tumor necrosis factor receptor 2 (TNFR2) and a potential role for this target in immunotherapy. A diverse phage display library was screened to find antibody mimetics with preferential binding to Treg cells, the most Treg-selective of which were all, without exception, found to bind specifically to TNFR2. A subset of these TNFR2 binders were found to agonise the receptor, inducing iκ-B degradation and NF-κB pathway signalling in vitro. TNFR2 was found to be expressed by tumor-infiltrating Treg cells, and to a lesser extent Teff cells, from three lung cancer patients, and a similar pattern was also observed in mice implanted with CT26 syngeneic tumors. In such animals, TNFR2-specific agonists inhibited tumor growth, enhanced tumor infiltration by CD8+ T cells and increased CD8+ T cell IFN-γ synthesis. Together, these data indicate a novel mechanism for TNF-α-independent TNFR2 agonism in cancer immunotherapy, and demonstrate the utility of target-agnostic screening in highlighting important targets during drug discovery.


Cancer Research | 2014

Abstract 4613: Identification of tumor necrosis factor receptor II as a regulatory T cell target for cancer immunotherapy using designed ankyrin repeat protein phenotypic selections

Geoff Williams; Judith Anderton; Vahe Bedian; Jane Coates Ulrichsen; Andrea González-Muñoz; Sandrine Guillard; Olivia Harris; James Hair; Andrew Leishman; Arthur Lewis; Jacques Moissan; Ralph Minter; Bina Mistry; Julie Parmentier; Edmund Poon; Amy Popple; Steve Rust; Alan Sandercock; Ross Stewart; Viia Valge-Archer; Robert W. Wilkinson

Regulatory T cells (Treg cells) are observed within multiple tumor types and contribute to immune evasion. Effective and specific targeting of Treg cells by antibodies is hampered by the lack of a unique surface marker. With the aim of identifying such a marker, human Treg cells were isolated from peripheral blood, expanded in vitro and used as a source of antigen in phage display selections with a diverse library of designed ankyrin repeat proteins (DARPins). The resulting DARPins were screened for binding to multiple cell-types, and approximately thirty with preferential binding to Treg cells were identified. All of these DARPins were determined to bind to the third and/or fourth cysteine-rich extracellular domains of tumor necrosis factor receptor II (TNFRII, also known as TNFRSF1B or CD120b). Expression of TNFRII has previously been demonstrated on Treg cells, and TNFRII is shown here also to be expressed on CD8+ T cells following exposure to cognate antigen and on the surface of tumor-infiltrating T cells in the 4T1 mouse tumor model. Furthermore, TNFRII mRNA was detected at increased levels in tumor-infiltrating leukocytes isolated from cancer patients. Functional characterisation of the anti-TNFRII DARPins and mAbs revealed an ability to enhance T-cell activation in response to polyclonal and antigen specific stimuli, and in Treg cell suppression assays. These studies demonstrate the utility of DARPins for phenotypic selections to identify drug targets using primary human lymphocytes and highlight TNFRII as a potential therapeutic target enriched on Treg cells. Citation Format: Geoff Williams, Judith Anderton, Vahe Bedian, Jane Coates Ulrichsen, Andrea Gonzalez-Munoz, Sandrine Guillard, Olivia Harris, James Hair, Andrew Leishman, Arthur Lewis, Jacques Moissan, Ralph Minter, Bina Mistry, Julie Parmentier, Edmund Poon, Amy Popple, Steve Rust, Alan Sandercock, Ross Stewart, Viia Valge-Archer, Robert W. Wilkinson. Identification of tumor necrosis factor receptor II as a regulatory T cell target for cancer immunotherapy using designed ankyrin repeat protein phenotypic selections. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4613. doi:10.1158/1538-7445.AM2014-4613


Cancer Research | 2013

Abstract 4328: Phenotypic selection for identification of functional antibodies and development of high throughput screening assays.

Carl Hay; Steven Rust; Erin Sult; Lori Clarke; Kim Rosenthal; Sandrine Guillard; David Lowne; Matt Flynn; Lutz Jermutus; Ralph Minter; Robert E. Hollingsworth; Kris Sachsenmeier

Discovery of therapeutic antibodies requires the continued identification of tractable targets, and we have employed a phenotypic screening strategy to this end. In this study, antibodies from phage display libraries were screened for their abilities to inhibit MDA-MB-231 (‘triple negative’) breast cancer cells. The screening cascade included tests for specific cell binding, antibody internalization, and cytotoxicity as an antibody-drug conjugate. Targets of these antibodies then were identified using immunoprecipitation and mass spectrometry, and confirmed by showing diminished antibody binding after siRNA-mediated knockdown of the putative target gene. One target was identified as NT5E (also known as CD73), a 5’-ectonucleotidase. NT5E catalyzes the conversion of adenosine monophosphate to adenosine, and its function has been linked with immunity, angiogenesis, and cancer. To screen for additional inhibitors of this target, a novel, high-throughput ectonucleotidase assay was developed. NT5E activity is typically assayed either by high performance liquid chromatography or by quantification of free phosphate using malachite green, and neither of these methods is suitable for high-throughput screening. One inhibitory antibody was found to significantly slow growth of MDA-MB-231 xenograft tumors in mice. In summary, our phenotypic screening approach provides a mechanism for rapidly discovering and evaluating new antibody targets and is being used to accelerate the discovery of new cancer drugs. Citation Format: Carl Hay, Steven Rust, Erin Sult, Lori Clarke, Kim Rosenthal, Sandrine Guillard, David Lowne, Matt Flynn, Lutz Jermutus, Ralph Minter, Robert Hollingsworth, Kris Sachsenmeier. Phenotypic selection for identification of functional antibodies and development of high throughput screening assays. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4328. doi:10.1158/1538-7445.AM2013-4328


Cancer Research | 2012

Abstract 974: New target discovery for oncology utilizing phenotypic selection and primary NSCLC patient tumors

Nicholas Holoweckyj; Alan Sandercock; Sandrine Guillard; Steven Rust; Carl Hay; Erin Brand; John Meekin; Qihui Huang; Kris Sachsenmeier; Robert E. Hollingsworth; Ralph Minter; Emma T. Bowden

Lung cancer is the most common type of cancer, with approximately 222,000 new cases expected each year in the United States. Approximately 72% of the individuals diagnosed with lung cancer will fall victim to the disease. Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, but current treatments will not cure the disease for most patients. In an effort to identify novel therapeutics to address this unmet need, a phenotypic screening campaign was initiated. This type of target identification approach allows the discovery of novel drug targets by selection of antibodies based on their ability to elicit a specific desired phenotype. Once antibodies with the desired function are isolated, the targets to which the “hit” antibodies bind can be identified using a variety of approaches including IP-mass spec. Although phenotypic screens have been performed previously, the current effort has important advantages over those described previously. Specifically, this included the use of several unique screening assays run in parallel to allow the evaluation of multiple phenotypes simultaneously. Also unique was the use of primary material from NSCLC patients for both selection and functional screening. Specifically, we demonstrate the use of primary tumor cells in high throughput screening assays. We show the utility of incorporating novel screening assays including anoikis and spheroids with multiple endpoint readouts. Finally we provide data describing a series of “hit” antibodies isolated in the screen which had a variety of activities across the screening assays. These studies show the utility of function-first screening in the identification of unique potential therapeutic targets for oncology. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 974. doi:1538-7445.AM2012-974

Collaboration


Dive into the Sandrine Guillard's collaboration.

Researchain Logo
Decentralizing Knowledge