Network


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

Hotspot


Dive into the research topics where Marie-Alix Poul is active.

Publication


Featured researches published by Marie-Alix Poul.


Journal of Immunological Methods | 2001

Expression of single-chain Fv-Fc fusions in Pichia pastoris

David B. Powers; Peter Amersdorfer; Marie-Alix Poul; Ulrik Nielsen; M.Refaat Shalaby; Gregory P. Adams; Louis M. Weiner; James D. Marks

Phage display technology makes possible the direct isolation of monovalent single-chain Fv antibody fragments. For many applications, however, it is useful to restore Fc mediated antibody functions such as avidity, effector functions and a prolonged serum half-life. We have constructed vectors for the convenient, rapid expression of a single-chain antibody Fv domain (scFv) fused to the Fc portion of human IgG1 in the methylotrophic yeast Pichia pastoris. The scFv-Fc fusion protein is secreted and recovered from the culture medium as a disulfide-linked, glycosylated homodimer. The increased size of the dimer (approximately 106 kDa vs. approximately 25 kDa for a scFv) results in a prolonged serum half-life in vivo, with t(1/2) of the beta phase of clearance increasing from 3.5 h for a typical scFv to 93 h for a scFv-Fc fusion in mice. The scFv-Fc fusion is capable of mediating antibody-dependent cellular cytotoxicity against tumor target cells using human peripheral blood mononuclear cells as effectors. Finally, the Fc domain is a convenient, robust affinity handle for purification and immunochemical applications, eliminating the need for proteolytically sensitive epitope and/or affinity tags on the scFv.


Molecular Cancer Research | 2009

The Aberrant Localization of Oncogenic Kit Tyrosine Kinase Receptor Mutants Is Reversed on Specific Inhibitory Treatment

Houcine Bougherara; Frédéric Subra; Ronan Crépin; Patrick Tauc; Christian Auclair; Marie-Alix Poul

Kit is a cell surface type III tyrosine kinase (TK) receptor implicated in cell transformation through overexpression or oncogenic mutation. Two categories of Kit mutants displaying mutations either in the juxtamembrane intracellular domain (regulatory mutants) or in the catalytic domain (catalytic mutants) have been described. To explore the effect of Kit oncogenic mutations on its subcellular localization, we constructed enhanced green fluorescent protein (EGFP)–tagged human Kit chimeras harboring mutations either in the regulatory (V560G) or in the catalytic (D816V) domain. When expressed in Chinese hamster ovary cells, EGFP-tagged wild-type Kit was activated on stem cell factor stimulation, whereas both EGFP-tagged Kit mutants displayed a constitutive TK activity. Constitutively activated mutants exhibited a high-mannose–type N-glycosylation pattern and an intracellular localization, suggesting that these mutants induce downstream oncogenic signaling without the need to reach the cell surface. Inhibition of constitutive Kit TK activity with dasatinib induced a complex, mature N-glycosylation pattern identical to unstimulated wild-type Kit and resulted in the redistribution of the mutants to the plasma membrane. This relocalization was clearly correlated to the inhibition of TK activity because imatinib, a specific inhibitor of the V560G mutant, inactive on the catalytic D816V mutant, induced only the relocalization of the V560G mutant. These data show that on TK inhibition, the aberrant localization of Kit mutants can be fully reversed. Kit mutants are then exported and/or stabilized at the cell surface as inactive and fully N-glycosylated isoforms. (Mol Cancer Res 2009;7(9):1525–33)


Journal of Biological Chemistry | 2011

Time-resolved Fluorescence Resonance Energy Transfer (TR-FRET) to Analyze the Disruption of EGFR/HER2 Dimers A NEW METHOD TO EVALUATE THE EFFICIENCY OF TARGETED THERAPY USING MONOCLONAL ANTIBODIES

Nadège Gaborit; Christel Larbouret; Julie Vallaghe; Frédéric Peyrusson; Caroline Bascoul-Mollevi; Evelyne Crapez; D. Azria; Thierry Chardès; Marie-Alix Poul; Hervé Bazin; André Pèlegrin

In oncology, simultaneous inhibition of epidermal growth factor receptor (EGFR) and HER2 by monoclonal antibodies (mAbs) is an efficient therapeutic strategy but the underlying mechanisms are not fully understood. Here, we describe a time-resolved fluorescence resonance energy transfer (TR-FRET) method to quantify EGFR/HER2 heterodimers on cell surface to shed some light on the mechanism of such therapies. First, we tested this antibody-based TR-FRET assay in NIH/3T3 cell lines that express EGFR and/or HER2 and in various tumor cell lines. Then, we used the antibody-based TR-FRET assay to evaluate in vitro the effect of different targeted therapies on EGFR/HER2 heterodimers in the ovarian carcinoma cell line SKOV-3. A simultaneous incubation with Cetuximab (anti-EGFR) and Trastuzumab (anti-HER2) disturbed EGFR/HER2 heterodimers resulting in a 72% reduction. Cetuximab, Trastuzumab or Pertuzumab (anti-HER2) alone induced a 48, 44, or 24% reduction, respectively. In contrast, the tyrosine kinase inhibitors Erlotinib and Lapatinib had very little effect on EGFR/HER2 dimers concentration. In vivo, the combination of Cetuximab and Trastuzumab showed a better therapeutic effect (median survival and percentage of tumor-free mice) than the single mAbs. These results suggest a correlation between the extent of the mAb-induced EGFR/HER2 heterodimer reduction and the efficacy of such mAbs in targeted therapies. In conclusion, quantifying EGFR/HER2 heterodimers using our antibody-based TR-FRET assay may represent a useful method to predict the efficacy and explain the mechanisms of action of therapeutic mAbs, in addition to other commonly used techniques that focus on antibody-dependent cellular cytotoxicity, phosphorylation, and cell proliferation.


Cancer Research | 2010

Development of human single-chain antibodies to the transferrin receptor that effectively antagonize the growth of leukemias and lymphomas.

Ronan Crépin; Anne-Laure Goenaga; Betsy Jullienne; Houcine Bougherara; Christine Legay; Karim Benihoud; James D. Marks; Marie-Alix Poul

The major route of iron uptake by cells occurs through transferrin receptor (TfR)-mediated endocytosis of diferric-charged plasma transferrin (holo-Tf). In this work, we pursued TfR antibodies as potential cancer therapeutics, characterizing human single-chain variable antibody fragments (scFv) specific for the human TfR isolated from a phage display library. We hypothesized that many of these antibodies would function as ligand mimetics because scFvs from the library were selected for binding and internalization into living cells. In support of this hypothesis, the anti-TfR scFvs identified were antagonists of TfR binding to holo-Tf, particularly two of the most potent antibodies, 3TF12 and 3GH7, which blocked the in vitro proliferation of a number of hematopoietic cancer cell lines. We optimized this activity of 3TF12 and 3GH7 by engineering 55-kDa bivalent antibody formats, namely, F12CH and H7CH, which could block cell proliferation with an IC(50) of 0.1 microg/mL. We found that the mechanism of the scFv antibody cytotoxicity was unique compared with cytotoxic anti-TfR monoclonal antibodies that have been described, causing cell surface upregulation of TfR along with the inhibition of holo-Tf cell uptake and induction of cell death. In a nude mouse model of erythroleukemia, administration of F12CH reduced tumor growth. Together, our findings define a new class of fully human anti-TfR antibodies suitable for immunotherapy against tumors whose proliferation relies on high levels of TfR and iron uptake, such as acute lymphoid and myeloid leukemias.


Immunotechnology | 1995

Design of cassette baculovirus vectors for the production of therapeutic antibodies in insect cells

Marie-Alix Poul; Martine Cerutti; Hassan Chaabihi; Gérard Devauchelle; Michel Kaczorek; Marie-Paule Lefranc

BACKGROUND Various systems have been described for the expression of recombinant monoclonal antibodies for therapeutical applications. Insect cells offer great advantages with respect to post-translational modifications, stability, yields and applications. OBJECTIVES To construct plasmid cassette transfer vectors in order to express chimeric, humanized or human antibodies in insect cells using baculovirus expression system. STUDY DESIGN Two transfer vectors, pBHuC kappa and pBHuC gamma 1, were designed. They contain a viral promoter (polyhedrin or p10 promoters, respectively), a signal peptide sequence and a human immunoglobulin light chain C kappa gene or heavy chain C gamma 1 sequence, respectively. Restriction sites have been introduced to allow insertion of rearranged variable genes, after amplification by polymerase chain reaction. RESULTS Recombinant baculoviruses expressing complete immunoglobulins have been generated by a double-recombination event between baculovirus DNA and the loaded cassette transfer vectors. CONCLUSION Our genetic cassette approach makes this system a very flexible and convenient one for the rapid production of therapeutic monoclonal antibodies with heavy and light chains of any human isotype. Specific variable regions selected by the antibody phage display technology can be easily transferred in these vectors to obtain a complete antibody.


Molecular Immunology | 1995

Inhibition of T cell activation with a humanized anti-β1 integrin chain mAb

Marie-Alix Poul; Michel Ticchioni; Alain Bernard; Marie-Paule Lefranc

The murine anti-CD29 mAb K20 (Mu-K20) is known to bind to the β1 chain of the human integrins and to inhibit activation and proliferation of T cells, implying an important potential for in vivo immunosuppression. However, use of K20 as an immunosuppressant drug would be impaired by the immunogenicity of mouse mAbs in man. We have therefore engineered K20 into (1) a mouse/human chimeric mAb (Ch-K20) that comprises the human κγ1 C regions and the K20 V regions; and (2) a humanized mAb (Hu-K20) combining the complementarity-determining regions (CDRs) of the K20 mAb with human framework (FR) and κγ1 C regions. Both chimeric and humanized Abs were able to reproduce a range of functional properties of the original mouse mAb K20 (Mu-K20), namely, specific binding of CD29, inhibition of T cell proliferation and elevation of second messenger phosphatidic acid (PA) induced via CD3 in a soluble form, and activation of T cell proliferation in a cross-linked form. When compared to Ch-K20, the avidity of Hu-K20 was only slightly reduced. This demonstrates the feasibility of a successful humanization performed on the sole basis of the primary amino acid sequence analysis of the original mouse antibody V regions.


Molecular Cancer Therapeutics | 2017

Neuregulin 1 allosterically enhances the anti-tumor effects of the non-competing anti-HER3 antibody 9F7-F11 by increasing its binding to HER3

Christophe Le Clorennec; Hervé Bazin; Olivier Dubreuil; Christel Larbouret; Charline Ogier; Yassamine Lazrek; Véronique Garambois; Marie-Alix Poul; Philippe Mondon; Jean-Marc Barret; Jean-François Prost; André Pèlegrin; Thierry Chardès

Exploratory clinical trials using therapeutic anti-HER3 antibodies strongly suggest that neuregulin (NRG1; HER3 ligand) expression at tumor sites is a predictive biomarker of anti-HER3 antibody efficacy in cancer. We hypothesized that in NRG1-expressing tumors, where the ligand is present before antibody treatment, anti-HER3 antibodies that do not compete with NRG1 for receptor binding have a higher receptor-neutralizing action than antibodies competing with the ligand for binding to HER3. Using time-resolved–fluorescence energy transfer (TR-FRET), we demonstrated that in the presence of recombinant NRG1, binding of 9F7-F11 (a nonligand-competing anti-HER3 antibody) to HER3 is increased, whereas that of ligand-competing anti-HER3 antibodies (H4B-121, U3-1287, Ab#6, Mab205.10.2, and MOR09825) is decreased. Moreover, 9F7-F11 showed higher efficacy than antibodies that compete with the ligand for binding to HER3. Specifically, 9F7-F11 inhibition of cell proliferation and of HER3/AKT/ERK1/2 phosphorylation as well as 9F7-F11–dependent cell-mediated cytotoxicity were higher in cancer cells preincubated with recombinant NRG1 compared with cells directly exposed to the anti-HER3 antibody. This translated in vivo into enhanced growth inhibition of NRG1-expressing BxPC3 pancreatic, A549 lung, and HCC-1806 breast cell tumor xenografts in mice treated with 9F7-F11 compared with H4B-121. Conversely, both antibodies had similar antitumor effect in NRG1-negative HPAC pancreatic carcinoma cells. In conclusion, the allosteric modulator 9F7-F11 shows increased anticancer effectiveness in the presence of NRG1 and thus represents a novel treatment strategy for NRG1-addicted tumors. Mol Cancer Ther; 16(7); 1312–23. ©2017 AACR.


Archive | 1995

Production of a Biologically Active Chimeric Mouse/Human Monoclonal Antibody in Insect Cells

F. X. Deramoudt; H Chaabihi; Marie-Alix Poul; C. Margaritte; Martine Cerutti; Gérard Devauchelle; Alain Bernard; Marie-Paule Lefranc; M. Kaczorek.

Murine K20 (m-K20) monoclonal antibody (MAb) has an immunosuppressive effect by inhibiting human T-cell proliferation after binding to the s1-subunit of human integrins (CD29). In order to express a chimeric mouse/human form of K20 with an insect cells/baculovirus system, sequences encoding the variable regions of mu-K20 have been cloned and inserted into plasmid cassettes. Each cassette contains a baculovirus strong late promoter, a sequence encoding an immunoglobulin signal peptide and a sequence encoding a human constant region (respectively κ and γl). Subsequent transfer by homologous recombination into the baculovirus genome was performed to construct a recombinant virus for the co-expression of the light and heavy chain sequences of the MAb. Insect cells infected with this virus co-produce both chimeric K20 (Chi-K20) light (L) and heavy (H) chains which are assembled and secreted into the culture medium. ch-K20 was produced in a 10 liter stirred bioreactor, and purified by affinity chromatography. In vitro comparative assays showed that Chi-K20 and parental mu -K20 are indistinguishable by means of binding to CD29, cell specific targeting and inhibition of T-cell proliferation.


Molecular Cancer Therapeutics | 2015

Neutralization of KIT oncogenic signaling in leukemia with antibodies targeting KIT membrane proximal domain 5

Marianne Le Gall; Ronan Crépin; Madeline Neiveyans; Christian Auclair; Yongfeng Fan; Yu Zhou; James D. Marks; André Pèlegrin; Marie-Alix Poul

KIT is a cell surface tyrosine kinase receptor whose ligand stem cell factor (SCF) triggers homodimerization and activation of downstream effector pathways involved in cell survival, proliferation, homing, or differentiation. KIT-activating mutations are major oncogenic drivers in subsets of acute myeloid leukemia (AML), in mast cell leukemia, and in gastrointestinal stromal tumors (GIST). The overexpression of SCF and/or wild-type (WT) KIT is also observed in a number of cancers, including 50% of AML and small cell lung cancer. The use of tyrosine kinase inhibitors (TKI) in these pathologies is, however, hampered by initial or acquired resistance following treatment. Using antibody phage display, we obtained two antibodies (2D1 and 3G1) specific for the most membrane proximal extracellular immunoglobulin domain (D5) of KIT, which is implicated in KIT homodimerization. Produced as single chain variable antibody fragments fused to the Fc fragment of a human IgG1, bivalent 2D1-Fc and 3G1-Fc inhibited KIT-dependent growth of leukemic cell lines expressing WT KIT (UT7/Epo) or constitutively active KIT mutants, including the TKI imatinib-resistant KIT D816V mutant (HMC1.2 cell line). In all models, either expressing WT KIT or mutated KIT, 2D1 and 3G1-Fc induced KIT internalization and sustained surface downregulation. However, interestingly, KIT degradation was only observed in leukemic cell lines with oncogenic KIT, a property likely to limit the toxicity of these antibodies in patients. These fully human antibody formats may represent therapeutic tools to target KIT signaling in leukemia or GIST, and to bypass TKI resistance of certain KIT mutants. Mol Cancer Ther; 14(11); 2595–605. ©2015 AACR.


Journal of Molecular Biology | 2000

Selection of tumor-specific internalizing human antibodies from phage libraries

Marie-Alix Poul; Baltazar Becerril; Ulrik Nielsen; Peter Morisson; James D. Marks

Collaboration


Dive into the Marie-Alix Poul's collaboration.

Top Co-Authors

Avatar

James D. Marks

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ulrik Nielsen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gérard Devauchelle

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Martine Cerutti

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Thierry Chardès

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Christian Auclair

École normale supérieure de Cachan

View shared research outputs
Top Co-Authors

Avatar

Houcine Bougherara

École normale supérieure de Cachan

View shared research outputs
Top Co-Authors

Avatar

Ronan Crépin

École normale supérieure de Cachan

View shared research outputs
Researchain Logo
Decentralizing Knowledge