59 results on '"Christel Larbouret"'
Search Results
2. Design and selection of optimal ErbB-targeting bispecific antibodies in pancreatic cancer
- Author
-
Emilia Rabia, Véronique Garambois, Christine Dhommée, Christel Larbouret, Laurie Lajoie, Yoan Buscail, Gabriel Jimenez-Dominguez, Sylvie Choblet-Thery, Emmanuelle Liaudet-Coopman, Martine Cerutti, Marta Jarlier, Patrice Ravel, Laurent Gros, Nelly Pirot, Gilles Thibault, Eugene A. Zhukovsky, Pierre-Emmanuel Gérard, André Pèlegrin, Jacques Colinge, and Thierry Chardès
- Subjects
ErbB ,systems biology ,antibody ,bispecific ,pancreatic cancer ,ADCC ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The ErbB family of receptor tyrosine kinases is a primary target for small molecules and antibodies for pancreatic cancer treatment. Nonetheless, the current treatments for this tumor are not optimal due to lack of efficacy, resistance, or toxicity. Here, using the novel BiXAb™ tetravalent format platform, we generated bispecific antibodies against EGFR, HER2, or HER3 by considering rational epitope combinations. We then screened these bispecific antibodies and compared them with the parental single antibodies and antibody pair combinations. The screen readouts included measuring binding to the cognate receptors (mono and bispecificity), intracellular phosphorylation signaling, cell proliferation, apoptosis and receptor expression, and also immune system engagement assays (antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity). Among the 30 BiXAbs™ tested, we selected 3Patri-1Cetu-Fc, 3Patri-1Matu-Fc and 3Patri-2Trastu-Fc as lead candidates. The in vivo testing of these three highly efficient bispecific antibodies against EGFR and HER2 or HER3 in pre-clinical mouse models of pancreatic cancer showed deep antibody penetration in these dense tumors and robust tumor growth reduction. Application of such semi-rational/semi-empirical approach, which includes various immunological assays to compare pre-selected antibodies and their combinations with bispecific antibodies, represents the first attempt to identify potent bispecific antibodies against ErbB family members in pancreatic cancer.
- Published
- 2023
- Full Text
- View/download PDF
3. ITCH-dependent proteasomal degradation of c-FLIP induced by the anti-HER3 antibody 9F7-F11 promotes DR5/caspase 8-mediated apoptosis of tumor cells
- Author
-
Christophe Le Clorennec, Yassamine Lazrek, Olivier Dubreuil, Carla Sampaio, Christel Larbouret, Romain Lanotte, Marie-Alix Poul, Jean-Marc Barret, Jean-François Prost, André Pèlegrin, and Thierry Chardès
- Subjects
Cancer ,HER3 ,Antibody ,Apoptosis ,C-FLIP ,ITCH ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background HER3/ErbB3 receptor deletion or blockade leads to tumor cell apoptosis, whereas its overexpression confers anti-cancer drug resistance through upregulation of protective mechanisms against apoptosis. We produced the anti-HER3 antibody 9F7-F11 that promotes HER3 ubiquitination and degradation via JNK1/2-dependent activation of the E3 ubiquitin ligase ITCH, and that induces apoptosis of cancer cells. Cellular FLICE-like inhibitory protein (c-FLIP) is a key regulator of apoptotic pathways. Here, we wanted to determine the mechanisms underlying the pro-apoptotic effect of 9F7-F11. Methods Anti-HER3 antibody-induced apoptosis was assessed by western blot, and by flow cytometry measurement of Annexin V/7-AAD-labelled tumor cells (BxPC3, MDA-MB-468 and DU145 cell lines). c-FLIP/ITCH interaction and subsequent degradation/ubiquitination were investigated by co-immunoprecipitation of ITCH-silenced vs scramble control cells. The relationship between ITCH-mediated c-FLIP degradation and antibody-induced apoptosis was examined by western blot and flow cytometry of tumor cells, after ITCH RNA interference or by pre-treatment with ITCH chemical inhibitor chlorimipramine (CI). Results Following incubation with 9F7-F11, cancer cell apoptosis occurs through activation of caspase-8, − 9 and − 3 and the subsequent cleavage of poly (ADP-ribose) polymerase (PARP). Moreover we showed that ubiquitination and proteasomal degradation of the anti-apoptotic protein c-FLIP was mediated by USP8-regulated ITCH recruitment. This effect was abrogated by ITCH- and USP8-specific RNA interference (siRNA), or by the ITCH chemical inhibitor CI. Specifically, ITCH silencing or CI blocked 9F7-F11-induced caspase-8-mediated apoptosis of tumor cells, and restored c-FLIP expression. ITCH-silencing or CI concomitantly abrogated HER3-specific antibody-induced apoptosis of Annexin V/7-AAD-labelled BxPC3 cells. 9F7-F11 favored the extrinsic apoptosis pathway by inducing TRAIL-R2/DR5 upregulation and TRAIL expression that promoted the formation of death-inducing signaling complex (DISC), leading to caspase-8-mediated apoptosis. Incubation with 9F7-F11 also induced BID cleavage, BAX upregulation and BIM expression, which initiated the caspase-9/3-mediated mitochondrial death pathway. The anti-HER3 antibody pro-apoptotic effect occurred concomitantly with downregulation of the pro-survival proteins c-IAP2 and XIAP. Conclusions The allosteric non-neuregulin competing modulator 9F7-F11, sensitizes tumor cells to DR5/caspase-8-mediated apoptosis through ITCH-dependent downregulation of c-FLIP. Graphical abstract
- Published
- 2019
- Full Text
- View/download PDF
4. Anti-tumoral activity of the Pan-HER (Sym013) antibody mixture in gemcitabine-resistant pancreatic cancer models
- Author
-
Emilia Rabia, Véronique Garambois, Julie Hubert, Marine Bruciamacchie, Nelly Pirot, Hélène Delpech, Morgane Broyon, Charles Theillet, Pierre-Emmanuel Colombo, Nadia Vie, Diego Tosi, Celine Gongora, Lakhdar Khellaf, Marta Jarlier, Nina Radosevic-Robin, Thierry Chardès, André Pèlegrin, and Christel Larbouret
- Subjects
EGFR ,HER2 ,HER3 ,pancreatic cancer ,gemcitabine ,Pan-Her ,Therapeutics. Pharmacology ,RM1-950 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Chemoresistance, particularly to gemcitabine, is a major challenge in pancreatic cancer. The epidermal growth factor receptor (EGFR) and human epidermal growth factor receptors 2 and 3 (HER2, HER3) are expressed in many tumors, and they are relevant therapeutic targets due to their synergistic interaction to promote tumor aggressiveness and therapeutic resistance. Cocktails of antibodies directed against different targets are a promising strategy to overcome these processes. Here, we found by immunohistochemistry that these three receptors were co-expressed in 11% of patients with pancreatic adenocarcinoma. We then developed gemcitabine-resistant pancreatic cancer cell models (SW-1990-GR and BxPC3-GR) and one patient-derived xenograft (PDX2846-GR) by successive exposure to increasing doses of gemcitabine. We showed that expression of EGFR, HER2 and HER3 was increased in these gemcitabine-resistant pancreatic cancer models, and that an antibody mixture against all three receptors inhibited tumor growth in mice and downregulated HER receptors. Finally, we demonstrated that the Pan-HER and gemcitabine combination has an additive effect in vitro and in mice xenografted with the gemcitabine-sensitive or resistant pancreatic models. The mixture of anti-EGFR, HER2 and HER3 antibodies is a good candidate therapeutic approach for gemcitabine-sensitive and -resistant pancreatic cancer.
- Published
- 2021
- Full Text
- View/download PDF
5. Rational development of synergistic combinations of chemotherapy and molecular targeted agents for colorectal cancer treatment
- Author
-
Diego Tosi, Esther Pérez-Gracia, Salima Atis, Nadia Vié, Eve Combès, Mélissa Gabanou, Christel Larbouret, Marta Jarlier, Caroline Mollevi, Adeline Torro, Maguy Del Rio, Pierre Martineau, and Céline Gongora
- Subjects
Colorectal cancer ,Drug combinations ,Phosphokinome ,Irinotecan ,Synergistic effect ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The irinotecan-induced phosphokinome changes in colorectal cancer (CRC) cells were used to guide the selection of targeted agents to be tested in combination with irinotecan. Methods Phosphokinome profiling with peptide arrays of tumour samples from nude mice xenografted with HT29 cells and treated or not with an effective dose of irinotecan was used to identify signalling pathways activated by irinotecan treatment. Then, drugs targeting these pathways were combined in vitro with irinotecan to test potential synergistic effect. The interactions between these drug combinations were assessed by a dose matrix approach. Confirmation of the most potential combination has been confirmed in vivo in xenografted mice. Results Irinotecan induced in vivo the activation of AKT and MEK1 phosphorylation. The dose matrix approach showed that BKM120 (PI3K inhibitor) and MEK162 (MEK inhibitor) are synergistic in vitro and in vivo with a cytostatic and cytotoxic effect, while combination of BKM120 and irinotecan or MEK162 and irinotecan are only additive or even antagonistic. However, the triple combination of SN38, BKM120 and MEK162 showed a better synergistic effect that BKM120 and MEK162, indicating that the cells need to inhibit both AKT and ERK pathways to become more sensitive to irinotecan-based chemotherapies. Conclusion Analysis of chemotherapy-induced phosphokinome changes helps to elucidate the mechanisms of drug resistance and to guide the selection of targets for combination therapies with synergistic activity.
- Published
- 2018
- Full Text
- View/download PDF
6. Anti-HER3 Domain 1 and 3 Antibodies Reduce Tumor Growth by Hindering HER2/HER3 Dimerization and AKT-Induced MDM2, XIAP, and FoxO1 Phosphorylation
- Author
-
Yassamine Lazrek, Olivier Dubreuil, Véronique Garambois, Nadège Gaborit, Christel Larbouret, Christophe Le Clorennec, Gaelle Thomas, Wilhem Leconet, Marta Jarlier, Martine Pugnière, Nadia Vié, Bruno Robert, Céline Monnet, Khalil Bouayadi, Hakim Kharrat, Philippe Mondon, André Pèlegrin, and Thierry Chardès
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Blockade of the human epidermal growth factor receptor 3 (HER3) and of the downstream phosphatidylinositide 3-kinase (PI3K)/AKT pathway is a prerequisite for overcoming drug resistance and to develop novel treatments for cancers that are not eligible for the currently approved targeted therapies. To this end, we generated specific antibodies (Abs) against domain 1 (D1) and domain 3 (D3) of HER3 that recognize epitopes that do not overlap with the neuregulin-binding site. The fully human H4B-121 Ab and the mouse monoclonal Abs 16D3-C1 and 9F7-F11 inhibited tumor growth in nude mice xenografted with epidermoid, pancreatic, or triple-negative breast cancer cells. The combination of one anti-HER3 Ab and trastuzumab improved tumor growth inhibition in mice xenografted with HER2low cancer cell lines, for which trastuzumab alone shows no or moderate efficiency. Ab-induced disruption of tumor growth was associated with G1 cell cycle arrest, proliferation inhibition, and apoptosis of cancer cells. Anti-HER3 Abs blocked HER2/HER3 heterodimerization and HER3 phosphorylation at the cell membrane, leading to inhibition of phosphorylation of the downstream AKT targets murine double minute 2, X-linked inhibitor of apoptosis, and forkhead box O1. This study demonstrates that anti-HER3 D1 and D3 Abs could represent a new option for immunotherapy of pancreatic and triple-negative breast cancers.
- Published
- 2013
- Full Text
- View/download PDF
7. In Pancreatic Carcinoma, Dual EGFR/HER2 Targeting with Cetuximab/Trastuzumab Is More Effective than Treatment with Trastuzumab/Erlotinib or Lapatinib Alone: Implication of Receptors' Down-regulation and Dimers' Disruption
- Author
-
Christel Larbouret, Nadège Gaborit, Thierry Chardès, Mickaël Coelho, Emmanuelle Campigna, Caroline Bascoul-Mollevi, Jean-Pierre Mach, David Azria, Bruno Robert, and André Pèlegrin
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
We previously demonstrated the synergistic therapeutic effect of the cetuximab (anti-epidermal growth factor receptor [EGFR] monoclonal antibody, mAb)-trastuzumab (anti-HER2 mAb) combination (2mAbs therapy) in HER2low human pancreatic carcinoma xenografts. Here, we compared the 2mAbs therapy, the erlotinib (EGFR tyrosine kinase inhibitor [TKI])-trastuzumab combination and lapatinib alone (dual HER2/EGFR TKI) and explored their possible mechanisms of action. The effects on tumor growth and animal survival of the three therapies were assessed in nude mice xenografted with the human pancreatic carcinoma cell lines Capan-1 and BxPC-3. After therapy, EGFR and HER2 expression and AKT phosphorylation in tumor cells were analyzed by Western blot analysis. EGFR/HER2 heterodimerization was quantified in BxPC-3 cells by time-resolved FRET. In K-ras-mutated Capan-1 xenografts, the 2mAbs therapy gave significantly higher inhibition of tumor growth than the erlotinib/trastuzumab combination, whereas in BxPC-3 (wild-type K-ras) xenografts, the erlotinib/trastuzumab combination showed similar growth inhibition but fewer tumor-free mice. Lapatinib showed no antitumor effect in both types of xenografts. The efficacy of the 2mAbs therapy was partly Fc-independent because F(ab′)2 fragments of the two mAbs significantly inhibited BxPC-3 growth, although with a time-limited therapeutic effect. The 2mAbs therapy was associated with a reduction of EGFR and HER2 expression and AKT phosphorylation. BxPC-3 cells preincubated with the two mAbs showed 50% less EGFR/HER2 heterodimers than controls. In pancreatic carcinoma xenografts, the 2mAbs therapy is more effective than treatments involving dual EGFR/HER2 TKIs. The mechanism of action may involve decreased AKT phosphorylation and/or disruption of EGFR/HER2 heterodimerization.
- Published
- 2012
- Full Text
- View/download PDF
8. Quantification of HER expression and dimerization in patients' tumor samples using time-resolved Förster resonance energy transfer.
- Author
-
Alexandre Ho-Pun-Cheung, Hervé Bazin, Nadège Gaborit, Christel Larbouret, Patrick Garnero, Eric Assenat, Florence Castan, Caroline Bascoul-Mollevi, Jeanne Ramos, Marc Ychou, André Pèlegrin, Gérard Mathis, and Evelyne Lopez-Crapez
- Subjects
Medicine ,Science - Abstract
Following the development of targeted therapies against EGFR and HER2, two members of the human epidermal receptor (HER) family of receptor tyrosine kinases, much interest has been focused on their expression in tumors. However, knowing the expression levels of individual receptors may not be sufficient to predict drug response. Here, we describe the development of antibody-based time-resolved Förster resonance energy transfer (TR-FRET) assays for the comprehensive analysis not only of EGFR and HER2 expression in tumor cryosections, but also of their activation through quantification of HER homo- or heterodimers. First, EGFR and HER2 expression levels were quantified in 18 breast tumors and the results were compared with those obtained by using reference methods. The EGFR number per cell determined by TR-FRET was significantly correlated with EGFR mRNA copy number (P
- Published
- 2012
- Full Text
- View/download PDF
9. Figure S3 from Neuregulin 1 Allosterically Enhances the Antitumor Effects of the Noncompeting Anti-HER3 Antibody 9F7-F11 by Increasing Its Binding to HER3
- Author
-
Thierry Chardès, André Pèlegrin, Jean-François Prost, Gérard Mathis, Jean-Marc Barret, Philippe Mondon, Marie-Alix Poul, Véronique Garambois, Yassamine Lazrek, Charline Ogier, Christel Larbouret, Olivier Dubreuil, Hervé Bazin, and Christophe Le Clorennec
- Abstract
Supplemental Fig.S3. In pancreatic cancer cells, ch9F7-F11-Emb is a negative allosteric modulator of NRG1-mediated signaling. The low-fucose non-NRG1 competing allosteric anti-HER3 antibody ch9F7-F11-Emb inhibits in a dose-dependent manner NRG1-mediated cell signaling more efficiently than the ligand-competing H4B-121-Emb antibody. BxPC3 cells were pre-stimulated with 1nM NRG1 for 5min, before adding antibodies at various concentrations for another 25min. After cell lysis, the expression levels of total and phosphorylated HER3 (Tyr1289, Tyr1197 and Tyr1222), and total and phosphorylated AKT (Ser473) were measured by western blotting (A) using the appropriate antibodies. Phosphorylation was then pixel-quantified with Image J (B) and shown relative to the maximal phosphorylation (100%) evaluated in NRG1-stimulated cells without antibody treatment (M). Results are the mean {plus minus} SD of three independent experiments
- Published
- 2023
10. Supplementary Figure S1-S6 from Therapeutic Activity of Anti-AXL Antibody against Triple-Negative Breast Cancer Patient-Derived Xenografts and Metastasis
- Author
-
Bruno Robert, Christel Larbouret, André Pèlegrin, Jean-Max Pasquet, Dany Chalbos, Charles Theillet, Nina Radosevic-Robin, Marta Jarlier, Muriel Busson, Imade Aït-Arsa, Audrey Sirvent, Clément Chevalier, Stanislas du Manoir, Myriam Chentouf, and Wilhem Leconet
- Abstract
Supplementary Figure 1. AXL, MER and TYRO-3 expression in Triple Negative Breast Cancer (TNBC) cell lines and PDXs by Western blot analysis. Supplementary Figure 2. AXL mRNA expression in a large dataset of breast cancer tumors. Supplementary Figure 3. The 100 genes most correlated with AXL expression in 254 basal-like breast cancers. Supplementary Figure 4. Effect of the anti-AXL monoclonal antibody 20G7-D9 on matrix degradation by breast cancer cell lines. Supplementary Figure 5. GAS6 regulates expression of EMT markers in TNBC - Western Blot quantifications. Supplementary Figure 6. E-cadherin expression in TNBC cell lines
- Published
- 2023
11. Supplementary Table S1 from Therapeutic Activity of Anti-AXL Antibody against Triple-Negative Breast Cancer Patient-Derived Xenografts and Metastasis
- Author
-
Bruno Robert, Christel Larbouret, André Pèlegrin, Jean-Max Pasquet, Dany Chalbos, Charles Theillet, Nina Radosevic-Robin, Marta Jarlier, Muriel Busson, Imade Aït-Arsa, Audrey Sirvent, Clément Chevalier, Stanislas du Manoir, Myriam Chentouf, and Wilhem Leconet
- Abstract
Suppl.Table 1: Analysis of the cellular localization, pathways and gene expression modification of the 100 genes most correlated with AXL expression in 254 basal-like breast cancers.
- Published
- 2023
12. Data from Therapeutic Activity of Anti-AXL Antibody against Triple-Negative Breast Cancer Patient-Derived Xenografts and Metastasis
- Author
-
Bruno Robert, Christel Larbouret, André Pèlegrin, Jean-Max Pasquet, Dany Chalbos, Charles Theillet, Nina Radosevic-Robin, Marta Jarlier, Muriel Busson, Imade Aït-Arsa, Audrey Sirvent, Clément Chevalier, Stanislas du Manoir, Myriam Chentouf, and Wilhem Leconet
- Abstract
Purpose: AXL receptor tyrosine kinase has been described as a relevant molecular marker and a key player in invasiveness, especially in triple-negative breast cancer (TNBC).Experimental Design: We evaluate the antitumor efficacy of the anti-AXL monoclonal antibody 20G7-D9 in several TNBC cell xenografts or patient-derived xenograft (PDX) models and decipher the underlying mechanisms. In a dataset of 254 basal-like breast cancer samples, genes correlated with AXL expression are enriched in EMT, migration, and invasion signaling pathways.Results: Treatment with 20G7-D9 inhibited tumor growth and bone metastasis formation in AXL-positive TNBC cell xenografts or PDX, but not in AXL-negative PDX, highlighting AXL role in cancer growth and invasion. In vitro stimulation of AXL-positive cancer cells by its ligand GAS6 induced the expression of several EMT-associated genes (SNAIL, SLUG, and VIM) through an intracellular signaling implicating the transcription factor FRA-1, important in cell invasion and plasticity, and increased their migration/invasion capacity. 20G7-D9 induced AXL degradation and inhibited all AXL/GAS6–dependent cell signaling implicated in EMT and in cell migration/invasion.Conclusions: The anti-AXL antibody 20G7-D9 represents a promising therapeutic strategy in TNBC with mesenchymal features by inhibiting AXL-dependent EMT, tumor growth, and metastasis formation. Clin Cancer Res; 23(11); 2806–16. ©2016 AACR.
- Published
- 2023
13. Abstract 5729: Imiqualines for pancreatic cancer: first-in-class potent and synergistic inhibitors of microtubule polymerisation
- Author
-
Kevin Bigot, Véronique Garambois, Nadia Vie, Marine Bruciamacchie, Pierre-Emmanuel Colombo, Diego Tosi, Cindy Patinote, Yann Maggipinto, Pierre-Antoine Bonnet, Céline Gongora, Carine Deleuze-Masquefa, and Christel Larbouret
- Subjects
Cancer Research ,Oncology - Abstract
Background: The survival rate for patients with Pancreatic Ductal Adenocarcinoma (PDAC) is dramatically poor with a five-year survival rate less than 10%. The research of new treatments, which could complement the current therapeutic arsenal constituted by Gemcitabine, FOLFIRINOX (fluorouracile, leucovorin, irinotecan, oxaliplatin) and nab-paclitaxel, is a major challenge. Imiqualines are new original small heterocyclic chemical molecules based on the quinoxalinic moiety. Among these first in class compounds, the lead EAPB02303 (1) displays outstanding nanomolar activities comparable to those of the current best anticancer agents on a panel of human cancer cell lines, notably on poorly sensitive cancer like PDAC and melanoma. We tested here if EAPB02303 could be an attractive first in class molecule in PDAC and we conducted in-deep molecular characterization and bioinformatics studies to decipher its mechanism of action. Methods: We characterized EAPB02303 effect on tumor growth in-vitro by conducting sulforhodamine B assay on a panel of PDAC cells including cells derived from PDX (Patient Derived Xenograft) and 3D models with Cancer Associated Fibroblasts. We assessed in-vivo activity on subcutaneous PDAC xenografts mouse models. We then studied EAPB02303 effect on cell cycle, apoptosis and microtubule polymerisation by flow cytometry and immunofluorescence. We analyzed mRNAseq and Reverse Phase Protein Assay (RPPA) data of PDAC cell lines treated with EAPB02303 at multiple time points and concentrations to identify signaling pathways and key proteins implicated in EAPB02303 effect. We performed differential gene expression and gene set enrichment analysis by using EdgeR, Deseq2 and fgsea packages. We also used PharmacoGx package to seek for similar transcriptomic profiles among the CMAP perturbational database. Results: We showed that EAPB02303 exerts activity at low nanomolar concentrations in-vitro in PDAC cell lines and 3D models, and is able to reduce tumor growth in our xenografts in-vivo mouse models. We also found a potent synergy with Paclitaxel at lower concentrations of both compounds. Furthermore, we found that EAPB02303 induces mitosis arrest and impairment of spindle assembly after 24h treatment. Cells also underwent apoptosis after 48h treatment. mRNAseq and RPPA data showed activation of several signaling pathways including MAPK kinases. CMAP database mining revealed a high connectivity score of transcriptomic signatures between EAPB02303 and inhibitors of microtubule polymerization. All these data suggest that EAPB02303 is a new microtubule-disrupting agent with in-vivo activity in PDAC and in-vitro synergy with Paclitaxel, showing potential for future clinical investigations. (1) Imidazo[1,2a]quinoxalines and derivatives thereof for treating cancers. WO 2009 043934A1. Deleuze-Masqeufa C. et al. Citation Format: Kevin Bigot, Véronique Garambois, Nadia Vie, Marine Bruciamacchie, Pierre-Emmanuel Colombo, Diego Tosi, Cindy Patinote, Yann Maggipinto, Pierre-Antoine Bonnet, Céline Gongora, Carine Deleuze-Masquefa, Christel Larbouret. Imiqualines for pancreatic cancer: first-in-class potent and synergistic inhibitors of microtubule polymerisation. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5729.
- Published
- 2023
14. Imiter la réponse immunitaire humorale polyclonale
- Author
-
Marie-Alix Poul, Christel Larbouret, Thierry Chardès, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
0301 basic medicine ,Drug ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,medicine.drug_class ,media_common.quotation_subject ,Monoclonal antibody ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,[SDV.SP.MED]Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,Immunity ,Medicine ,media_common ,biology ,business.industry ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,General Medicine ,[SDV.SP]Life Sciences [q-bio]/Pharmaceutical sciences ,3. Good health ,030104 developmental biology ,Polyclonal antibodies ,Cell culture ,030220 oncology & carcinogenesis ,Immunology ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,[SDV.IB]Life Sciences [q-bio]/Bioengineering ,Antibody ,business ,Cell bank - Abstract
International audience; Monoclonal antibodies have revolutionized the treatment of many diseases, but their clinical effectiveness remains limited in some cases. Associations of antibodies binding to the same target (homo-combination) or to several different targets (hetero-combination), thereby mimicking a polyclonal humoral immune response, have demonstrated a therapeutic improvement in pre-clinical and clinical trials, mainly in the field of oncology and infectious diseases. The combinations increase the efficacy of the biological responses and override resistance mechanisms observed with antibody monotherapy. The most common method of formulating and administering antibody combinations is a separate formulation, with sequential injection of each antibody as individual drug substance. Alternatively, combined formulations are developed where the separately-produced antibodies are mixed before administration or produced simultaneously by a single cell line, or a mixture of cell lines as a polyclonal master cell bank. The regulation, the toxicity and the injection sequence of these oligoclonal antibody-based mixtures remain points to be clarified and optimized for a better therapeutic effect.; Les anticorps monoclonaux ont révolutionné le traitement de nombreuses maladies mais leur efficacité clinique reste limitée dans certains cas. Des associations d'anticorps se liant à une même cible (homo-combinaisons) ou à plusieurs cibles différentes (hétéro-combinaisons), mimant ainsi une réponse immunitaire humorale polyclonale, ont conduit à une amélioration thérapeutique dans des essais précliniques et cliniques, essentiellement en cancérologie et en infectiologie. Ces combinaisons augmentent l'efficacité des réponses biologiques et court-circuitent les mécanismes de résistances observés lors d'une monothérapie par anticorps. Le procédé de formulation et d'administration des combinaisons d'anticorps le plus fréquent est une formulation séparée, avec injection séquentielle de chaque anticorps « principe actif ». Alternativement, se développent des formulations combinées, où les anticorps produits séparément sont mélangés avant administration, ou produits simultanément par une lignée cellulaire unique ou un mélange de lignées cellulaires correspondant à une master-bank 2 cellulaire polyclonale. La réglementation, la toxicité et la séquence d'injection des mélanges oligoclonaux restent des points à éclaircir et à optimiser pour un meilleur effet thérapeutique.
- Published
- 2019
15. Improving Biologics' Effectiveness in Clinical Oncology: From the Combination of Two Monoclonal Antibodies to Oligoclonal Antibody Mixtures
- Author
-
Christel Larbouret, Thierry Chardès, Laurent Gros, André Pèlegrin, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), and Chardès, Thierry
- Subjects
Cancer Research ,[SDV.SP.MED] Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,medicine.drug_class ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Ipilimumab ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Review ,Monoclonal antibody ,03 medical and health sciences ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,[SDV.SP.MED]Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,Trastuzumab ,antibody ,medicine ,cancer ,RC254-282 ,030304 developmental biology ,combination ,0303 health sciences ,biology ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,3. Good health ,[SDV.BIO] Life Sciences [q-bio]/Biotechnology ,[SDV] Life Sciences [q-bio] ,mixture ,oligoclonal ,Oncology ,Polyclonal antibodies ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Pertuzumab ,immunotherapy ,Antibody ,Nivolumab ,[SDV.IMM.IMM] Life Sciences [q-bio]/Immunology/Immunotherapy ,business ,biologic ,medicine.drug - Abstract
Simple Summary The approval of the two antibody combinations trastuzumab/pertuzumab and ipilimumab/nivolumab in oncology has paved the way for novel antibody combinations or oligoclonal antibody mixtures to improve their efficacy in cancer. The underlying biological mechanisms and challenges of these strategies will be discussed using data from clinical trials listed in databases. These therapeutic combinations also lead to questions on how to optimize their formulation and delivery to induce a therapeutic polyclonal response in patients with cancer. Abstract Monoclonal antibodies have revolutionized the treatment of many diseases, but their clinical efficacy remains limited in some other cases. Pre-clinical and clinical trials have shown that combinations of antibodies that bind to the same target (homo-combinations) or to different targets (hetero-combinations) to mimic the polyclonal humoral immune response improve their therapeutic effects in cancer. The approval of the trastuzumab/pertuzumab combination for breast cancer and then of the ipilimumab/nivolumab combination for melanoma opened the way to novel antibody combinations or oligoclonal antibody mixtures as more effective biologics for cancer management. We found more than 300 phase II/III clinical trials on antibody combinations, with/without chemotherapy, radiotherapy, small molecules or vaccines, in the ClinicalTrials.gov database. Such combinations enhance the biological responses and bypass the resistance mechanisms observed with antibody monotherapy. Usually, such antibody combinations are administered sequentially as separate formulations. Combined formulations have also been developed in which separately produced antibodies are mixed before administration or are produced simultaneously in a single cell line or a single batch of different cell lines as a polyclonal master cell bank. The regulation, toxicity and injection sequence of these oligoclonal antibody mixtures still need to be addressed in order to optimize their delivery and their therapeutic effects.
- Published
- 2021
16. Abstract 2589: Effect of folfirinox with an ATR inhibitor on pancreatic tumor cells and its microenvironment
- Author
-
Marine Bruciamacchie, Nadia Vie, Véronique Garambois, Diego Tosi, Pierre-Emmanuel Colombo, Céline Gongora, and Christel Larbouret
- Subjects
Cancer Research ,Oncology - Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is an extremely aggressive disease.There is a clear need of new strategies and new researches to treat and diagnose these patients. Regarding treatments, surgery is possible in only 20% of cases, and the chemotherapeutic molecule Gemcitabine is unfortunately lacking a good response rate. Recently, a new polychemotherapy oxaliplatin-based (FOLFIRINOX), which is a combination of 4 drugs: oxaliplatin, irinotecan, fluorouracil and leucovorin, has been approved. It has showed a significant increase of the overall survival inpatients compared to gemcitabine, but associated with more toxicity and still limited efficiency. Most of the drugs induce their toxicity by provoking DNA damages and replication stress, leading to the activation of DNA repair pathways. In this context,our research project proposes to find a synergistic association of FOLFIRINOX with specific inhibitors of DNA damage repair -Ataxia Telangiectasia and Rad3 related inhibitor (ATRi)- to increase the efficiency of the chemotherapy while reducing itstoxicity. The resistance to chemotherapy can come from the stroma that represents up to 80% of the tumor mass. The impact of the chemotherapies on the microenvironment can be a key to increase the treatments efficiency. That’s why in this project, we studied co-culture models to look at the effect of this new polychemotherapy on tumor cells and its microenvironment, in particular Cancer-Associated Fibroblasts (CAFs). Viability matrix in 2D & 3D in vitro co-culture of tumor cells with primary CAFs were carried out. DNA damage and proteins from the DNA damage repair pathways were analysed after treatments. Cell death and autophagy pathways were studied. In vivo, immunodeficient mice xenografted with ATCC and Patient Derived Xenograft models were treated with FOLFIRINOX and ATRi to evaluate the effect on tumor progression. A synergistic effect of the association was demonstrated in vitro independently of the KRAS, ATM, TP53, BRCA1/2 mutation statuts in several pancreatic models (ATCC and derived from PDX) and in co-culture with CAFs. A higher apoptosis and DNA damages were observed in tumor cells treated with the associated drugs. These results were associated with a decrease of DNA damage repair pathways leading to more apoptosis compared to the chemotherapy alone and an inhibition of the autophagy flux. Also, a phenotypic change in the cells was found after treating with ATRi and with an increase of this particular phenotype when the chemotherapy was added. A protective effect of the CAFs on tumor cells was observed and CAF secretome was analysed. In vivo, the association inhibits significantly the tumor growth compared to each treatment alone and no toxicity was observed. Now, validation of this polychemotherapy in vivo using co-culture models in immunodeficient and immunocompetent mice are crucial to confirm the therapeutic potential of this new treatment for PDAC. Citation Format: Marine Bruciamacchie, Nadia Vie, Véronique Garambois, Diego Tosi, Pierre-Emmanuel Colombo, Céline Gongora, Christel Larbouret. Effect of folfirinox with an ATR inhibitor on pancreatic tumor cells and its microenvironment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2589.
- Published
- 2022
17. Biasing human epidermal growth factor receptor 4 (HER4) tyrosine kinase signaling with antibodies: Induction of cell death by antibody-dependent HER4 intracellular domain trafficking
- Author
-
Romain, Lanotte, Véronique, Garambois, Nadège, Gaborit, Christel, Larbouret, Astrid, Musnier, Pierre, Martineau, André, Pèlegrin, and Thierry, Chardès
- Subjects
animal structures ,Receptor, ErbB-4 ,Cell Death ,Dose-Response Relationship, Drug ,Neuregulin-1 ,Intracellular Space ,Antibodies, Monoclonal ,Original Articles ,neuregulin ,Mitochondria ,Mice ,Protein Transport ,Drug Discovery and Delivery ,Cell Line, Tumor ,4ICD ,antibody ,Animals ,Humans ,cancer ,Original Article ,HER4 ,Reactive Oxygen Species ,Epitope Mapping ,Signal Transduction - Abstract
Human epidermal growth factor receptor 4 (HER4) isoforms have oncogenic or tumor suppressor functions depending on their susceptibility to proteolytic cleavage and HER4 intracellular domain (4ICD) translocation. Here, we report that the neuregulin 1 (NRG1) tumor suppressor mechanism through the HER4 JMa/CYT1 isoform can be mimicked by the agonist anti‐HER4 Ab C6. Neuregulin 1 induced cleavage of poly(ADP‐ribose) polymerase (PARP) and sub‐G1 DNA fragmentation, and also reduced the metabolic activity of HER3−/HER4+ cervical (C‐33A) and ovarian (COV318) cancer cells. This effect was confirmed in HER4 JMa/CYT1‐, but not JMa/CYT2‐transfected BT549 triple‐negative breast cancer cells. Neuregulin 1 favored 4ICD cleavage and retention in mitochondria in JMa/CYT1‐transfected BT549 cells, leading to reactive oxygen species (ROS) production through mitochondrial depolarization. Similarly, the anti‐HER4 Ab C6, which binds to a conformational epitope located on a.a. 575‐592 and 605‐620 of HER4 domain IV, induced 4ICD cleavage and retention in mitochondria, and mimicked NRG1‐mediated effects on PARP cleavage, ROS production, and mitochondrial membrane depolarization in cancer cells. In vivo, C6 reduced growth of COV434 and HCC1187 tumor cell xenografts in nude mice. Biasing 4ICD trafficking to mitochondria with anti‐HER4 Abs to mimic NRG1 suppressor functions could be an alternative anticancer strategy., Neuregulin 1 (NRG1) induced cleavage of poly(ADP‐ribose) polymerase (PARP) in human epidermal growth factor receptor 4 (HER4) JMa/CYT1‐expressing cancer cells. NRG1 favored HER4 intracellular domain (4ICD) cleavage and retention into mitochondria leading to reactive oxygen species (ROS) production through mitochondrial depolarization in HER4 JMa/CYT1‐expressing cancer cells. Phage‐displayed selected anti‐HER4 Ab C6 induced 4ICD cleavage and retention into mitochondria, mimicking NRG1‐mediated effects on PARP cleavage, ROS production, and mitochondrial membrane depolarization. C6 Ab reduced in vivo growth of ovarian COV434 and breast HCC1187 tumor cell xenografts in nude mice.
- Published
- 2020
18. Imiter la réponse immunitaire humorale polyclonale : de l'association de deux anticorps monoclonaux aux productions oligoclonales
- Author
-
Christel, Larbouret, Marie-Alix, Poul, Thierry, Chardès, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), and CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)
- Subjects
[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,Molecular Mimicry ,Oligoclonal Bands ,Antibodies, Monoclonal ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,[SDV.SP]Life Sciences [q-bio]/Pharmaceutical sciences ,Communicable Diseases ,Immunity, Humoral ,Drug Combinations ,Antineoplastic Agents, Immunological ,[SDV.SP.MED]Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,Neoplasms ,Antibody Formation ,Antineoplastic Combined Chemotherapy Protocols ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Animals ,Humans ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,[SDV.IB]Life Sciences [q-bio]/Bioengineering ,Immunotherapy - Abstract
Monoclonal antibodies have revolutionized the treatment of many diseases, but their clinical effectiveness remains limited in some cases. Associations of antibodies binding to the same target (homo-combination) or to several different targets (hetero-combination), thereby mimicking a polyclonal humoral immune response, have demonstrated a therapeutic improvement in pre-clinical and clinical trials, mainly in the field of oncology and infectious diseases. The combinations increase the efficacy of the biological responses and override resistance mechanisms observed with antibody monotherapy. The most common method of formulating and administering antibody combinations is a separate formulation, with sequential injection of each antibody as individual drug substance. Alternatively, combined formulations are developed where the separately-produced antibodies are mixed before administration or produced simultaneously by a single cell line, or a mixture of cell lines as a polyclonal master cell bank. The regulation, the toxicity and the injection sequence of these oligoclonal antibody-based mixtures remain points to be clarified and optimized for a better therapeutic effect.Imiter la réponse immunitaire humorale polyclonale - De l’association de deux anticorps monoclonaux aux productions oligoclonales.Les anticorps monoclonaux ont révolutionné le traitement de nombreuses maladies mais leur efficacité clinique reste limitée dans certains cas. Des associations d’anticorps se liant à une même cible (homo-combinaisons) ou à plusieurs cibles différentes (hétéro-combinaisons), mimant ainsi une réponse immunitaire humorale polyclonale, ont conduit à une amélioration thérapeutique dans des essais précliniques et cliniques, essentiellement en cancérologie et en infectiologie. Ces combinaisons augmentent l’efficacité des réponses biologiques et court-circuitent les mécanismes de résistances observés lors d’une monothérapie par anticorps. Le procédé de formulation et d’administration des combinaisons d’anticorps le plus fréquent est une formulation séparée, avec injection séquentielle de chaque anticorps « principe actif ». Alternativement, se développent des formulations combinées, où les anticorps produits séparément sont mélangés avant administration, ou produits simultanément par une lignée cellulaire unique ou un mélange de lignées cellulaires correspondant à une master-bank cellulaire polyclonale. La réglementation, la toxicité et la séquence d’injection des mélanges oligoclonaux restent des points à éclaircir et à optimiser pour un meilleur effet thérapeutique.
- Published
- 2019
19. Impact of cathepsin B-sensitive triggers and hydrophilic linkers onin vitroefficacy of novel site-specific antibody–drug conjugates
- Author
-
Nicolas Joubert, Stéphanie Letast, Cyril Colas, Marie-Claude Viaud-Massuard, Eva Lles, Camille Martin, Christel Larbouret, Inmaculada Viéitez-Villemin, Emilie Allard-Vannier, Anaïs Rousseau, Marie Brachet-Botineau, Francesca Bryden, Groupe innovation et ciblage cellulaire (GICC), EA 7501 [2018-...] (GICC EA 7501), Université de Tours (UT), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Institut de Chimie Organique et Analytique (ICOA), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université d'Orléans (UO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Nanomédicaments et Nanosondes, EA 6295 (NMNS), Génétique, Immunothérapie, Chimie et Cancer ( GICC ), Université de Tours-Centre National de la Recherche Scientifique ( CNRS ), Institut de recherche en cancérologie de Montpellier ( IRCM ), Université Montpellier 1 ( UM1 ) -CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Université de Montpellier ( UM ), Institut de Chimie Organique et Analytique ( ICOA ), Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Commissariat à l'énergie atomique et aux énergies alternatives ( CEA ) -Centre National de la Recherche Scientifique ( CNRS ) -Université d'Orléans ( UO ), Nanomédicaments et Nanosondes, EA 6295 ( NMNS ), Université de Tours, and Université d'Orléans (UO)-Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut de Chimie du CNRS (INC)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Immunoconjugates ,Cell Survival ,Receptor, ErbB-2 ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Chemistry Techniques, Synthetic ,[ SDV.BBM.BM ] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Biochemistry ,Cathepsin B ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,03 medical and health sciences ,chemistry.chemical_compound ,Antineoplastic Agents, Immunological ,Drug Delivery Systems ,[SDV.SP.MED]Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,[CHIM.ANAL]Chemical Sciences/Analytical chemistry ,Cell Line, Tumor ,Neoplasms ,Humans ,Physical and Theoretical Chemistry ,ComputingMilieux_MISCELLANEOUS ,Cathepsin ,Bioconjugation ,Dipeptide ,Chemistry ,[ SDV.SP.MED ] Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,Organic Chemistry ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Trastuzumab ,Combinatorial chemistry ,In vitro ,3. Good health ,030104 developmental biology ,PEGylation ,[ CHIM.ANAL ] Chemical Sciences/Analytical chemistry ,Hydrophobic and Hydrophilic Interactions ,Oligopeptides ,Linker ,Conjugate - Abstract
Herein we describe the synthesis and evaluation of four novel HER2-targeting, cathepsin B-sensitive antibody-drug conjugates bearing a monomethylauristatin E (MMAE) cytotoxic payload, constructed via the conjugation of cleavable linkers to trastuzumab using a site-specific bioconjugation methodology. These linkers vary by both cleavable trigger motif and hydrophilicity, containing one of two cathepsin B sensitive dipeptides (Val-Cit and Val-Ala), and engendered with either hydrophilic or hydrophobic character via application of a PEG12 spacer. Through evaluation of physical properties, in vitro cytotoxicity, and receptor affinity of the resulting antibody-drug conjugates (ADCs), we have demonstrated that while both dipeptide triggers are effective, the increased hydrophobicity of the Val-Ala pair limits its utility within this type of linker. In addition, while PEGylation augments linker hydrophilicity, this change does not translate to more favourable ADC hydrophilicity or potency. While all described structures demonstrated excellent and similar in vitro cytotoxicity, the ADC with the ValCitPABMMAE linker shows the most promising combination of in vitro potency, structural homogeneity, and hydrophilicity, warranting further evaluation into its therapeutic potential.
- Published
- 2018
20. An auristatin‐based antibody‐drug conjugate targeting HER3 enhances the radiation response in pancreatic cancer
- Author
-
Laura Bourillon, Céline Bourgier, Christel Larbouret, Nadège Gaborit, Pierre-Emmanuel Colombo, Alexandre Zampieri, Nina Radosevic-Robin, Charles Theillet, Eva Llès, Thierry Chardès, Hélène Delpech, Véronique Garambois, André Pèlegrin, Marta Jarlier, Béatrice Orsetti, D. Azria, Charline Ogier, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), UNICANCER - Institut régional du Cancer Montpellier Val d'Aurelle (ICM), CRLCC Val d'Aurelle - Paul Lamarque, Imagerie Moléculaire et Stratégies Théranostiques (IMoST), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Clermont Auvergne [2017-2020] (UCA [2017-2020]), Centre National de la Recherche Scientifique (CNRS), This work was supported by the program 'Investissement d’Avenir' (grant agreement: Labex MabImprove, ANR-10-LABX-53-01, A. Pèlegrin), by INSERM Transfert (CoPoC grant HER3ADC, T. Chardès) and the SIRIC Montpellier Cancer (SIRIC Montpellier Cancer Grant INCa_Inserm_DGOS_12553, T. Chardès)., ANR-10-LABX-0053,MAbImprove,Optimization of therapeutic monoclonal antibodies development Better antibodies, better developed AND better used(2010), Chardès, Thierry, and Laboratoires d'excellence - Optimization of therapeutic monoclonal antibodies development Better antibodies, better developed AND better used - - MAbImprove2010 - ANR-10-LABX-0053 - LABX - VALID
- Subjects
Cancer Research ,Immunoconjugates ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Cell ,pancreatic cancer ,Antibodies, Monoclonal, Murine-Derived ,Mice ,0302 clinical medicine ,Pancreatic tumor ,Phosphorylation ,skin and connective tissue diseases ,irradiation ,Chemistry ,Chemoradiotherapy ,Cell cycle ,3. Good health ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,HER3/ErbB3 ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Oligopeptides ,Carcinoma, Pancreatic Ductal ,STAT3 Transcription Factor ,Radiosensitizer ,Antibody-drug conjugate ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Cell Survival ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Immunologic Factors ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Cell Proliferation ,monomethylauristatin ,medicine.disease ,Xenograft Model Antitumor Assays ,[SDV.BIO] Life Sciences [q-bio]/Biotechnology ,Radiation therapy ,Pancreatic Neoplasms ,body regions ,ADC ,Apoptosis ,Cancer research - Abstract
International audience; Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer characterized by poor response to chemo- and radiotherapy due to the lack of efficient therapeutic tools and early diagnostic markers. We previously generated the non-ligand competing anti-HER3 antibody 9F7-F11 that binds to pancreatic tumor cells and induces tumor regression in vivo in experimental models. Here, we asked whether coupling 9F7-F11 with a radiosensitizer, such as monomethylauristatin E (MMAE), by using the antibody-drug conjugate (ADC) technology could improve radiation therapy efficacy in PDAC. We found that the MMAE-based HER3 antibody-drug conjugate (HER3-ADC) was efficiently internalized in tumor cells, increased the fraction of cells arrested in G2/M, which is the most radiosensitive phase of the cell cycle, and promoted programmed cell death of irradiated HER3-positive pancreatic cancer cells (BxPC3 and HPAC cell lines). HER3-ADC decreased the clonogenic survival of irradiated cells by increasing DNA double-strand break formation (based on γH2AX level), and by modulating DNA damage repair. Tumor radiosensitization with HER3-ADC favored the inhibition of the AKT-induced survival pathway, together with more efficient caspase 3/PARP-mediated apoptosis. Incubation with HER3-ADC before irradiation synergistically reduced the phosphorylation of STAT3, which is involved in chemoradiation resistance. In vivo, the combination of HER3-ADC with radiation therapy increased the overall survival of mice harboring BxPC3, HPAC cell xenografts or patient-derived xenografts, and reduced proliferation (KI67-positive cells). Combining auristatin radiosensitizer delivery via an HER3-ADC with radiotherapy is a new promising therapeutic strategy in PDAC. This article is protected by copyright. All rights reserved.
- Published
- 2019
21. The anti-HER3 (ErbB3) therapeutic antibody 9F7-F11 induces HER3 ubiquitination and degradation in tumors through JNK1/2- dependent ITCH/AIP4 activation
- Author
-
Christel Larbouret, Marie Alix Poul, Thierry Chardès, André Pèlegrin, Gerry Melino, Philippe Mondon, Christophe Le Clorennec, Yassamine Lazrek, Olivier Dubreuil, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Institut Pasteur de la Guyane, Réseau International des Instituts Pasteur (RIIP), Millegen SA, MILEGEN-Immeuble BIOSTEP, GamaMabs Pharma SA [Toulouse] ( Centre Pierre Potier), Oncopole de Toulouse-Centre Pierre Potier [Toulouse], LFB Biotechnologies [Lille, France], Università degli Studi di Roma Tor Vergata [Roma], University of Leicester, and Herrada, Anthony
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,Receptor, ErbB-3 ,MESH: Down-Regulation ,MESH: Antibodies, Monoclonal ,Antibodies, Monoclonal, Murine-Derived ,0302 clinical medicine ,Ubiquitin ,antibody ,Medicine ,skin and connective tissue diseases ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,biology ,treatment ,Antibodies, Monoclonal ,3. Good health ,Ubiquitin ligase ,ITCH/AIP4 ,MESH: Receptor, ErbB-3 ,Oncology ,MESH: Repressor Proteins ,030220 oncology & carcinogenesis ,MESH: Mitogen-Activated Protein Kinase 9 ,Phosphorylation ,MESH: Mitogen-Activated Protein Kinase 8 ,Research Paper ,MESH: Cell Line, Tumor ,MAP Kinase Signaling System ,Ubiquitin-Protein Ligases ,Down-Regulation ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Antineoplastic Agents ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,HER3 ,Cell Line, Tumor ,cancer ,Humans ,Mitogen-Activated Protein Kinase 9 ,Mitogen-Activated Protein Kinase 8 ,Settore BIO/10 ,Protein kinase B ,PI3K/AKT/mTOR pathway ,MESH: Humans ,business.industry ,MESH: MAP Kinase Signaling System ,Ubiquitination ,MESH: Ubiquitin-Protein Ligases ,body regions ,Repressor Proteins ,030104 developmental biology ,USP9X ,MESH: Antibodies, Monoclonal, Murine-Derived ,Cancer cell ,Immunology ,biology.protein ,Cancer research ,MESH: Ubiquitination ,MESH: Antineoplastic Agents ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
International audience; We characterized the mechanism of action of the neuregulin-non-competitive anti-HER3 therapeutic antibody 9F7-F11 that blocks the PI3K/AKT pathway, leading to cell cycle arrest and apoptosis in vitro and regression of pancreatic and breast cancer in vivo. We found that 9F7-F11 induces rapid HER3 down-regulation. Specifically, 9F7-F11-induced HER3 ubiquitination and degradation in pancreatic, breast and prostate cancer cell lines was driven mainly by the itchy E3 ubiquitin ligase (ITCH/AIP4). Overexpression of the ITCH/AIP4 inhibitor N4BP1 or small-interfering RNA-mediated knockdown of ITCH/AIP4 inhibited HER3 ubiquitination/degradation and PI3K/AKT signaling blockade induced by 9F7-F11. Moreover, 9F7-F11-mediated JNK1/2 phosphorylation led to ITCH/AIP4 activation and recruitment to HER3 for receptor ubiquitination and degradation. ITCH/AIP4 activity was activated by the deubiquitinases USP8 and USP9X, as demonstrated by RNA interference. Taken together, our results suggest that 9F7-F11-induced HER3 ubiquitination and degradation in cancer cells mainly occurs through JNK1/2-dependent ITCH/AIP4 activation.
- Published
- 2016
22. Rational development of synergistic combinations of chemotherapy and molecular targeted agents for colorectal cancer treatment
- Author
-
Nadia Vie, Mélissa Gabanou, Diego Tosi, Céline Gongora, Adeline Torro, Caroline Mollevi, Christel Larbouret, Pierre Martineau, Marta Jarlier, Eve Combes, Salima Atis, Esther Pérez-Gracia, Maguy Del Rio, Institut du Cancer de Montpellier (ICM), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), The research leading to this publication was funded by Novartis, the FrenchNational Research Agency under the programme 'Investissements d’avenir'(Grant Agreement LabEx MAbImprove, ANR-10-LABX-53) and SIRIC MontpellierCancer (INCA-DGOS-Inserm 6045)., ANR-10-LABX-0053,MAbImprove,Optimization of therapeutic monoclonal antibodies development Better antibodies, better developed AND better used(2010), Duchange, Nathalie, and Laboratoires d'excellence - Optimization of therapeutic monoclonal antibodies development Better antibodies, better developed AND better used - - MAbImprove2010 - ANR-10-LABX-0053 - LABX - VALID
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Colorectal cancer ,medicine.medical_treatment ,MAP Kinase Kinase 1 ,Aminopyridines ,Mice ,Phosphokinome ,Molecular Targeted Therapy ,Chemistry ,Drug combinations ,MEK inhibitor ,Drug Synergism ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Oncology ,Colorectal Neoplasms ,HT29 Cells ,Research Article ,Signal Transduction ,medicine.drug ,Morpholines ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Irinotecan ,lcsh:RC254-282 ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,In vivo ,Genetics ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,neoplasms ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Chemotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,digestive system diseases ,030104 developmental biology ,Drug Resistance, Neoplasm ,Synergistic effect ,Cancer research ,Benzimidazoles ,Camptothecin - Abstract
Background The irinotecan-induced phosphokinome changes in colorectal cancer (CRC) cells were used to guide the selection of targeted agents to be tested in combination with irinotecan. Methods Phosphokinome profiling with peptide arrays of tumour samples from nude mice xenografted with HT29 cells and treated or not with an effective dose of irinotecan was used to identify signalling pathways activated by irinotecan treatment. Then, drugs targeting these pathways were combined in vitro with irinotecan to test potential synergistic effect. The interactions between these drug combinations were assessed by a dose matrix approach. Confirmation of the most potential combination has been confirmed in vivo in xenografted mice. Results Irinotecan induced in vivo the activation of AKT and MEK1 phosphorylation. The dose matrix approach showed that BKM120 (PI3K inhibitor) and MEK162 (MEK inhibitor) are synergistic in vitro and in vivo with a cytostatic and cytotoxic effect, while combination of BKM120 and irinotecan or MEK162 and irinotecan are only additive or even antagonistic. However, the triple combination of SN38, BKM120 and MEK162 showed a better synergistic effect that BKM120 and MEK162, indicating that the cells need to inhibit both AKT and ERK pathways to become more sensitive to irinotecan-based chemotherapies. Conclusion Analysis of chemotherapy-induced phosphokinome changes helps to elucidate the mechanisms of drug resistance and to guide the selection of targets for combination therapies with synergistic activity. Electronic supplementary material The online version of this article (10.1186/s12885-018-4712-z) contains supplementary material, which is available to authorized users.
- Published
- 2018
23. Targeting the NRG1/HER3 pathway in tumor cells and cancer-associated fibroblasts with an anti-neuregulin 1 antibody inhibits tumor growth in pre-clinical models of pancreatic cancer
- Author
-
Christel Larbouret, Thierry Chardès, Bruno Robert, Gaëlle Thomas, Pierre-Emmanuel Colombo, Véronique Garambois, Martine Pugnière, Charline Ogier, Pierre Martineau, Marta Jarlier, Céline Gongora, Nelly Pirot, Pierre Sicard, André Pèlegrin, Nadège Gaborit, Nadia Vie, Corinne Bousquet, Lucile Canterel-Thouennon, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM), UNICANCER - Institut régional du Cancer [Montpellier] (ICM), CRLCC Val d'Aurelle - Paul Lamarque, Centre de Recherches en Cancérologie de Toulouse (CRCT), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Physiologie & médecine expérimentale du Cœur et des Muscles [U 1046] (PhyMedExp), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Laboratoire de Physiopathologie et de Pharmacologie Cardiovasculaire Expérimentale (LPPCE), Université de Bourgogne (UB), Institut de recherche en cancérologie de Montpellier (IRCM - U896 Inserm - UM1), Université Montpellier 1 (UM1)-CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Institut Charles Gerhardt Montpellier - Institut de Chimie Moléculaire et des Matériaux de Montpellier (ICGM ICMMM), Université Montpellier 1 (UM1)-Université Montpellier 2 - Sciences et Techniques (UM2)-Ecole Nationale Supérieure de Chimie de Montpellier (ENSCM)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Immunociblage et radiobiologie en oncologie, Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM), CRLCC Val d'Aurelle - Paul Lamarque-Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 1 (UM1), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Institut du Cancer de Montpellier (ICM), and Chardès, Thierry
- Subjects
0301 basic medicine ,Cancer Research ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,Receptor, ErbB-3 ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Apoptosis ,[SDV.BC.IC] Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Mice ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Pancreatic tumor ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Tumor Cells, Cultured ,Tumor Microenvironment ,ComputingMilieux_MISCELLANEOUS ,Mice, Inbred BALB C ,biology ,Chemistry ,Antibodies, Monoclonal ,3. Good health ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,[SDV.IB]Life Sciences [q-bio]/Bioengineering ,Female ,immunotherapy ,[SDV.IMM.IMM] Life Sciences [q-bio]/Immunology/Immunotherapy ,Carcinoma, Pancreatic Ductal ,Signal Transduction ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Cancer-associated fibroblast ,Neuregulin-1 ,Mice, Nude ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,03 medical and health sciences ,Stroma ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,HER3 ,Pancreatic cancer ,mental disorders ,Neuregulin 1 ,medicine ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Animals ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Cell Proliferation ,[SDV.IB] Life Sciences [q-bio]/Bioengineering ,Immunotherapy ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Coculture Techniques ,[SDV.BIO] Life Sciences [q-bio]/Biotechnology ,Pancreatic Neoplasms ,030104 developmental biology ,Tumor progression ,Cancer cell ,biology.protein ,Cancer research - Abstract
International audience; Neuregulin 1 (NRG1), a ligand for HER3 and HER4 receptors, is secreted by both pancreatic tumor cells (PC) and cancer-associated fibroblasts (CAFs), the latter representing the most abundant compound of pancreatic stroma. This desmoplastic stroma contributes to Pancreatic Ductal Adenocarcinoma (PDAC) aggressiveness and therapeutic failure by promoting tumor progression, invasion and resistance to chemotherapies. In the present work, we aimed at disrupting the complex crosstalk between PC and CAF in order to prevent tumor cell proliferation. To do so, we demonstrated the promising tumor growth inhibitory effect of the 7E3, an original antibody directed to NRG1. This antibody promotes antibody dependent cellular cytotoxicity in NRG1-positive PC and CAFs and inhibits NRG1-associated signaling pathway induction, by blocking NRG1-mediated HER3 activation. Moreover, 7E3 inhibits migration and growth of pancreatic cancer cells co-cultured with CAFs, both in vitro and in vivo using orthotopic pancreatic tumor xenografts. Our preclinical results demonstrate that the anti-NRG1 antibody 7E3 could represent a promising approach to target pancreatic stroma and cancer cells, thereby providing novel therapeutic options for PDAC.
- Published
- 2018
24. Neuregulin 1 Allosterically Enhances the Antitumor Effects of the Noncompeting Anti-HER3 Antibody 9F7-F11 by Increasing Its Binding to HER3
- Author
-
Véronique Garambois, Philippe Mondon, André Pèlegrin, Olivier Dubreuil, Yassamine Lazrek, Christel Larbouret, Marie-Alix Poul, Gérard Mathis, Jean-Marc Barret, Thierry Chardès, Jean-François Prost, Christophe Le Clorennec, Charline Ogier, Hervé Bazin, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Institut du Cancer de Montpellier (ICM), Cisbio Bioassays [Codolet, France] (Innovation Management / CbB), GamaMabs Pharma SA [Toulouse] ( Centre Pierre Potier), Oncopole de Toulouse-Centre Pierre Potier [Toulouse], Millegen SA, and MILEGEN-Immeuble BIOSTEP
- Subjects
0301 basic medicine ,Cancer Research ,Allosteric modulator ,Receptor, ErbB-3 ,Neuregulin-1 ,[SDV]Life Sciences [q-bio] ,Biology ,Pharmacology ,neuregulin ,Antibodies, Monoclonal, Murine-Derived ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,HER3 ,Neoplasms ,antibody ,mental disorders ,Biomarkers, Tumor ,Fluorescence Resonance Energy Transfer ,Animals ,Humans ,Phosphorylation ,Neuregulin 1 ,Cytotoxicity ,skin and connective tissue diseases ,Cell Proliferation ,Cancer ,allostery ,treatment ,Cell growth ,Ligand (biochemistry) ,Xenograft Model Antitumor Assays ,Antibodies, Anti-Idiotypic ,3. Good health ,Gene Expression Regulation, Neoplastic ,body regions ,030104 developmental biology ,Oncology ,A549 Cells ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Female ,Antibody ,Protein Binding ,Signal Transduction - Abstract
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.
- Published
- 2017
25. Therapeutic Activity of Anti-AXL Antibody against Triple-Negative Breast Cancer Patient-Derived Xenografts and Metastasis
- Author
-
Imade Ait-Arsa, Muriel Busson, André Pèlegrin, Christel Larbouret, Nina Radosevic-Robin, Jean-Max Pasquet, Dany Chalbos, Audrey Sirvent, Clément Chevalier, Charles Theillet, Marta Jarlier, Wilhem Leconet, Bruno Robert, Stanislas du Manoir, Myriam Chentouf, Centre Jean Perrin [Clermont-Ferrand] (UNICANCER/CJP), UNICANCER, Imagerie Moléculaire et Stratégies Théranostiques (IMoST), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Clermont Auvergne [2017-2020] (UCA [2017-2020]), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Institute of Mathematical Statistics and Actuarial Science [Bern] (IMSV), University of Bern, Centre de recherche en Biologie Cellulaire (CRBM), Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 1 (UM1), Transfert de Genes a Visee Therapeutique Dans les Cellules Souches, Université Bordeaux Segalen - Bordeaux 2-Institut National de la Santé et de la Recherche Médicale (INSERM), Immunociblage des Tumeurs et Ingenierie des Anticorps, Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de recherche en Biologie cellulaire de Montpellier (CRBM), and Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Cancer Research ,Cell signaling ,Epithelial-Mesenchymal Transition ,Triple Negative Breast Neoplasms ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Triple-negative breast cancer ,ComputingMilieux_MISCELLANEOUS ,Cell Proliferation ,AXL receptor tyrosine kinase ,GAS6 ,Receptor Protein-Tyrosine Kinases ,Bone metastasis ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Axl Receptor Tyrosine Kinase ,Antibodies, Anti-Idiotypic ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Immunology ,Cancer research ,Heterografts ,Female ,Signal Transduction - Abstract
Purpose: AXL receptor tyrosine kinase has been described as a relevant molecular marker and a key player in invasiveness, especially in triple-negative breast cancer (TNBC). Experimental Design: We evaluate the antitumor efficacy of the anti-AXL monoclonal antibody 20G7-D9 in several TNBC cell xenografts or patient-derived xenograft (PDX) models and decipher the underlying mechanisms. In a dataset of 254 basal-like breast cancer samples, genes correlated with AXL expression are enriched in EMT, migration, and invasion signaling pathways. Results: Treatment with 20G7-D9 inhibited tumor growth and bone metastasis formation in AXL-positive TNBC cell xenografts or PDX, but not in AXL-negative PDX, highlighting AXL role in cancer growth and invasion. In vitro stimulation of AXL-positive cancer cells by its ligand GAS6 induced the expression of several EMT-associated genes (SNAIL, SLUG, and VIM) through an intracellular signaling implicating the transcription factor FRA-1, important in cell invasion and plasticity, and increased their migration/invasion capacity. 20G7-D9 induced AXL degradation and inhibited all AXL/GAS6–dependent cell signaling implicated in EMT and in cell migration/invasion. Conclusions: The anti-AXL antibody 20G7-D9 represents a promising therapeutic strategy in TNBC with mesenchymal features by inhibiting AXL-dependent EMT, tumor growth, and metastasis formation. Clin Cancer Res; 23(11); 2806–16. ©2016 AACR.
- Published
- 2017
26. FOXO3a and the MAPK p38 are activated by cetuximab to induce cell death and inhibit cell proliferation and their expression predicts cetuximab efficacy in colorectal cancer
- Author
-
Vincent Denis, Christel Larbouret, Laetitia K. Linares, Esther Pérez-Gracia, Delphine Desigaud, Adeline Ayrolles-Torro, Nadia Vie, M. del Rio, Marta Jarlier, Clara Montagut, Diego Tosi, E W-F Lam, Laetitia Marzi, Mar Iglesias, Céline Gongora, Pierre Martineau, Eve Combes, Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Department of Medical Oncology [Barcelona, Spain], Hospital del Mar [Barcelona, Spain], Department of Surgery & Cancer [London, UK], Imperial College London-Cancer Imaging Centre [London, UK], This work was supported by INSERM and ICM Val D’Aurelle. LM had a PhD studentship from the French government. EC has a PhD studentship from the Labex MabImprove. AT had a MabImprove post-doctoral fellowship. The research leading to this publication was funded by the French National Research Agency under the programme ‘Investissements d’avenir’ Grant Agreement LabEx MAbImprove: ANR-10-LABX-53 and SIRIC (INCA-DGOS-Inserm 6045)., ANR-10-LABX-53-01/10-LABX-0053,MAbImprove,Optimization of therapeutic monoclonal antibodies development - Better antibodies, better developed and better used(2010), IMIM-Hospital del Mar, Generalitat de Catalunya, Imperial College London, ANR-10-LABX-0053,MAbImprove,Optimization of therapeutic monoclonal antibodies development Better antibodies, better developed AND better used(2010), Imperial College Trust, Herrada, Anthony, and Laboratoires d'excellence - Optimization of therapeutic monoclonal antibodies development Better antibodies, better developed AND better used - - MAbImprove2010 - ANR-10-LABX-0053 - LABX - VALID
- Subjects
0301 basic medicine ,Oncology ,MESH: Cell Death ,MESH: Signal Transduction ,Cancer Research ,Colorectal cancer ,Cetuximab ,medicine.disease_cause ,p38 Mitogen-Activated Protein Kinases ,MESH: ErbB Receptors ,MESH: Antibodies, Monoclonal ,Mice ,0302 clinical medicine ,MESH: Cetuximab ,MESH: Up-Regulation ,MAPK p38 ,MESH: Animals ,signalling ,Cell Death ,Forkhead Box Protein O3 ,Antibodies, Monoclonal ,MESH: Gene Expression Regulation, Neoplastic ,3. Good health ,Up-Regulation ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,[SDV.SP.PHARMA] Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,Biomarker (medicine) ,Female ,KRAS ,MESH: Caco-2 Cells ,Colorectal Neoplasms ,MESH: ras Proteins ,medicine.drug ,Signal Transduction ,medicine.medical_specialty ,Programmed cell death ,MESH: Cell Line, Tumor ,Mice, Nude ,Context (language use) ,Antineoplastic Agents ,colorectal cancer ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,MESH: Forkhead Box Protein O3 ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Internal medicine ,Cell Line, Tumor ,MESH: Cell Proliferation ,medicine ,MESH: Mice, Nude ,Animals ,Humans ,Oncology & Carcinogenesis ,FOXO3a ,MESH: Mice ,neoplasms ,Cell Proliferation ,MESH: Humans ,Cell growth ,business.industry ,[SDV.MHEP.HEG]Life Sciences [q-bio]/Human health and pathology/Hépatology and Gastroenterology ,medicine.disease ,[SDV.MHEP.HEG] Life Sciences [q-bio]/Human health and pathology/Hépatology and Gastroenterology ,digestive system diseases ,MESH: p38 Mitogen-Activated Protein Kinases ,030104 developmental biology ,Apoptosis ,ras Proteins ,[SDV.SP.PHARMA]Life Sciences [q-bio]/Pharmaceutical sciences/Pharmacology ,MESH: Antineoplastic Agents ,Caco-2 Cells ,business ,Translational Therapeutics ,1112 Oncology And Carcinogenesis ,MESH: Female ,MESH: Colorectal Neoplasms ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
BACKGROUND: Cetuximab, a monoclonal antibody against EGFR used for the treatment of colorectal cancer (CRC), is ineffective in many patients. The aim of this study was to identify the signalling pathways activated by cetuximab in CRC cells and define new biomarker of response. METHODS: We used in vitro, in vivo models and clinical CRC samples to assess the role of p38 and FOXO3a in cetuximab mechanism of action. RESULTS: We show that cetuximab activates the MAPK p38. Specifically, p38 inhibition reduced cetuximab efficacy on cell growth and cell death. At the molecular level, cetuximab activates the transcription factor FOXO3a and promotes its nuclear translocation via p38-mediated phosphorylation, leading to the upregulation of its target genes p27 and BIM and the subsequent induction of apoptosis and inhibition of cell proliferation. Finally, we found that high FOXO3a and p38 expression levels are associated with better response rate and improved outcome in cetuximab-treated patients with CRC harbouring WT KRAS. CONCLUSIONS: We identify FOXO3a as a key mediator of cetuximab mechanism of action in CRC cells and define p38 as its activator in this context. Moreover, high FOXO3a and p38 expression could predict the response to cetuximab in patients with CRC harbouring WT KRAS.British Journal of Cancer advance online publication, 29 September 2016; doi:10.1038/bjc.2016.313 www.bjcancer.com.
- Published
- 2016
27. Modulation biologique des radiations ionisantes par les agonistes des Toll-like receptors : vers une amélioration de l’index thérapeutique de la radiothérapie ?
- Author
-
Olivier Riou, David Azria, Christel Larbouret, and Bruno Robert
- Subjects
Cancer Research ,biology ,medicine.medical_treatment ,Hematology ,General Medicine ,3. Good health ,Ionizing radiation ,Radiation therapy ,03 medical and health sciences ,0302 clinical medicine ,Therapeutic index ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,biology.protein ,Radiology, Nuclear Medicine and imaging ,Radiosensitivity ,Receptor ,Flagellin ,030215 immunology - Abstract
Toll-like receptors are ubiquitous and very well conserved throughout evolution, with important functions mediating innate and adaptative immunological mechanisms. The importance of these receptors and their agonists has been recently pointed out in immunology and cancerology, although the accurate underlying mechanisms are still under investigation. The association of agonists of these receptors with ionizing radiation has been studied in preclinical experiments with promising results. Part of these compounds is flagellin, which seems to be able to modulate the radiosensitivity of both tumors and healthy tissues.
- Published
- 2012
28. Time-resolved Fluorescence Resonance Energy Transfer (TR-FRET) to Analyze the Disruption of EGFR/HER2 Dimers
- Author
-
Caroline Bascoul-Mollevi, Christel Larbouret, Julie Vallaghe, Herve Bazin, David Azria, Nadège Gaborit, Thierry Chardès, Marie-Alix Poul, Gérard Mathis, Evelyne Crapez, Frédéric Peyrusson, and André Pèlegrin
- Subjects
0303 health sciences ,biology ,Cetuximab ,Cell Biology ,Lapatinib ,Biochemistry ,Molecular biology ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Trastuzumab ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Erlotinib ,Epidermal growth factor receptor ,Pertuzumab ,skin and connective tissue diseases ,Erlotinib Hydrochloride ,neoplasms ,Molecular Biology ,Tyrosine kinase ,030304 developmental biology ,medicine.drug - Abstract
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.
- Published
- 2011
29. Keystone Symposium on Antibodies as Drugs
- Author
-
Bruno Robert, Thierry Wurch, and Christel Larbouret
- Subjects
Gerontology ,Research groups ,business.industry ,Immunology ,Immunology and Allergy ,Medicine ,Library science ,University medical ,business ,Hoffmann-LaRoche - Abstract
The symposium on Antibodies as Drugs, organized by Keystone Symposia and chaired by J. Marks, (University of California Los Angeles, USA), E.S. Ward (University of Texas Southwestern Medical Center, USA) and L. Weiner (Georgetown University Medical Center, USA), was held in Whistler, British Columbia. This Canadian Rockies village, which will host the 2010 Olympic Games, served as an enchanting backdrop to the meeting. The >350 speakers and attendees included scientists from major pharmaceutical firms, e.g., Abbott, MedImmune/Astra Zeneca, Bristol-Myers Squibb, Merck & Co, Pfizer, Sanofi-Aventis, Schering, GlaxoSmithKline, Eli Lilly, Hoffmann LaRoche, Novartis, Wyeth, and biotechnology companies, e.g., Ablynx, Medarex, Morphosys, GenMab, Amgen, Genentech, ImmunoGen, Agensys, Domantis, Biogen Idec, Centocor, LFB, Micromet, PDL Biopharma, Borean Pharma, Dyax Corp, Symphogen, Syntonix. Academic research groups at Imperial College London, University of Oxford, ETH Zurich, Scripps, Institute Cochin, Karolinsk...
- Published
- 2009
30. Association d’anticorps anti-EGFR et anti-HER2
- Author
-
David Azria, Isabelle Teulon, Christel Larbouret, Bruno Robert, and André Pèlegrin
- Subjects
Chemotherapy ,Pancreatic disease ,Anticorps monoclonal ,business.industry ,medicine.medical_treatment ,medicine ,Cancer ,General Medicine ,medicine.disease ,business ,Molecular biology ,General Biochemistry, Genetics and Molecular Biology - Published
- 2007
31. Examination of HER3 targeting in cancer using monoclonal antibodies
- Author
-
Moshit Lindzen, Christel Larbouret, Myriam Chentouf, Manjusha Ghosh, Lucile Mounier, Yosef Yarden, Sai Dunoyer, Sara Lavi, Ali Abdul-Hai, Thierry Chardès, André Pèlegrin, Michael Sela, Hervé Bazin, Nadège Gaborit, Orith Leitner, Maicol Mancini, Department of Biological Regulation [Rehovot, Israel], Weizmann Institute of Science [Rehovot, Israël], Department of Internal Medicine [Rehovot, Israel], Kaplan Medical Center [Rehovot, Israel], Department of Biological Services [Rehovot, Israel], Cisbio Bioassays [Codolet, France] (Innovation Management / CbB), Institut de recherche en cancérologie de Montpellier (IRCM - U896 Inserm - UM1), Université Montpellier 1 (UM1)-CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Department of Immunology [Rehovot, Israël], Our laboratories are supported by the Israel Cancer Research Fund, the M. D. Moross Cancer Research Institute, and the Dr. Miriam and Sheldon G. Adelson Medical Research Foundation., Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), and KARLI, Mélanie
- Subjects
Receptor, ErbB-3 ,medicine.drug_class ,antibody combination ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Pharmacology ,Monoclonal antibody ,Receptor tyrosine kinase ,03 medical and health sciences ,0302 clinical medicine ,Drug Delivery Systems ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,HER3 ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Humans ,Kinase activity ,Autocrine signalling ,skin and connective tissue diseases ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,Cancer ,Antibodies, Monoclonal ,tyrosine kinase ,Biological Sciences ,medicine.disease ,3. Good health ,body regions ,030220 oncology & carcinogenesis ,biology.protein ,cancer therapy ,Antibody ,Tyrosine kinase ,signal transduction - Abstract
International audience; The human EGF receptor (HER/EGFR) family of receptor tyrosine kinases serves as a key target for cancer therapy. Specifically, EGFR and HER2 have been repeatedly targeted because of their genetic aberrations in tumors. The therapeutic potential of targeting HER3 has long been underestimated, due to relatively low expression in tumors and impaired kinase activity. Nevertheless, in addition to serving as a dimerization partner of EGFR and HER2, HER3 acts as a key player in tumor cells' ability to acquire resistance to cancer drugs. In this study, we generated several monoclonal antibodies to HER3. Comparisons of their ability to degrade HER3, decrease downstream signaling, and inhibit growth of cultured cells, as well as recruit immune effector cells, selected an antibody that later emerged as the most potent inhibitor of pancreatic cancer cells grown as tumors in animals. Our data predict that anti-HER3 antibodies able to intercept autocrine and stroma-tumor interactions might strongly inhibit tumor growth, in analogy to the mechanism of action of anti-EGFR antibodies routinely used now to treat colorectal cancer patients.
- Published
- 2015
32. A bispecific antibody to enhance radiotherapy by tumor necrosis factor-α in human CEA–expressing digestive tumors
- Author
-
Christel Larbouret, David Azria, Jean-Bernard Dubois, André Pèlegrin, Sophie Gourgou, Pierre Martineau, Bruno Robert, and Véronique Garambois
- Subjects
Radiation-Sensitizing Agents ,Cancer Research ,medicine.medical_specialty ,Pathology ,CD30 ,medicine.medical_treatment ,Urology ,Mice, Nude ,Antineoplastic Agents ,Tumor M2-PK ,Mice ,Carcinoembryonic antigen ,Cell Line, Tumor ,Pancreatic cancer ,Antibodies, Bispecific ,medicine ,Carcinoma ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Clonogenic assay ,Tumor Stem Cell Assay ,Radiation ,biology ,Tumor Necrosis Factor-alpha ,business.industry ,Radiotherapy Dosage ,medicine.disease ,Carcinoembryonic Antigen ,Neoplasm Proteins ,Pancreatic Neoplasms ,Radiation therapy ,Oncology ,Tumor progression ,Colonic Neoplasms ,biology.protein ,Female ,business - Abstract
Tumor necrosis factor-alpha (TNF-alpha) enhances X-ray killing of human tumor cells in vitro and enhances tumor control when combined with radiotherapy (RT) in animal tumor models. In multiple Phase I studies, intravenous injection of TNF-alpha appeared to have severe systemic side effects. To overcome these limitations, we used a bispecific antibody (BAb) directed against carcinoembryonic antigen and human TNF-alpha to target this cytokine in human digestive carcinoma treated with simultaneous RT. We used human digestive carcinoma cell lines (colon cancer, LS174T, and pancreatic cancer, BxPC-3) to determine the interaction of TNF-alpha and RT on clonogenic cytotoxicity. Isobolograms were established to confirm additive or supra-additive effects between both treatments. LS174T and BxPC-3 cells were grafted subcutaneously at Day 0 into female nude mice (7-8 weeks old). When the tumors reached a volume of about 80 mm(3), the mice were randomly assigned to treatment: Group 1, normal saline i.v. injection (control group); Group 2, TNF-alpha at 1 microg/i.v. injection; Group 3, BAb at 25 microg/i.v. injection; Group 4, BAb plus TNF-alpha (ratio 25 microg to 1 microg) i.v. injection; Group 5, local RT plus normal saline (0.5 Gy. min(-1)) at a total dose of 30 Gy delivered in five fractions; Group 6, local RT plus TNF-alpha injections 3 h before RT; Group 7, local RT plus BAb plus TNF-alpha co-injected 24 h before RT. Tumor growth delay was used as the end point for all groups. In the LS174T experiments, TNF-alpha added 12 h before RT showed a statistically significant decrease in the survival fraction at 2 Gy compared with RT alone (0.23 vs. 0.42 Gy, p = 0.0017). These results were largely confirmed with the BxPC-3 cell lines (0.29 vs. 0.72, p0.00001). Isobolograms confirmed the additivity between TNF-alpha and RT in both cell lines. At 50% survival, the data points were within the envelope of additivity. In the LS174T and BxPC-3 xenografts, RT as a single agent (Group 5) slowed tumor progression compared with Group 1 (p0.027 and p = 0.00001, respectively). TNF-alpha alone, BAb alone, or BAb plus TNF-alpha (Groups 2, 3, and 4) had no effect. In the LS174T model, TNF-alpha plus RT enhanced the delay to reach 2000 mm(3) compared with RT alone but without statistical significance. This delay was significantly longer when BAb was added (p = 0.0033, for Group 6 vs. Group 7). In the BxPC-3 experiments, the median delay to reach 2000 mm(3) was similar between the RT and TNF-alpha plus RT groups (93 days). The use of our BAb in combination with TNF-alpha and RT dramatically enhanced this median delay (177 days, p = 0.0013). No body weight loss was observed in any group. Our data could be used as a solid preclinical rationale on which to base a clinical study of locally advanced pancreatic or rectal cancers in the near future.
- Published
- 2004
33. Potentiation of ionising radiation by targeting tumour necrosis factor alpha using a bispecific antibody in human pancreatic cancer
- Author
-
André Pèlegrin, Véronique Garambois, Andrew Kramar, Marc Ychou, Christel Larbouret, Jean-Bernard Dubois, Pierre Martineau, Bruno Robert, David Azria, and N Aillères
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Pancreatic disease ,medicine.medical_treatment ,Antineoplastic Agents ,Adenocarcinoma ,Radiation Tolerance ,S Phase ,tumour necrosis factor alpha ,Causes of cancer ,pancreas cancer ,Radiation, Ionizing ,Internal medicine ,Pancreatic cancer ,Antibodies, Bispecific ,Tumor Cells, Cultured ,medicine ,Humans ,Experimental Therapeutics ,Survival rate ,Chemotherapy ,Tumor Necrosis Factor-alpha ,business.industry ,radiation enhancement ,Cancer ,medicine.disease ,Carcinoembryonic Antigen ,Pancreatic Neoplasms ,Radiation therapy ,bispecific antibody ,business - Abstract
The aim of this study was to treat carcinoembryonic antigen (CEA)-expressing pancreatic carcinoma cells with tumour necrosis factor alpha (TNFalpha) and simultaneous radiation therapy (RT), using a bispecific antibody (BAb) anti-TNFalpha/anti-CEA. TNFalpha used alone produced a dose-dependent inhibition of the clonogenic capacity of the cultured cells. Flow cytometry analysis of cell cycle progression confirmed the accumulation of cells in G(1) phase after exposure to TNFalpha. When TNFalpha was added 12 h before RT, the surviving fraction at 2 Gy was 60% lower than that obtained with irradiation alone (0.29 vs 0.73, respectively, P0.00001). In combination treatment, cell cycle analysis demonstrated that TNFalpha reduced the number of cells in radiation-induced G(2) arrest, blocked irreversibly the cells in G(1) phase, and showed an additive decrease of the number of cells in S phase. In mice, RT as a single agent slowed tumour progression as compared with the control group (P0.00001). BAb+TNFalpha+RT combination enhanced the delay for the tumour to reach 1500 mm(3) as compared with RT alone or with RT+TNFalpha (P=0.0011). Median delays were 90, 93, and 142 days for RT alone, RT+TNFalpha, and RT+BAb+TNFalpha groups, respectively. These results suggest that TNFalpha in combination with BAb and RT may be beneficial for the treatment of pancreatic cancer in locally advanced or adjuvant settings.
- Published
- 2003
34. Preclinical validation of AXL receptor as a target for antibody-based pancreatic cancer immunotherapy.: Anti-AXL mAb for pancreatic cancer immunotherapy
- Author
-
Madeline Neiveyans, Muriel Busson, Bruno Robert, Christel Larbouret, Thierry Chardès, Marta Jarlier, Florence Bernex, Jean-Max Pasquet, Gaëlle Thomas, Frédérique Penault-Llorca, Martine Pugnière, Wilhem Leconet, Nina Radosevic-Robin, André Pèlegrin, Institut de recherche en cancérologie de Montpellier (IRCM - U896 Inserm - UM1), Université Montpellier 1 (UM1)-CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), Département d'anatomopathologie, Centre Jean Perrin [Clermont-Ferrand] (UNICANCER/CJP), UNICANCER-UNICANCER-UNICANCER, Hématopoïèse Leucémique et Cible Thérapeutique, Université Bordeaux Segalen - Bordeaux 2, Laboratoire d'hématologie, CHU Bordeaux [Bordeaux], Transfert de gènes à visée thérapeutique dans les cellules souches, Université Bordeaux Segalen - Bordeaux 2-Institut National de la Santé et de la Recherche Médicale (INSERM), INSERM, and Oribase Pharma
- Subjects
Cancer Research ,medicine.medical_treatment ,pancreatic cancer ,medicine.disease_cause ,0302 clinical medicine ,Pancreatic tumor ,Cell Movement ,Molecular Targeted Therapy ,Phosphorylation ,0303 health sciences ,Antibodies, Monoclonal ,targeted therapy ,3. Good health ,030220 oncology & carcinogenesis ,Intercellular Signaling Peptides and Proteins ,Female ,Immunotherapy ,monoclonal antibodies ,Carcinoma, Pancreatic Ductal ,Cell Survival ,RTK ,Mice, Nude ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,Article ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,In vivo ,Pancreatic cancer ,Proto-Oncogene Proteins ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Protein kinase B ,030304 developmental biology ,AXL receptor tyrosine kinase ,GAS6 ,Receptor Protein-Tyrosine Kinases ,medicine.disease ,Xenograft Model Antitumor Assays ,Axl Receptor Tyrosine Kinase ,Pancreatic Neoplasms ,Immunology ,Cancer research ,ras Proteins ,Carcinogenesis ,Proto-Oncogene Proteins c-akt - Abstract
International audience; AXL receptor tyrosine kinase (RTK) is implicated in proliferation and invasion of many cancers, particularly in pancreatic ductal adenocarcinoma (PDAC), for which new therapeutic options are urgently required. We investigated whether inhibition of AXL activity by specific monoclonal antibodies (mAbs) is efficient in limiting proliferation and migration of pancreatic cancer cells. Expression of AXL was evaluated by immunohistochemistry in 42 PDAC. The AXL role in oncogenesis was studied using the short hairpin RNA approach in a pancreatic carcinoma cell line. We further generated antihuman AXL mAbs and evaluated their inhibitory effects and the AXL downstream signaling pathways first in vitro, in a panel of pancreatic cancer cell lines and then in vivo, using subcutaneous or orthotopic pancreatic tumor xenografts. AXL receptor was found expressed in 76% (32/42) of PDAC and was predominantly present in invasive cells. The AXL-knockdown Panc-1 cells decreased in vitro cell migration, survival and proliferation, and reduced in vivo tumor growth. Two selected anti-AXL mAbs (D9 and E8), which inhibited phosphorylation of AXL and of its downstream target AKT without affecting growth arrest-specific factor 6 (GAS6) binding, induced downexpression of AXL by internalization, leading to an inhibition of proliferation and migration in the four pancreatic cancer cell lines studied. In vivo, treatment by anti-AXL mAbs significantly reduced growth of both subcutaneous and orthotopic pancreatic tumor xenografts independently of their KRAS mutation status. Our in vitro and preclinical in vivo data demonstrate that anti-human AXL mAbs could represent a new approach to the pancreatic cancer immunotherapy.Oncogene advance online publication, 18 November 2013; doi:10.1038/onc.2013.487.
- Published
- 2014
35. In Pancreatic Carcinoma, Dual EGFR/HER2 Targeting with Cetuximab/Trastuzumab Is More Effective than Treatment with Trastuzumab/Erlotinib or Lapatinib Alone: Implication of Receptors' Down-regulation and Dimers' Disruption
- Author
-
André Pèlegrin, Jean-Pierre Mach, Mickael Coelho, Emmanuelle Campigna, David Azria, Caroline Bascoul-Mollevi, Nadège Gaborit, Christel Larbouret, Bruno Robert, Thierry Chardès, Institut de recherche en cancérologie de Montpellier (IRCM - U896 Inserm - UM1), CRLCC Val d'Aurelle - Paul Lamarque-Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 1 (UM1), CRLC Val d'Aurelle-Paul Lamarque, CRLCC Val d'Aurelle - Paul Lamarque, Institut de Biochimie, Université de Lausanne (UNIL), Le Ster, Yves, Université Montpellier 1 (UM1)-CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), and Université de Lausanne = University of Lausanne (UNIL)
- Subjects
Cancer Research ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,Receptor, ErbB-2 ,pancreatic cancer ,Cetuximab ,Kaplan-Meier Estimate ,Mice, SCID ,Tyrosine-kinase inhibitor ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,tyrosine kinase inhibitor ,Trastuzumab ,Antineoplastic Combined Chemotherapy Protocols ,Phosphorylation ,[INFO.INFO-BT]Computer Science [cs]/Biotechnology ,skin and connective tissue diseases ,0303 health sciences ,Antibodies, Monoclonal ,[SDV.SP]Life Sciences [q-bio]/Pharmaceutical sciences ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,[SDV.SP] Life Sciences [q-bio]/Pharmaceutical sciences ,ErbB Receptors ,030220 oncology & carcinogenesis ,Female ,Erlotinib ,monoclonal antibodies ,Growth inhibition ,Research Article ,medicine.drug ,medicine.drug_class ,EGFR ,Down-Regulation ,Mice, Nude ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Antibodies, Monoclonal, Humanized ,Lapatinib ,lcsh:RC254-282 ,Erlotinib Hydrochloride ,Inhibitory Concentration 50 ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Growth factor receptor ,Cell Line, Tumor ,Pancreatic cancer ,HER2 ,HERdimerization ,medicine ,Animals ,Humans ,neoplasms ,030304 developmental biology ,business.industry ,Carcinoma ,medicine.disease ,Xenograft Model Antitumor Assays ,[SDV.BIO] Life Sciences [q-bio]/Biotechnology ,Pancreatic Neoplasms ,[INFO.INFO-BT] Computer Science [cs]/Biotechnology ,chemistry ,Quinazolines ,Cancer research ,Protein Multimerization ,business ,Proto-Oncogene Proteins c-akt - Abstract
International audience; We previously demonstrated the synergistic therapeutic effect of the cetuximab (anti-epidermal growth factor receptor [EGFR] monoclonal antibody, mAb)-trastuzumab (anti-HER2 mAb) combination (2mAbs therapy) in HER2(low) human pancreatic carcinoma xenografts. Here, we compared the 2mAbs therapy, the erlotinib (EGFR tyrosine kinase inhibitor [TKI])-trastuzumab combination and lapatinib alone (dual HER2/EGFR TKI) and explored their possible mechanisms of action. The effects on tumor growth and animal survival of the three therapies were assessed in nude mice xenografted with the human pancreatic carcinoma cell lines Capan-1 and BxPC-3. After therapy, EGFR and HER2 expression and AKT phosphorylation in tumor cells were analyzed by Western blot analysis. EGFR/HER2 heterodimerization was quantified in BxPC-3 cells by time-resolved FRET. In K-ras-mutated Capan-1 xenografts, the 2mAbs therapy gave significantly higher inhibition of tumor growth than the erlotinib/trastuzumab combination, whereas in BxPC-3 (wild-type K-ras) xenografts, the erlotinib/trastuzumab combination showed similar growth inhibition but fewer tumor-free mice. Lapatinib showed no antitumor effect in both types of xenografts. The efficacy of the 2mAbs therapy was partly Fc-independent because F(ab')(2) fragments of the two mAbs significantly inhibited BxPC-3 growth, although with a time-limited therapeutic effect. The 2mAbs therapy was associated with a reduction of EGFR and HER2 expression and AKT phosphorylation. BxPC-3 cells preincubated with the two mAbs showed 50% less EGFR/HER2 heterodimers than controls. In pancreatic carcinoma xenografts, the 2mAbs therapy is more effective than treatments involving dual EGFR/HER2 TKIs. The mechanism of action may involve decreased AKT phosphorylation and/or disruption of EGFR/HER2 heterodimerization.
- Published
- 2012
36. Quantification of HER expression and dimerization in patients' tumor samples using time-resolved Förster resonance energy transfer
- Author
-
Hervé Bazin, Gérard Mathis, Nadège Gaborit, Marc Ychou, Caroline Bascoul-Mollevi, Patrick Garnero, Jeanne Ramos, Florence Castan, Alexandre Ho-Pun-Cheung, Evelyne Lopez-Crapez, Christel Larbouret, André Pèlegrin, and Eric Assenat
- Subjects
Receptor, ErbB-2 ,Cell ,Cancer Treatment ,Invasive Ductal Carcinoma ,lcsh:Medicine ,Breast Neoplasms ,In Vitro Techniques ,Biochemistry ,Receptor tyrosine kinase ,Mice ,Antibody Therapy ,Cell Line, Tumor ,Growth Factors ,Breast Cancer ,Breast Tumors ,medicine ,Fluorescence Resonance Energy Transfer ,Animals ,Humans ,Receptor ,Protein Interactions ,lcsh:Science ,Biology ,Messenger RNA ,Multidisciplinary ,biology ,lcsh:R ,Proteins ,Obstetrics and Gynecology ,Cancers and Neoplasms ,Molecular biology ,ErbB Receptors ,Transmembrane Proteins ,Förster resonance energy transfer ,medicine.anatomical_structure ,Oncology ,Cell culture ,biology.protein ,Immunohistochemistry ,Medicine ,Female ,lcsh:Q ,Antibody ,Protein Multimerization ,Research Article - Abstract
Following the development of targeted therapies against EGFR and HER2, two members of the human epidermal receptor (HER) family of receptor tyrosine kinases, much interest has been focused on their expression in tumors. However, knowing the expression levels of individual receptors may not be sufficient to predict drug response. Here, we describe the development of antibody-based time-resolved Forster resonance energy transfer (TR-FRET) assays for the comprehensive analysis not only of EGFR and HER2 expression in tumor cryosections, but also of their activation through quantification of HER homo- or heterodimers. First, EGFR and HER2 expression levels were quantified in 18 breast tumors and the results were compared with those obtained by using reference methods. The EGFR number per cell determined by TR-FRET was significantly correlated with EGFR mRNA copy number (P
- Published
- 2012
37. Abstract B16: Cetuximab activates Foxo3a via the MAPK p38 to induce apoptosis and reduce cell proliferation in colorectal cancer cells
- Author
-
Vincent Denis, Céline Gongora, Christel Larbouret, Pierre Martineau, Nadia Vie, Eve Combes, Maguy Del Rio, Eric Lam, Clara Montagut, Laetitia K. Linares, Adeline Ayrolles-Torro, Mar Iglesias, Diego Tosi, and Laetitia Marzi
- Subjects
MAPK/ERK pathway ,Cancer Research ,Cetuximab ,Cell growth ,business.industry ,Colorectal cancer ,medicine.disease_cause ,medicine.disease ,digestive system diseases ,Oncology ,Apoptosis ,Immunology ,medicine ,Cancer research ,KRAS ,Signal transduction ,business ,neoplasms ,Protein kinase B ,medicine.drug - Abstract
Cetuximab is a monoclonal antibody against EGFR and is associated with 5-FU and irinotecan for the treatment of patients with colorectal cancer harboring wild type KRAS. However, cetuximab is ineffective in patients with constitutively active mutated KRAS and also in half of the patients with wild type KRAS. As cetuximab downstream cellular targets are not fully characterized, the aim of this study was to identify the signaling pathways activated by cetuximab in CRC cells. Our results show that in addition to inhibiting ERK and AKT, cetuximab also activates the mitogen-activated protein kinase (MAPK) p38. We have showed that p38 inhibition reduced cetuximab efficacy on cell growth and cell death. At the molecular level, cetuximab activates the transcription factor FOXO3a and promotes its nuclear translocation via p38-mediated phosphorylation. FOXO3a phosphorylation and activation by p38 led to the up-regulation of its target genes p27 and BIM and the subsequent induction of apoptosis and inhibition of cell proliferation. Finally, FOXO3a silencing reduced cetuximab cytotoxicity, further confirming FOXO3a role in cetuximab effect. Collectively, our results identify FOXO3a as a key mediator of cetuximab mechanism of action in CRC cells KRAS WT and define p38 as its main activator in this context. Citation Format: Laetitia Marzi, Eve Combes, Nadia VIE, Diego Tosi, Adeline Ayrolles-Torro, Christel Larbouret, Vincent Denis, Laetitia K. Linares, Clara Montagut, Mar Iglesias, Pierre Martineau, Eric W-F Lam, Maguy Del Rio, Céline Gongora. Cetuximab activates Foxo3a via the MAPK p38 to induce apoptosis and reduce cell proliferation in colorectal cancer cells. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr B16.
- Published
- 2015
38. 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
- Author
-
Nadège, Gaborit, Christel, Larbouret, Julie, Vallaghe, Frédéric, Peyrusson, Caroline, Bascoul-Mollevi, Evelyne, Crapez, David, Azria, Thierry, Chardès, Marie-Alix, Poul, Gérard, Mathis, Hervé, Bazin, and André, Pèlegrin
- Subjects
Antibodies, Neoplasm ,Receptor, ErbB-2 ,Antineoplastic Agents ,Lapatinib ,Cell Biology ,Receptors, Fibroblast Growth Factor ,Antibodies, Monoclonal, Murine-Derived ,Erlotinib Hydrochloride ,Mice ,Cell Line, Tumor ,Neoplasms ,Fluorescence Resonance Energy Transfer ,NIH 3T3 Cells ,Quinazolines ,Animals ,Humans ,Drug Screening Assays, Antitumor ,Phosphorylation ,Protein Multimerization ,skin and connective tissue diseases ,neoplasms ,Protein Kinase Inhibitors ,Cell Proliferation - Abstract
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.
- Published
- 2011
39. Bispecific Antibodies for the Retargeting of Cytokines
- Author
-
Christel Larbouret, David Azria, Jean-Pierre Mach, Bruno Robert, and André Pèlegrin
- Subjects
Bispecific antibody ,business.industry ,Melanoma ,medicine.medical_treatment ,Cancer therapy ,Cancer ,medicine.disease ,Cancer immunotherapy ,Renal cell carcinoma ,Retargeting ,medicine ,Cancer research ,business ,Adjuvant - Abstract
The use of cytokines as adjuvant for different forms of cancer therapy represents one of the most promising areas of applied cancer research (Seruga et al.2008). As examples, Interleukin-2 is now been a classical treatment for renal cell carcinoma (FDA approved in 1992) and always evaluated for other cancer immunotherapy, interferon α for melanoma therapy (FDA approved in 1995) and others, such as IL-15 emerged as good candidate since IL-15 was ranked 1st/124 immunomodulatory drugs in clinical and preclinical trials to treat cancer by an NCI consortium (Cheever 2008).
- Published
- 2011
40. Radiocurability by targeting tumor necrosis factor-alpha using a bispecific antibody in carcinoembryonic antigen transgenic mice
- Author
-
André Pèlegrin, Bruno Robert, David Azria, Lore Santoro, Frédéric Bibeau, Sophie Gourgou, Christel Larbouret, Pierre Martineau, Isabelle Teulon, Christine Linard, Jean-Pierre Pouget, Immunociblage et radiobiologie en oncologie, Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Unité de biostatistiques en Oncologie, CRLC Val d'Aurelle, Laboratoire d'anatomo-pathologie, CRLCC Val d'Aurelle - Paul Lamarque, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), 'Comité de l'Hérault de la Ligue Nationale Contre le Cancer' and 'Fondation Gustave et Simone Prévot', and Le Ster, Yves
- Subjects
Cancer Research ,Pathology ,MESH: Combined Modality Therapy ,Necrosis ,medicine.medical_treatment ,Bispecific antibody ,MESH: Random Allocation ,Drug Evaluation, Preclinical ,Mice ,Random Allocation ,0302 clinical medicine ,Carcinoembryonic antigen ,Antibodies, Bispecific ,MESH: Immunocompromised Host ,MESH: Animals ,Tumor Stem Cell Assay ,Radiation ,biology ,MESH: Tumor Stem Cell Assay ,synergism ,Combined Modality Therapy ,3. Good health ,Cytokine ,Oncology ,MESH: Cell Survival ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,MESH: Drug Evaluation, Preclinical ,Tumor necrosis factor alpha ,Antibody ,medicine.symptom ,medicine.medical_specialty ,MESH: Carcinoembryonic Antigen ,Cell Survival ,MESH: Mice, Transgenic ,Mice, Transgenic ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Immunocompromised Host ,03 medical and health sciences ,CEA-transgenic mice ,MESH: Antibodies, Bispecific ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Antigen ,In vivo ,Tumor Necrosis FactorΑ ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,RNA, Messenger ,Clonogenic assay ,MESH: Mice ,MESH: RNA, Messenger ,MESH: Colonic Neoplasms ,MESH: Humans ,Tumor Necrosis Factor-alpha ,business.industry ,radiotherapy enhancement ,Carcinoembryonic Antigen ,MESH: Tumor Necrosis Factor-alpha ,biology.protein ,Cancer research ,business ,030215 immunology - Abstract
International audience; PURPOSE: Tumor necrosis factor-alpha (TNF-alpha) enhances radiotherapy (RT) killing of tumor cells in vitro and in vivo. To overcome systemic side effects, we used a bispecific antibody (BsAb) directed against carcinoembryonic antigen (CEA) and TNF-alpha to target this cytokine in a CEA-expressing colon carcinoma. We report the evaluation of this strategy in immunocompetent CEA-transgenic mice. METHODS AND MATERIALS: The murine CEA-transfected colon carcinoma MC-38 was used for all experiments. In vitro, clonogenic assays were performed after RT alone, TNF-alpha alone, and RT plus TNF-alpha. In vivo, the mice were randomly assigned to treatment groups: control, TNF-alpha, BsAb, BsAb plus TNF-alpha, RT, RT plus TNF-alpha, and RT plus BsAb plus TNF-alpha. Measurements of endogenous TNF-alpha mRNA levels and evaluation of necrosis (histologic evaluation) were assessed per treatment group. RESULTS: In vitro, combined RT plus TNF-alpha resulted in a significant decrease in the survival fraction at 2 Gy compared with RT alone (p < 0.00001). In vivo, we observed a complete response in 5 (50%) of 10, 2 (20%) of 10, 2 (18.2%) of 11, and 0 (0%) of 12 treated mice in the RT plus BsAb plus TNF-alpha, RT plus TNF-alpha, RT alone, and control groups, respectively. This difference was statistically significant when TNF-alpha was targeted with the BsAb (p = 0.03). The addition of exogenous TNF-alpha to RT significantly increased the endogenous TNF-alpha mRNA level, particularly when TNF-alpha was targeted with BsAb (p < 0.01). The percentages of necrotic area were significantly augmented in the RT plus BsAb plus TNF-alpha group. CONCLUSION: These results suggest that targeting TNF-alpha with the BsAb provokes RT curability in a CEA-expressing digestive tumor syngenic model and could be considered as a solid rationale for clinical trials.
- Published
- 2007
41. In vivo Therapeutic Synergism of Anti-Epidermal Growth Factor Receptor and Anti-HER2 Monoclonal Antibodies against Pancreatic Carcinomas
- Author
-
David Azria, Maha-Zohra Ladjemi, Sébastien Morisseau, Emmanuelle Campigna, Christel Larbouret, Bruno Robert, André Pèlegrin, Simon Thezenas, Frédéric Bibeau, Isabelle Navarro-Teulon, Jean-Pierre Mach, Immunociblage des Tumeurs et Ingenierie des Anticorps, Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM), Unité de biostatistiques, CRLCC Val d'Aurelle - Paul Lamarque, Laboratoire d'anatomo-pathologie, Département de Biochimie, Université de Lausanne = University of Lausanne (UNIL), Département de radiothérapie, INSERM EMI0227 was supported for this project by the CAMPLP. C. Larbouret and this study were supported by the French National League against Cancer and by Merck AG Laboratories, Germany., and Le Ster, Yves
- Subjects
Cancer Research ,medicine.medical_specialty ,Pancreatic disease ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Receptor, ErbB-2 ,medicine.drug_class ,EGFR ,pancreatic cancer ,Mice, Nude ,Antineoplastic Agents ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,Antibodies, Monoclonal, Humanized ,Monoclonal antibody ,Mice ,Growth factor receptor ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,In vivo ,Cell Line, Tumor ,Internal medicine ,HER2 ,medicine ,Carcinoma ,Animals ,Humans ,Epidermal growth factor receptor ,Mice, Inbred BALB C ,Matuzumab ,Antibodies, Monoclonal ,Drug Synergism ,Trastuzumab ,medicine.disease ,ErbB Receptors ,Pancreatic Neoplasms ,Endocrinology ,therapeutic synergism ,Oncology ,Tumor progression ,Cancer research ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,monoclonal antibodies ,Neoplasm Transplantation ,medicine.drug - Abstract
Purpose: Pancreatic carcinoma is highly resistant to therapy. Epidermal growth factor receptor (EGFR) and HER2 have been reported to be both dysregulated in this cancer. To evaluate the in vivo effect of binding both EGFR and HER2 with two therapeutic humanized monoclonal antibodies (mAb), we treated human pancreatic carcinoma xenografts, expressing high EGFR and low HER2 levels. Experimental Design: Nude mice, bearing xenografts of BxPC-3 or MiaPaCa-2 human pancreatic carcinoma cell lines, were injected twice weekly for 4 weeks with different doses of anti-EGFR (matuzumab) and anti-HER2 (trastuzumab) mAbs either alone or in combination. The effect of the two mAbs, on HER receptor phosphorylation, was also studied in vitro by Western blot analysis. Results: The combined mAb treatment significantly inhibited tumor progression of the BxPC-3 xenografts compared with single mAb injection (P = 0.006) or no treatment (P = 0.0004) and specifically induced some complete remissions. The two mAbs had more antitumor effect than 4-fold greater doses of each mAb. The significant synergistic effect of the two mAbs was confirmed on the MiaPaCa-2 xenograft and on another type of carcinoma, SK-OV-3 ovarian carcinoma xenografts. In vitro, the cooperative effect of the two mAbs was associated with a decrease in EGFR and HER2 receptor phosphorylation. Conclusions: Anti-HER2 mAb has a synergistic therapeutic effect when combined with an anti-EGFR mAb on pancreatic carcinomas with low HER2 expression. These observations may open the way to the use of these two mAbs in a large panel of carcinomas expressing different levels of the two HER receptors.
- Published
- 2007
42. MHC class I-related chain A conjugated to antitumor antibodies can sensitize tumor cells to specific lysis by natural killer cells
- Author
-
Claire Germain, Valérie Cesson, Alena Donda, Werner Held, Bruno Robert, Jean-Pierre Mach, André Pèlegrin, and Christel Larbouret
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,medicine.drug_class ,Antibodies, Neoplasm ,medicine.medical_treatment ,Monoclonal antibody ,Natural killer cell ,Cell Line ,Immunoglobulin Fab Fragments ,Cancer immunotherapy ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,Neoplasms ,MHC class I ,medicine ,Animals ,Humans ,Lymphokine-activated killer cell ,biology ,Histocompatibility Antigens Class I ,Antibodies, Monoclonal ,Immunotherapy ,Cytotoxicity Tests, Immunologic ,Flow Cytometry ,Killer Cells, Natural ,medicine.anatomical_structure ,Oncology ,Immunology ,biology.protein ,Cancer research ,Antibody - Abstract
Purpose: As a first step for the development of a new cancer immunotherapy strategy, we evaluated whether antibody-mediated coating by MHC class I–related chain A (MICA) could sensitize tumor cells to lysis by natural killer (NK) cells. Experimental Design: Recombinant MICA (rMICA) was chemically conjugated to Fab′ fragments from monoclonal antibodies specific for tumor-associated antigens, such as carcinoembryonic antigen, HER2, or CD20. Results: Flow cytometry analysis showed an efficient coating of MICA-negative human cancer cell lines with the Fab-rMICA conjugates. This was strictly dependent on the expression of the appropriate tumor-associated antigens in the target cells. Importantly, preincubation of the tumor cells with the appropriate Fab-rMICA conjugate resulted in NK cell–mediated tumor cell lysis. Antibody blocking of the NKG2D receptor in NK cells prevented conjugate-mediated tumor cell lysis. Conclusions: These results open the way to the development of immunotherapy strategies based on antibody-mediated targeting of MICA.
- Published
- 2005
43. [Radiotherapy and inhibitors of epidermal growth factor receptor: preclinical findings and preliminary clinical trials]
- Author
-
David, Azria, Christel, Larbouret, Bruno, Robert, Stéphane, Culine, Marc, Ychou, Pierre, Verrelle, Jean-Bernard, Dubois, and André, Pèlegrin
- Subjects
Radiation-Sensitizing Agents ,Receptor, ErbB-3 ,Receptor, ErbB-2 ,Antibodies, Monoclonal ,Cetuximab ,Antineoplastic Agents ,Gefitinib ,Trastuzumab ,Antibodies, Monoclonal, Humanized ,Combined Modality Therapy ,ErbB Receptors ,Neoplasms ,Quinazolines ,Animals ,Humans - Abstract
Recent studies have demonstrated that ionizing radiation activate existing cellular response pathways involving protein kinases. These pathways mediate the cytotoxic and cytoprotective responses of cell death and cell survival, respectively. Cytoprotective responses involve dominantly mitogen-activated protein kinase (MAPK) through radiation-induced activation of EGF receptors and may stimulate cell proliferation if radiation-induced damage is successfully repaired. Similarly, overexpression of EGF receptor family members or their activation by ligands expressed at normal levels may also confer radioresistance. Recent encouraging results indicate that EGF receptor inhibitors such as antibodies or small molecule tyrosine-kinase inhibitors may be effective radiosensitizers in tumors. Within the antibody class of EGF receptor inhibitors are monoclonal antibodies such as cetuximab and trastuzumab. These agents have a common target of the extracellular domain of the EGF receptor. Striking synergistic antitumor effects on human epidermoid and on adenocarcinoma cancer-cell xenografts have been observed when cetuximab treatment is combined with radiotherapy. Promising results have also been obtained from the first clinical trial with cetuximab and radiotherapy in squamous-cell carcinoma of the head and neck. Trastuzumab has been poorly studied in combination with radiotherapy but showed an increased radiosensitivity of HER2-overexpressing breast cancer cells as measured by in vitro colony-forming assays. The mechanism of radiosensitization appears to involve DNA repair. There are well over a dozen agents in the small molecule tyrosine-kinase inhibitor category but the preclinical studies in combination with radiotherapy exist only for ZD1839 and CI1033. Preliminary studies confirm the capacity of ZD1839 and radiotherapy to produce a highly significant increase in tumor growth inhibition when compared to treatment with either modality alone. Another member of the quinazoline class of small molecule tyrosine-kinase inhibitors (CI1033) has recently been examined for its impact in conjunction with radiation in a series of HER-overexpressing breast cancer cell lines. This molecule inhibits tyrosine-kinase activity in all four members of the HER family, and preclinical studies showed a synergistic interaction of CI1033 with ionizing radiation. Finally, EGF receptor family member inhibitors may themselves be effective radiosensitizers and their use in future clinical investigations are based on a solid radiobiological rational.
- Published
- 2004
44. Monoclonal and Bispecific Antibodies in Combination with Radiotherapy for Cancer Treatment
- Author
-
Jean-Bernard Dubois, Bruno Robert, Christel Larbouret, André Pèlegrin, David Azria, and Mahmut Ozsahin
- Subjects
Bispecific antibody ,biology ,business.industry ,medicine.medical_treatment ,THERAPEUTIC RADIOPHARMACEUTICALS ,Cancer treatment ,Radiation therapy ,Monoclonal ,biology.protein ,medicine ,Cancer research ,Epidermal Growth Factor Receptor Family ,Epidermal growth factor receptor ,Antibody ,business - Abstract
In recent years, the advent of new tumor-targeting vectors such as monoclonal or bispecific antibodies has resulted in the development of a new generation of therapeutic radiopharmaceuticals and an increasing research interest in this field. However, the emphasis in the overwhelming majority of such studies has been on efficacy in clinical conditions or in animal models of human cancers, and little consideration has been given to the effects of the combination of these antibodies and radiation therapy.
- Published
- 2004
45. Abstract A62: p38MAPK implication in cetuximab response
- Author
-
Christel Larbouret, Vincent Denis, Nadia Vezzio-Vie, Clara Montagut, Pierre Martineau, Maguy Del Rio, Eve Combes, Laetitia K. Linares, Laetitia Marzi, Gaëlle Thomas, Céline Gongora, and Mar Iglesias
- Subjects
Cancer Research ,Cetuximab ,Chemistry ,Cell cycle ,Pharmacology ,Lapatinib ,medicine.disease_cause ,digestive system diseases ,Oncology ,Growth factor receptor ,medicine ,Cytotoxic T cell ,Erlotinib ,KRAS ,Kinase activity ,neoplasms ,medicine.drug - Abstract
Cetuximab is used in colorectal cancer (CRC), as targeted therapy against the Epithelial Growth Factor receptor (EGFR), in association with chemotherapy (5-FU and irinotecan). Its binding inhibits signalling pathways downstream to the receptor leading to a decrease in proliferation and surviving. In KRAS mutated CRC patients, cetuximab is ineffective, and half of KRAS Wild Type (WT) patients does not respond to cetuximab either. Thus, a better knowledge of cetuximab mechanism of action will help to improve response rate. We have previously shown that activation of the MAP Kinase p38 (p38MAPK) induces irinotecan resistance in vitro and in vivo. The aim of our project is to determine, in KRAS WT colorectal cells, if p38MAPK is involved as well in the cetuximab effect. Experiments were done on two KRAS WT CRC cell lines which respond differently to cetuximab: Caco2 cells (30% of survival inhibition, p53 mutated) and DiFi cells (80% of survival inhibition, p53 Wild Type). Cytotoxic experiments combining cetuximab treatment and inhibition of p38MAPK by a pharmacological inhibitor (SB202190) were performed. We assessed apoptosis and inhibition of proliferation by FACS analysis of cell cycle and DNA synthesis. In addition, Erk, Bim and p27 were analysed by Western Blot. Our results showed that inhibition of p38MAPK by SB202190 enhances cetuximab cytotoxic effect in Caco2 cells but impairs it in DiFi cells. We also observed that inhibition of p38MAPK decreases cetuximab induced apoptosis (from 40% of cells in subG1 to 15%) and anti-proliferative effect (from 10% EdU positive cells to 40%) in DiFi cells. The prevention of cell death by SB202190 in cetuximab treated DiFi cells could be explained by Erk pathway activation and the decrease of p27 and Bim respectively involved in cellular proliferation and mitochondrial apoptosis. We have shown that genetic inhibition of p53 also prevents cetuximab cytotoxicity in DiFi cells and we observed a synergistic effect with p38MAPK inhibition leading to higher impairment of cetuximab effect. Our results have shown the same inhibiting effect of SB202190 on the cytotoxic effect of two tyrosine-kinase inhibitors targeting EGFR (lapatinib and erlotinib) in DiFi cells indicating that p38MAPK implication is linked to inhibition of EGFR kinase activity not to cetuximab only. We have shown that p38MAPK is involved in response to the inhibition of EGFR activity. However, it seems to have different role according to the cell line used and genetic background (p53 status). Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A62. Citation Format: Laetitia Marzi, Eve Combès, Nadia Vezzio-Vié, Gaelle Thomas, Christel Larbouret, Laetitia Linares, Clara Montagut, Mar Iglesias, Vincent Denis, Maguy Del Rio, Pierre Martineau, Céline Gongora. p38MAPK implication in cetuximab response. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A62.
- Published
- 2013
46. Anti-HER3 Domain 1 and 3 Antibodies Reduce Tumor Growth by Hindering HER2/HER3 Dimerization and AKT-Induced MDM2, XIAP, and FoxO1 Phosphorylation
- Author
-
Christophe Le Clorennec, Khalil Bouayadi, Nadège Gaborit, André Pèlegrin, Marta Jarlier, Véronique Garambois, Bruno Robert, Christel Larbouret, Olivier Dubreuil, Yassamine Lazrek, Wilhem Leconet, Nadia Vie, Gaëlle Thomas, Hakim Kharrat, Thierry Chardès, Philippe Mondon, Martine Pugnière, and Céline Monnet
- Subjects
Cancer Research ,Receptor, ErbB-3 ,Receptor, ErbB-2 ,Apoptosis ,FOXO1 ,Epitopes ,Mice ,0302 clinical medicine ,Antibody Specificity ,Trastuzumab ,Neoplasms ,Phosphorylation ,skin and connective tissue diseases ,0303 health sciences ,Forkhead Box Protein O1 ,Antibodies, Monoclonal ,Forkhead Transcription Factors ,Proto-Oncogene Proteins c-mdm2 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Tumor Burden ,3. Good health ,XIAP ,030220 oncology & carcinogenesis ,Mdm2 ,Female ,Dimerization ,Protein Binding ,Research Article ,medicine.drug ,Molecular Sequence Data ,X-Linked Inhibitor of Apoptosis Protein ,Biology ,Antibodies, Monoclonal, Humanized ,Inhibitor of apoptosis ,lcsh:RC254-282 ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,030304 developmental biology ,Cell Cycle Checkpoints ,body regions ,Cancer cell ,biology.protein ,Cancer research ,Proto-Oncogene Proteins c-akt - Abstract
Blockade of the human epidermal growth factor receptor 3 (HER3) and of the downstream phosphatidylinositide 3-kinase (PI3K)/AKT pathway is a prerequisite for overcoming drug resistance and to develop novel treatments for cancers that are not eligible for the currently approved targeted therapies. To this end, we generated specific antibodies (Abs) against domain 1 (D1) and domain 3 (D3) of HER3 that recognize epitopes that do not overlap with the neuregulin-binding site. The fully human H4B-121 Ab and the mouse monoclonal Abs 16D3-C1 and 9F7-F11 inhibited tumor growth in nude mice xenografted with epidermoid, pancreatic, or triple-negative breast cancer cells. The combination of one anti-HER3 Ab and trastuzumab improved tumor growth inhibition in mice xenografted with HER2low cancer cell lines, for which trastuzumab alone shows no or moderate efficiency. Ab-induced disruption of tumor growth was associated with G1 cell cycle arrest, proliferation inhibition, and apoptosis of cancer cells. Anti-HER3 Abs blocked HER2/HER3 heterodimerization and HER3 phosphorylation at the cell membrane, leading to inhibition of phosphorylation of the downstream AKT targets murine double minute 2, X-linked inhibitor of apoptosis, and forkhead box O1. This study demonstrates that anti-HER3 D1 and D3 Abs could represent a new option for immunotherapy of pancreatic and triple-negative breast cancers.
- Published
- 2013
47. Letrozole (Femara®) sensitises breast cancer cells to gamma irradiation
- Author
-
André Pèlegrin, P Pujol, Jean-Bernard Dubois, D. Azria, S Cunat, Christel Larbouret, A Bastie, Dean B. Evans, and Gilles Romieu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Radiation ,business.industry ,Letrozole ,Internal medicine ,medicine ,Radiology, Nuclear Medicine and imaging ,Breast cancer cells ,business ,Gamma irradiation ,medicine.drug - Published
- 2003
48. R77 – Oral: Détection de l’oligomérisation des récepteurs de la famille HER par Transfert d’Énergie de Fluorescence en Temps Résolu (TR-FRET) : un nouveau biomarqueur potentiel de la réponse au traitement anti-HER
- Author
-
Hervé Bazin, A. Ho-Pun-Cheung, E. Lopez-Crapez, N. Gaborit, Christel Larbouret, André Pèlegrin, J. Vallague, and Patrick Garnero
- Subjects
Cancer Research ,Oncology ,Radiology, Nuclear Medicine and imaging ,Hematology ,General Medicine - Abstract
Introduction L’oligomerisation des Recepteurs a Tyrosine-Kinase (RTKs) HER1(EGFR) et HER2 (erbB-2) est un facteur critique dans les mecanismes d’oncogenese. La surexpression de HER2, utilisee pour selectionner les patients en vue d’un traitement a l’herceptine (anti-HER2) est communement evaluee par Immunohistochimie. Cette technique n’est pas quantitative et seulement 60 % des patients repondent a l’herceptine ce qui suggere que d’autres facteurs sont impliques, entre autre les heterodimeres HER1-HER2. Methode Nous avons mesure l’oligomerisation des recepteurs HER par TR-FRET sur des cellules vivantes surexprimant HER1 et/ou HER2 (lignees SKBR3, SKOV3) ainsi que des cellules NIH-3T3 transfectees de facon stable par ces recepteurs. Les cellules en cultures sont transferees dans les puits de plaques de microtitration et incubees avec des anticorps anti-HER ou de l’EGF marques avec un donneur (Terbium) et un accepteur (d2) puis analyse a l’aide d’un fluorimetre en mode temps resolu. Resultats En utilisant un couple d’anticorps tel que l’herceptine (anti-HER2) et le cetuximab (anti-HER1) ou l’EGF marque, nous pouvons quantifier les homodimeres HER1-HER1 ainsi que les heterodimeres HER1-HER2 sur des cellules vivantes. Le signal mesure par TR-FRET augmente en fonction du niveau d’expression des recepteurs HER. Discussion L’oligomerisation des recepteurs HER peut etre mesuree quantitativement par TR-FRET et pourrait etre utilisee pour ameliorer la prediction de la reponse aux therapies anti-HER.
- Published
- 2010
49. R144: Imagerie in vivo des cancers et anticorps monoclonaux
- Author
-
Jean-Pierre Pouget, Muriel Busson, I. Teulon, Christel Larbouret, Pierre-Olivier Kotzki, and André Pèlegrin
- Subjects
Cancer Research ,Oncology ,Radiology, Nuclear Medicine and imaging ,Hematology ,General Medicine - Published
- 2010
50. New biomolecules for cancer therapy (88.2)
- Author
-
Emmanuelle Campigna, Sebastien Morisseau, Veronique Garambois, Claire Germain, Christel Larbouret, Jean-Pierre Mach, and André Pèlegrin
- Subjects
Immunology ,Immunology and Allergy - Abstract
It has been previously demonstrated that tumor cells expressing MICA are no more able to grow in vivo due to the killing of tumor cells by both NK and CD8+ T cells. In order to restore the presence of the MICA ligand at the surface of tumor cells, we designed biomolecules composed of MICA and an scFv (single chain fragment variable) that can recognise tumor associated antigens (TAA). The biomolecules should target tumor cells and re-direct the specific lysis of tumor cells by NK cells after interaction between NKG2D and exogenous MICA at the surface of tumor cells. In vitro cytometry experiments showed that the biomolecules specifically bind different tumor cells expressing TAA. Using 51Cr release assay, we demonstrated that the biomolecules induce an efficient and specific killing of tumor cells mediated by NK cells or human PMBCs. In vivo, biodistribution studies showed that the biomolecules could target xenografted tumor when injected intravenously. Consequently, a protocol is investigated for therapy.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.