14 research outputs found

    HER3 as biomarker and therapeutic target in pancreatic cancer: new insights in pertuzumab therapy in preclinical models.

    Get PDF
    International audienceThe anti-HER2 antibody pertuzumab inhibits HER2 dimerization and affects HER2/HER3 dimer formation and signaling. As HER3 and its ligand neuregulin are implicated in pancreatic tumorigenesis, we investigated whether HER3 expression could be a predictive biomarker of pertuzumab efficacy in HER2low-expressing pancreatic cancer. We correlated in vitro and in vivo HER3 expression and neuregulin dependency with the inhibitory effect of pertuzumab on cell viability and tumor progression. HER3 knockdown in BxPC-3 cells led to resistance to pertuzumab therapy. Pertuzumab treatment of HER3-expressing pancreatic cancer cells increased HER3 at the cell membrane, whereas the anti-HER3 monoclonal antibody 9F7-F11 down-regulated it. Both antibodies blocked HER3 and AKT phosphorylation and inhibited HER2/HER3 heterodimerization but affected differently HER2 and HER3 homodimers. The pertuzumab/9F7-F11 combination enhanced tumor inhibition and the median survival time in mice xenografted with HER3-expressing pancreatic cancer cells. Finally, HER2 and HER3 were co-expressed in 11% and HER3 alone in 27% of the 45 pancreatic ductal adenocarcinomas analyzed by immunohistochemistry. HER3 is essential for pertuzumab efficacy in HER2low-expressing pancreatic cancer and HER3 expression might be a predictive biomarker of pertuzumab efficacy in such cancers. Further studies in clinical samples are required to confirm these findings and the interest of combining anti-HER2 and anti-HER3 therapeutic antibodies

    Signaling and targeting of the Tyrosine Kinase Receptor AXL in cancer

    No full text
    AXL est un récepteur tyrosine kinase (RTK) impliqué dans de nombreux mécanismes cellulaires tels que la migration, l'invasion, l'angiogenèse et la prolifération des cellules. Sa surexpression a été observée dans de nombreux cancers et est souvent liée à un mauvais pronostic vital pour le patient. De plus, ce récepteur semble agir dans un mécanisme important dans la formation de métastases et la résistance aux thérapies anticancéreuses : la transition épithélio-mésenchymateuse (EMT). Nous avons dans un premier temps généré des anticorps monoclonaux murins spécifiques du récepteur AXL. Deux de ces anticorps ont ensuite été sélectionnés pour leurs propriétés inhibitrices de l'expression d'AXL à la surface et de l'activation de ce récepteur par son ligand GAS6. En effet ces deux anticorps, le 20G7D9 et le 3E3E8, entraine l'internalisation et la dégradation lysosomale d'AXL.Nous avons dans un deuxième temps étudié l'expression et le rôle de ce récepteur dans le cancer du pancréas qui possède un manque cruel de solutions thérapeutiques aujourd'hui et dont le taux de survie reste très faible (moins 5% des patients survivent 5 ans après son diagnostic). Nous avons ainsi observé une expression d'AXL dans une majorité des tumeurs de patients (76%), notamment au niveau du front invasif de ces tumeurs. Le ciblage d'AXL par nos deux anticorps inhibe sa signalisation et permet une réduction in vitro et in vivo de la croissance tumorale.Enfin, l'importante expression d'AXL dans le front invasif des tumeurs nous a incité à étudier le rôle d'AXL au cours de la transition épithélio-mésenchymateuse. Nous avons ainsi démontré que le couple AXL/GAS6 induit l'EMT dans des modèles invasifs de cancer du sein triple négatifs. De plus, l'expression du récepteur dans des tumeurs de cancer du sein de type basal-like est corrélée à celle de différents marqueurs importants dans l'EMT. L'application de nos anticorps anti-AXL dans ce type de cancer permet d'inhiber l'induction de l'EMT par le récepteur ainsi que l'invasion cellulaire in vitro et in vivo.Cette thèse a ainsi permis de démontrer l'importance du récepteur tyrosine kinase AXL dans des mécanismes oncogéniques clés et l'efficacité de son ciblage par des anticorps monoclonaux dans des modèles précliniques de cancer.The Tyrosine Kinase Receptor (TKR) AXL is implicated in various cellular mechanisms (migration, invasion, angiogenesis and cell proliferation). Its overexpression has been observed in many cancers and is often correlated with poor prognosis. Moreover, this receptor seems to be important in Epithelial to Mesenchymal Transition (EMT), a mechanism related to metastasis formation and resistance to anticancer therapies.We have generated several AXL specific murine monoclonal antibodies. Two of them, 20G7D9 and 3E3E8, have been selected for their inhibition properties in AXL expression and activation by its ligand GAS6. In fact, both antibodies induce internalization and lysosomal degradation of AXL.Then we decided to study AXL expression and role in pancreatic cancer, which is characterized by a dramatic overall survival (<5%, 5 years after diagnosis) and a lack of efficient therapeutic solutions. We observed an ectopic expression of AXL in a majority of patient' pancreatic tumors (76%), notably in the invasive front of the tumor. Targeting AXL with both 20G7D9 and 3E3E8 inhibits its signaling and decreases tumor growth in vitro and in vivo.As AXL is mainly expressed in the invasive front of tumors, we analyzed its role during EMT. We observed that AXL/GAS6 signaling induces EMT in triple negative breast cancer cell lines. Furthermore, its expression is correlated with well-defined EMT markers in basal-like breast cancer tumors. In vitro and in vivo application of our antibodies inhibits AXL-dependant EMT signaling and cellular migration and invasion.In conclusion, this thesis demonstrates the importance of AXL Tyrosine Kinase Receptor in oncogenic processes and the efficacy of targeting this receptor with monoclonal antibodies in cancer preclinical models

    Preclinical validation of AXL receptor as a target for antibody-based pancreatic cancer immunotherapy.: Anti-AXL mAb for pancreatic cancer immunotherapy

    No full text
    International audienceAXL 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

    Intra-articular delivery of full-length antibodies through the use of an in situ forming depot

    No full text
    International audienceMonoclonal antibodies (mAbs) are large size molecules that have demonstrated high therapeutic potential for the treatment of cancer or autoimmune diseases. Despite some excellent results, their intravenous administration results in high plasma concentration. This triggers off-target effects and sometimes poor targeted tissue distribution. To circumvent this issue, we investigated a local controlled-delivery approach using an in situ forming depot technology. Two clinically relevant mAbs, rituximab (RTX) and daratumumab (DARA), were formulated using an injectable technology based on biodegradable PEG-PLA copolymers. The stability and controlled release features of the formulations were investigated. HPLC and mass spectrometry revealed the preservation of the protein structure. In vitro binding of formulated antibodies to their target antigens and to their cellular FcÎłRIIIa natural killer cell receptor was fully maintained. Furthermore, encapsulated RTX was as efficient as classical intravenous RTX treatment to inhibit the in vivo tumor growth of malignant human B cells in immunodeficient NSG mice. Finally, the intra-articular administration of the formulated mAbs yielded a sustained local release associated with a lower plasma concentration compared to the intra-articular delivery of non-encapsulated mAbs. Our results demonstrate that the utilization of this polymeric technology is a reliable alternative for the local delivery of fully functional clinically relevant mAbs

    Anti-HER3 Domain 1 and 3 Antibodies Reduce Tumor Growth by Hindering HER2/HER3 Dimerization and AKT-Induced MDM2, XIAP, and FoxO1 Phosphorylation

    Get PDF
    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
    corecore