60 research outputs found

    Imiter la réponse immunitaire humorale polyclonale : de l'association de deux anticorps monoclonaux aux productions oligoclonales

    Get PDF
    International audienceMonoclonal 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 rĂ©volutionnĂ© le traitement de nombreuses maladies mais leur efficacitĂ© clinique reste limitĂ©e dans certains cas. Des associations d'anticorps se liant Ă  une mĂȘme cible (homo-combinaisons) ou Ă  plusieurs cibles diffĂ©rentes (hĂ©tĂ©ro-combinaisons), mimant ainsi une rĂ©ponse immunitaire humorale polyclonale, ont conduit Ă  une amĂ©lioration thĂ©rapeutique dans des essais prĂ©cliniques et cliniques, essentiellement en cancĂ©rologie et en infectiologie. Ces combinaisons augmentent l'efficacitĂ© des rĂ©ponses biologiques et court-circuitent les mĂ©canismes de rĂ©sistances observĂ©s lors d'une monothĂ©rapie par anticorps. Le procĂ©dĂ© de formulation et d'administration des combinaisons d'anticorps le plus frĂ©quent est une formulation sĂ©parĂ©e, avec injection sĂ©quentielle de chaque anticorps « principe actif ». Alternativement, se dĂ©veloppent des formulations combinĂ©es, oĂč les anticorps produits sĂ©parĂ©ment sont mĂ©langĂ©s avant administration, ou produits simultanĂ©ment par une lignĂ©e cellulaire unique ou un mĂ©lange de lignĂ©es cellulaires correspondant Ă  une master-bank 2 cellulaire polyclonale. La rĂ©glementation, la toxicitĂ© et la sĂ©quence d'injection des mĂ©langes oligoclonaux restent des points Ă  Ă©claircir et Ă  optimiser pour un meilleur effet thĂ©rapeutique

    Letrozole sensitizes breast cancer cells to ionizing radiation

    Get PDF
    INTRODUCTION: Radiotherapy (RT) is considered a standard treatment option after surgery for breast cancer. Letrozole, an aromatase inhibitor, is being evaluated in the adjuvant setting. We determined the effects of the combination of RT and letrozole in the aromatase-expressing breast tumour cell line MCF-7CA, stably transfected with the CYP19 gene. METHODS: Irradiations were performed using a cobalt-60 source with doses ranging from 0 to 4 Gy. Cells were incubated with androstenedione in the presence or absence of letrozole. Effects of treatment were evaluated using clonogenic assays, tetrazolium salt colorimetric (MTT) assays, and cell number determinations. Cell-cycle analyses were conducted using flow cytometry. RESULTS: The survival fraction at 2 Gy was 0.66 for RT alone and was 0.44 for RT plus letrozole (P = 0.02). Growth of MCF-7CA cells as measured by the cell number 6 days after radiotherapy (2 and 4 Gy) was decreased by 76% in those cells treated additionally with letrozole (0.7 ÎŒM) compared with those receiving radiotherapy alone (P = 0.009). Growth inhibition, assessed either by cell number (P = 0.009) or by the MTT assay (P = 0.02), was increased after 12 days of the combination treatment. Compared with radiation alone, the combination of radiation and letrozole produced a significant decrease in radiation-induced G(2 )phase arrest and a decrease of cells in the S phase, with cell redistribution in the G(1 )phase. CONCLUSIONS: These radiobiological results may form the basis for concurrent use of letrozole and radiation as postsurgical adjuvant therapy for breast cancer

    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

    Design and selection of optimal ErbB-targeting bispecific antibodies in pancreatic cancer

    Get PDF
    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 BiXAbℱ 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 BiXAbsℱ 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

    Anticorps bispécifiques pour la thérapie des cancers (ciblage tumoral du TNFa associé à la radiothérapie, ciblage simultané de deux récepteurs, EGFR et HER2)

    No full text
    MONTPELLIER-BU MĂ©decine UPM (341722108) / SudocPARIS-BIUP (751062107) / SudocMONTPELLIER-BU MĂ©decine (341722104) / SudocSudocFranceF

    Ciblage de l'entité HER2/HER3 par des anticorps monoclonaux pour le traitement des cancers du pancréas exprimant faiblement HER2.

    No full text
    Avec un taux de survie Ă  5 ans infĂ©rieur Ă  5%, l'adĂ©nocarcinome pancrĂ©atique (PDAC) est l'un des cancers les plus agressifs et pour lequel les thĂ©rapies existantes sont largement insuffisantes. Ce cancer est caractĂ©risĂ© par un tissu fibreux dense et un stroma trĂšs dĂ©veloppĂ©, en constante interaction avec la tumeur, favorisant le dĂ©veloppement d'un environnement propice Ă  la progression tumorale. Actuellement, la gemcitabine est la seule chimiothĂ©rapie approuvĂ©e capable de prolonger lĂ©gĂšrement la survie des patients. Une thĂ©rapie ciblĂ©e dirigĂ©e contre l'EGFR, l'erlotinib, a prouvĂ© que le ciblage de cette famille pourrait ĂȘtre une stratĂ©gie intĂ©ressante dans cette pathologie mais qu'une sĂ©lection des patients Ă©tait indispensable pour augmenter les rĂ©ponses thĂ©rapeutiques. Deux rĂ©cepteurs de cette famille, HER2 et HER3, dimĂ©risent pour former une entitĂ© particuliĂšrement agressive et impliquĂ©e dans la croissance tumorale des PDACs. Diverses techniques rĂ©cemment dĂ©veloppĂ©es sont utilisĂ©es pour quantifier ces dimĂšres, afin d'Ă©tudier leur rĂŽle, mais Ă©galement pour tenter de les cibler, et empĂȘcher leur signalisation dans diffĂ©rents cancers. A l'heure actuelle, le pertuzumab est le seul anticorps monoclonal dirigĂ© contre le rĂ©cepteur HER2, capable de bloquer sa dimĂ©risation, et utilisĂ© en clinique. Dans un premier temps, nous nous sommes intĂ©ressĂ©s au rĂŽle de HER3 en tant que cible et marqueur pronostique de l'effet du pertuzumab sur les PDAC exprimant faiblement HER2. Puis dans une deuxiĂšme partie, nous avons Ă©tudiĂ© et comparĂ© les effets de diffĂ©rents anticorps monoclonaux dirigĂ©s contre HER2 et/ou HER3, sur la prolifĂ©ration tumorale de tumeurs du pancrĂ©as. L'ensemble de ces rĂ©sultats a permis d'Ă©tablir que le ciblage du dimĂšre HER2/HER3 s'avĂšre ĂȘtre une stratĂ©gie prometteuse pour inhiber la croissance des tumeurs du pancrĂ©as exprimant faiblement HER2, et que le rĂ©cepteur HER3 pourrait ĂȘtre un marqueur pronostique de l'effet du pertuzumab.With a 5-year survival lower than 5%, PDAC is one of the worst cancers in terms of mortality, and for which existing therapies are unsatisfying. This cancer is characterized by a dense fibrotic tissue and an over-developed stroma, in continual interaction with the tumor, promoting the development of an ideal environment for tumor progression.To date, gemcitabine is the only approved chemotherapy able to slightly increase patients' survival. The use of erlotinib, an EGFR targeting therapy, underlined that EGFR family targeting could be an interesting treatment strategy in this pathology, but that a better patients' selection is essential to increase therapeutic response. Two receptors of this family, HER2 and HER3, are able to dimerize to consititute an aggressive entity, involved in PDAC tumoral growth. Various recently developed techniques are used to quantify those dimers, in order to study their role, but also to target them and block their signaling in cancer cells. Pertuzumab is currently the only HER2-targeting monoclonal antibody able to block its dimerization and used in clinic. We first evaluated the role of HER3 as therapeutic target and prognostic marker of pertuzumab efficacy on HER2-low expressing PDACs. We then studied and compared therapeutic effects on pancreatic tumor proliferation of different antibodies targeting HER2 and/or HER3. Taken together, those results demonstrated that HER2/HER3 dimers targeting is a promising strategy to inhibit low-HER2 expressing pancreatic tumor growth, and that HER3 could be a pronostic marker for pertuzumab efficacy in those cancers.MONTPELLIER-BU MĂ©decine UPM (341722108) / SudocSudocFranceF

    Imiter la réponse immunitaire humorale polyclonale

    No full text
    Les anticorps monoclonaux ont rĂ©volutionnĂ© le traitement de nombreuses maladies mais leur efficacitĂ© clinique reste limitĂ©e dans certains cas. Des associations d’anticorps se liant Ă  une mĂȘme cible (homo-combinaisons) ou Ă  plusieurs cibles diffĂ©rentes (hĂ©tĂ©ro-combinaisons), mimant ainsi une rĂ©ponse immunitaire humorale polyclonale, ont conduit Ă  une amĂ©lioration thĂ©rapeutique dans des essais prĂ©cliniques et cliniques, essentiellement en cancĂ©rologie et en infectiologie. Ces combinaisons augmentent l’efficacitĂ© des rĂ©ponses biologiques et court-circuitent les mĂ©canismes de rĂ©sistances observĂ©s lors d’une monothĂ©rapie par anticorps. Le procĂ©dĂ© de formulation et d’administration des combinaisons d’anticorps le plus frĂ©quent est une formulation sĂ©parĂ©e, avec injection sĂ©quentielle de chaque anticorps « principe actif ». Alternativement, se dĂ©veloppent des formulations combinĂ©es, oĂč les anticorps produits sĂ©parĂ©ment sont mĂ©langĂ©s avant administration, ou produits simultanĂ©ment par une lignĂ©e cellulaire unique ou un mĂ©lange de lignĂ©es cellulaires correspondant Ă  une master-bank cellulaire polyclonale. La rĂ©glementation, la toxicitĂ© et la sĂ©quence d’injection des mĂ©langes oligoclonaux restent des points Ă  Ă©claircir et Ă  optimiser pour un meilleur effet thĂ©rapeutique

    Improving Biologics’ Effectiveness in Clinical Oncology: From the Combination of Two Monoclonal Antibodies to Oligoclonal Antibody Mixtures

    No full text
    International audienceMonoclonal 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

    Biologic modulation of ionizing radiation by Toll-like receptors agonists: Towards an increase in the therapeutic index of radiotherapy?

    No full text
    International audienceToll-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
    • 

    corecore