13 research outputs found

    AGuIX® from bench to bedside-Transfer of an ultrasmall theranostic gadolinium-based nanoparticle to clinical medicine

    Get PDF
    International audienceAGuIX® are sub-5 nm nanoparticles made of a polysiloxane matrix and gadolinium chelates. This nanoparticle has been recently accepted in clinical trials in association with radiotherapy. This review will summarize the principal preclinical results that have led to first in man administration. No evidence of toxicity has been observed during regulatory toxicity tests on two animal species (rodents and monkeys). Biodistributions on different animal models have shown passive uptake in tumours due to enhanced permeability and retention effect combined with renal elimination of the nanoparticles after intravenous administration. High radiosensitizing effect has been observed with different types of irradiations in vitro and in vivo on a large number of cancer types (brain, lung, melanoma, head and neck…). The review concludes with the second generation of AGuIX nanoparticles and the first preliminary results on human

    Evaluation de l’effet sensibilisant des nanoparticules AGuIX® associées à une irradiation photonique dans un modèle de chondrosarcome

    No full text
    International audienceLes chondrosarcomes (CHS), tumeurs malignes du cartilage chimio- et radio-résistantes, représentent environ 20% des tumeurs osseuses primitives de l’adulte. Les nanoparticules AGuIX® associées à l’irradiation photonique ayant conduit à la radiosensibilisation de plusieurs types de tumeurs radiorésistantes (cancer de la tête et du cou, du col utérin, mélanome …), cette stratégie a été testée dans un modèle de CHS in vitro sur cultures 3D en conditions normoxique et physioxique et in vivo dans des souris avec xenogreffe tumorale de CHS (ANR CHONDRAD). L’effet radiosensibilisant des AGuIX® associées à une irradiation photonique a été analysé sur la lignée HEMC-SS dérivée d’un chondrosarcome extra squelettique myxoïde humain.La microscopie en temps réel (IncuCyte®) a permis de suivre l’évolution des sphéroïdes fluorescents HEMC-SS-mKate2 après irradiation associée ou non aux AGuIX®. La combinaison d’une irradiation unique de 4Gy ou d’une irradiation fractionnée (1Gy x 4 jours consécutifs) avec un traitement par AGuIX® a conduit à une radiosensibilisation que ce soit en condition normoxique ou physioxique (2% O2).Pour soutenir l’application clinique potentielle en radiothérapie, des études in vivo ont été réalisées sur un modèle de souris nude avec des xénogreffes HEMC-SS irradiées par des photons avec une dose unique de 4 Gy après une injection intratumorale d’AGuIX®. Les résultats (croissance tumorale et survie) montrent un effet radiosensibilisant significatif des AGuIX®.L’ensemble de ces résultats apportent la preuve de concept de l’effet radiosensibilisant des AGuIX® dans le chondrosarcome

    AGuIX radiosensitizing nanoparticles: from molecular events to clinical applications

    No full text
    International audienceAGuIX (Activation and Guidance by Irradiation X) is a non-toxic gadolinium-based nanoparticle (GBN) (due to gadolinium entrapped in DOTA) developed by the Lyon University. It accumulates in the tumor through the enhanced permeability and retention (EPR) effect and clears rapidly through the kidneys due to its small size (sub-5nm). Furthermore, AGuIX demonstrates good performance as a contrast agent for MRI.We performed a proof of concept on head and neck squamous cell carcinoma (HNSCC), metastatic melanoma and chondrosarcoma tumors, known for their low survival rates, demonstrating the radiosensitizing efficacy of the AGuIX nanoparticles in cellular (2D and 3D cultures) and preclinical models. GBNs enter HNSCC cancer cells by passive diffusion and macropinocytosis (Rima et al, 2013), localize in cytoplasm, as free particle or entrapped in lysosomes, in close vicinity to the mitochondria. Gadolinium combined with irradiation can produce a large variety of secondary emissions such as secondary, Auger, and Compton electrons leading to the production of reactive oxygen species (ROS) that trigger an intra‐mitochondrial stress (ROS production, transmembrane potential decrease, mtDNA deletion) and nuclear DNA damage leading to cell death.Relative biological efficiency (RBE) in cancer cells is quite comparable to that observed in response to carbon ions, suggesting the existence of common mechanisms through the amplification of the local dose (Miladi et al. Al., Nanomedicine 2015, Wozny et al., submitted).The efficacy of AGuIX has also been demonstrated in orthotopic xenograft models of HNSCC and metastatic melanoma (Miladi et al., 2015, Kotb et al., Theranostics 2016); the experiments are ongoing for chondrosarcoma.Regulatory toxicity studies were conducted in rats and monkeys and a first clinical study was initiated in patients with multiple brain metastases (clinicaltrial.gov).This presentation will summarize our current results with AGuIX nanoparticles used in radiobiology and imaging as well as their first clinical applications.Supported by LabEx PRIMES (ANR-11-LABX-0063

    Nanocomposite sponges for enhancing intestinal residence time following oral administration

    No full text
    In this work, nanocomposites that combine mucopenetrating and mucoadhesive properties in a single system are proposed as innovative strategy to increase drug residence time in the intestine following oral administration. To this aim, novel mucoadhesive chitosan (CH) sponges loaded with mucopenetrating nanoemulsions (NE) were developed via freeze-casting technique. The NE mucopenetration ability was determined studying the surface affinity and thermodynamic binding of the nanosystem with mucins. The ability of nanoparticles to penetrate across a preformed mucins layer was validated by 3D-time laps Confocal Laser Scanning Microscopy imaging. Microscopy observations (Scanning Electron Microscopy and Optical Microscopy) showed that NE participated in the structure of the sponge affecting its stability and in vitro release kinetics. When incubated with HCT 116 and Caco-2 cell lines, the NE proved to be cytocompatible over a wide concentration range. Finally, the in vivo biodistribution of the nanocomposite was evaluated after oral gavage in healthy mice. The intestinal retention of NE was highly enhanced when loaded in the sponge compared to the NE suspension. Overall, our results demonstrated that the developed nanocomposite sponge is a promising system for sustained drug intestinal delivery.Portuguese Foundati on for Science and Technology and European Commission: 42306YB; Portuguese Foundation for Science and Technology: UID/Multi/04326/2020info:eu-repo/semantics/publishedVersio

    Transfer of the ultrasmall theranostic gadolinium-based nanoparticle AGuIX® to clinical medicine

    No full text
    International audienceAGuIX® (Activation-and-Guidance by Irradiation X) are sub-5nm theranostic nanoparticles composed of a polysiloxane matrix and gadolinium chelates. Developed by the Lyon University, they underwent extensive pre-clinical evaluation and were recently translated to clinical evaluation in the treatment of brain metastases and advanced cervical cancer. This presentation will summarize the principal in cellulo and pre-clinical results that led to their first-in-man administration.In collaboration with other teams, we did the proof of concept of their radiosensitizing efficacy in cellular (2D/3D cultures) and preclinical models of three radioresistant tumors: head and neck squamous cell carcinoma, metastatic melanoma and chondrosarcoma tumors (Miladi et al, Nanomedicine 2015; Kotb et al, Theranostics 2016; Ollier et al, in preparation). AGuIX® enter cancer cells by passive diffusion and macropinocytosis, localize in cytoplasm as free particle or entrapped in lysosomes, in close vicinity to the mitochondria (Rima et al, Biomaterials 2013). The interaction of radiation with gadolinium atoms produce a variety of secondary emissions, such as Auger shower, leading to the production of high reactive oxygen species levels that can trigger an intra‐mitochondrial stress and nuclear DNA damage leading to cell death (Simonet al., submitted). Relative biological efficiency (RBE) in cancer cells is quite comparable to that observed in response to carbon ions, suggesting the existence of common mechanisms through the amplification of the local dose (Wozny et al, Nanomedicine 2017). AGuIX®nanoparticles have already shown their efficiency as a radiosensitizer in at least 12 preclinical models of cancer (Lux et al, BJR 2018). Biodistribution studies after intravenous administration have shown passive uptake of the nanoparticles in tumors due to enhanced permeability and retention (EPR) effect combined with rapid renal elimination. We are now testing the next generation of AGuIX® nanoparticles containing other metals or functionalized with therapeutic or targeting moieties (D. Vernos’ poster).Supported by LabEx PRIME

    Transfer of the ultrasmall theranostic gadolinium-based nanoparticle AGuIX® to clinical medicine

    No full text
    International audienceAGuIX® (Activation-and-Guidance by Irradiation X) are sub-5nm theranostic nanoparticles composed of a polysiloxane matrix and gadolinium chelates. Developed by the Lyon University, they underwent extensive pre-clinical evaluation and were recently translated to clinical evaluation in the treatment of brain metastases and advanced cervical cancer. This presentation will summarize the principal in cellulo and pre-clinical results that led to their first-in-man administration.In collaboration with other teams, we did the proof of concept of their radiosensitizing efficacy in cellular (2D/3D cultures) and preclinical models of three radioresistant tumors: head and neck squamous cell carcinoma, metastatic melanoma and chondrosarcoma tumors (Miladi et al, Nanomedicine 2015; Kotb et al, Theranostics 2016; Ollier et al, in preparation). AGuIX® enter cancer cells by passive diffusion and macropinocytosis, localize in cytoplasm as free particle or entrapped in lysosomes, in close vicinity to the mitochondria (Rima et al, Biomaterials 2013). The interaction of radiation with gadolinium atoms produce a variety of secondary emissions, such as Auger shower, leading to the production of high reactive oxygen species levels that can trigger an intra‐mitochondrial stress and nuclear DNA damage leading to cell death (Simonet al., submitted). Relative biological efficiency (RBE) in cancer cells is quite comparable to that observed in response to carbon ions, suggesting the existence of common mechanisms through the amplification of the local dose (Wozny et al, Nanomedicine 2017). AGuIX®nanoparticles have already shown their efficiency as a radiosensitizer in at least 12 preclinical models of cancer (Lux et al, BJR 2018). Biodistribution studies after intravenous administration have shown passive uptake of the nanoparticles in tumors due to enhanced permeability and retention (EPR) effect combined with rapid renal elimination. We are now testing the next generation of AGuIX® nanoparticles containing other metals or functionalized with therapeutic or targeting moieties (D. Vernos’ poster).Supported by LabEx PRIME

    Hybrid core-shell particles for mRNA systemic delivery

    No full text
    International audiencemRNA based infectious disease vaccines have opened the venue for development of novel nucleic acids-based therapeutics. For all mRNA therapeutics dedicated delivery systems are required, where different functionalities and targeting abilities need to be optimized for the respective applications. One option for advanced formulations with tailored properties are lipid-polymer hybrid nanoparticles with complex nanostructure, which allow to combine features of several already well described nucleic acid delivery systems. Here, we explored hyaluronic acid (HA) as coating of liposome-mRNA complexes (LRCs) to investigate effects of the coating on surface charge, physicochemical characteristics and biological activity. HA was electrostatically attached to positively charged complexes, forming hybrid LRCs (HLRCs). At different N/P ratios, physico-chemical characterization of the two sets of particles showed similarity in size (around 200 nm) and mRNA binding abilities, while the presence of the HA shell conferred a negative surface charge to otherwise positive complexes. High transfection efficiency of LRCs and HLRCs in vitro has been obtained in THP-1 and human monocytes derived from PBMC, an interesting target cell population for cancer and immune related pathologies. In mice, quantitative biodistribution of radiolabeled LRC and HLRC particles, coupled with bioluminescence studies to detect the protein translation sites, hinted towards both particles' accumulation in the hepatic reticuloendothelial system (RES). mRNA translated proteins though was found mainly in the spleen, a major source for immune cells, with preference for expression in macrophages. The results showed that surface modifications of liposome-mRNA complexes can be used to fine-tune nanoparticle physico-chemical characteristics. This provides a tool for assembly of stable and optimized nanoparticles, which are prerequisite for future therapeutic interventions using mRNA-based nanomedicines

    From netrin‐1‐targeted <scp>SPECT</scp> / <scp>CT</scp> to internal radiotherapy for management of advanced solid tumors

    No full text
    International audienceTargeted radionuclide therapy is a revolutionary tool for the treatment of highly spread metastatic cancers. Most current approaches rely on the use of vectors to deliver radionuclides to tumor cells, targeting membrane-bound cancer-specific moieties. Here, we report the embryonic navigation cue netrin-1 as an unanticipated target for vectorized radiotherapy. While netrin-1, known to be reexpressed in tumoral cells to promote cancer progression, is usually characterized as a diffusible ligand, we demonstrate here that netrin-1 is actually poorly diffusible and bound to the extracellular matrix. A therapeutic anti-netrin-1 monoclonal antibody (NP137) has been preclinically developed and was tested in various clinical trials showing an excellent safety profile. In order to provide a companion test detecting netrin-1 in solid tumors and allowing the selection of therapy-eligible patients, we used the clinical-grade NP137 agent and developed an indium-111-NODAGA-NP137 single photon emission computed tomography (SPECT) contrast agent. NP137-111 In provided specific detection of netrin-1-positive tumors with an excellent signal-to-noise ratio using SPECT/CT imaging in different mouse models. The high specificity and strong affinity of NP137 paved the way for the generation of lutetium-177-DOTA-NP137, a novel vectorized radiotherapy, which specifically accumulated in netrin-1-positive tumors. We demonstrate here, using tumor cell-engrafted mouse models and a genetically engineered mouse model, that a single systemic injection of NP137-177 Lu provides important antitumor effects and prolonged mouse survival. Together, these data support the view that NP137-111 In and NP137-177 Lu may represent original and unexplored imaging and therapeutic tools against advanced solid cancers
    corecore