Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Adv Healthc Mater ; : e2401306, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39031098

RÉSUMÉ

Cancer immunotherapy has emerged as a promising approach for the induction of an antitumor response. While immunotherapy response rates are very high in some cancers, the efficacy against solid tumors remains limited caused by the presence of an immunosuppressive tumor microenvironment. Induction of immunogenic cell death (ICD) in the tumor can be used to boost immunotherapy response in solid cancers by eliciting the release of immune-stimulatory components. However, the delivery of components inducing ICD to tumor sites remains a challenge. Here, a novel delivery method is described for antitumor therapy based on MLKL (Mixed Lineage Kinase Domain-Like), a key mediator of necroptosis and inducer of ICD. A novel highly branched poly (ß-amino ester)s (HPAEs) system is designed to efficiently deliver MLKL plasmid DNA to the tumor with consequent enhancement of immune antigen presentation for T cell responses in vitro, and improved antitumor response and prolonged survival in tumor-bearing mice. Combination of the therapy with anti-PD-1 treatment revealed significant changes in the composition of the tumor microenvironment, including increased infiltration of CD8+ T cells and tumor-associated lymphocytes. Overall, the HPAEs delivery system can enhance MLKL-based cancer immunotherapy and promote antitumor immune responses, providing a potential treatment to boost cancer immunotherapies.

2.
ACS Appl Mater Interfaces ; 16(1): 66-83, 2024 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-38163254

RÉSUMÉ

Over the past decades, the medical exploitation of nanotechnology has been largely increasing and finding its way into translational research and clinical applications. Despite their biomedical potential, uncertainties persist regarding the intricate role that nanomaterials may play on altering physiology in healthy and diseased tissues. Extracellular vesicles (EVs) are recognized as an important pathway for intercellular communication and known to be mediators of cellular stress. EVs are currently explored for targeted delivery of therapeutic agents, including nanoformulations, to treat and diagnose cancer or other diseases. Here, we aimed to investigate whether nanomaterials could have a possible impact on EV functionality, their safety, and whether EVs can play a role in nanomaterial toxicity profiles. To evaluate this, the impact of inorganic nanomaterial administration on EVs derived from murine melanoma and human breast cancer cells was tested. Cells were incubated with subtoxic concentrations of 4 different biomedically relevant inorganic nanoparticles (NPs): gold, silver, silicon dioxide, or iron oxide. The results displayed a clear NP and cell-type-dependent effect on increasing or decreasing EV secretion. Furthermore, the expression pattern of several EV-derived miRNAs was significantly changed upon NP exposure, compared to nontreated cells. Detailed pathway analysis and additional studies confirmed that EVs obtained from NP-exposed cells could influence immunological responses and cellular physiology. Together, these data reveal that NPs can have wide-ranging effects which can result in toxicity concerns or enhanced therapeutic potential as a secondary enhanced effect mediated and enhanced by EVs.


Sujet(s)
Vésicules extracellulaires , microARN , Nanoparticules , Tumeurs , Humains , Souris , Animaux , Vésicules extracellulaires/métabolisme , Tumeurs/traitement médicamenteux , microARN/métabolisme , Communication cellulaire
3.
Adv Healthc Mater ; 12(24): e2300594, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37247322

RÉSUMÉ

The ability to improve nanoparticle delivery to solid tumors is an actively studied domain, where various mechanisms are looked into. In previous work, the authors have looked into nanoparticle size, tumor vessel normalization, and disintegration, and here it is aimed to continue this work by performing an in-depth mechanistic study on the use of ciRGD peptide co-administration. Using a multiparametric approach, it is observed that ciRGD can improve nanoparticle delivery to the tumor itself, but also to tumor cells specifically better than vessel normalization strategies. The effect depends on the level of tumor perfusion, hypoxia, neutrophil levels, and vessel permeability. This work shows that upon characterizing tumors for these parameters, conditions can be selected that can optimally benefit from ciRGD co-administration as a means to improve NP delivery to solid tumors.


Sujet(s)
Nanoparticules , Tumeurs , Humains , Neuropiline 1/usage thérapeutique , Granulocytes neutrophiles , Systèmes de délivrance de médicaments , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Nanoparticules/composition chimique , Hypoxie
5.
J Nanobiotechnology ; 21(1): 87, 2023 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-36915084

RÉSUMÉ

Nanoparticle-mediated cancer immunotherapy holds great promise, but more efforts are needed to obtain nanoformulations that result in a full scale activation of innate and adaptive immune components that specifically target the tumors. We generated a series of copper-doped TiO2 nanoparticles in order to tune the kinetics and full extent of Cu2+ ion release from the remnant TiO2 nanocrystals. Fine-tuning nanoparticle properties resulted in a formulation of 33% Cu-doped TiO2 which enabled short-lived hyperactivation of dendritic cells and hereby promoted immunotherapy. The nanoparticles result in highly efficient activation of dendritic cells ex vivo, which upon transplantation in tumor bearing mice, exceeded the therapeutic outcomes obtained with classically stimulated dendritic cells. Efficacious but simple nanomaterials that can promote dendritic cancer cell vaccination strategies open up new avenues for improved immunotherapy and human health.


Sujet(s)
Vaccins anticancéreux , Nanoparticules , Tumeurs , Vaccins , Animaux , Souris , Humains , Tumeurs/traitement médicamenteux , Nanoparticules/composition chimique , Immunothérapie/méthodes , Cellules dendritiques , Vaccins anticancéreux/usage thérapeutique
6.
EMBO Rep ; 24(3): e56310, 2023 03 06.
Article de Anglais | MEDLINE | ID: mdl-36597777

RÉSUMÉ

Macrophages undergo plasma membrane fusion and cell multinucleation to form multinucleated giant cells (MGCs) such as osteoclasts in bone, Langhans giant cells (LGCs) as part of granulomas or foreign-body giant cells (FBGCs) in reaction to exogenous material. How multinucleation per se contributes to functional specialization of mature mononuclear macrophages remains poorly understood in humans. Here, we integrate comparative transcriptomics with functional assays in purified mature mononuclear and multinucleated human osteoclasts, LGCs and FBGCs. Strikingly, in all three types of MGCs, multinucleation causes a pronounced downregulation of macrophage identity. We show enhanced lysosome-mediated intracellular iron homeostasis promoting MGC formation. The transition from mononuclear to multinuclear state is accompanied by cell specialization specific to each polykaryon. Enhanced phagocytic and mitochondrial function associate with FBGCs and osteoclasts, respectively. Moreover, human LGCs preferentially express B7-H3 (CD276) and can form granuloma-like clusters in vitro, suggesting that their multinucleation potentiates T cell activation. These findings demonstrate how cell-cell fusion and multinucleation reset human macrophage identity as part of an advanced maturation step that confers MGC-specific functionality.


Sujet(s)
Macrophages , Ostéoclastes , Humains , Macrophages/métabolisme , Ostéoclastes/métabolisme , Os et tissu osseux , Cellules géantes , Antigènes B7/métabolisme
7.
J Nanobiotechnology ; 20(1): 518, 2022 Dec 09.
Article de Anglais | MEDLINE | ID: mdl-36494816

RÉSUMÉ

Nanoparticle (NP) delivery to solid tumors remains an actively studied field, where several recent studies have shed new insights into the underlying mechanisms and the still overall poor efficacy. In the present study, Au NPs of different sizes were used as model systems to address this topic, where delivery of the systemically administered NPs to the tumor as a whole or to tumor cells specifically was examined in view of a broad range of tumor-associated parameters. Using non-invasive imaging combined with histology, immunohistochemistry, single-cell spatial RNA expression and image-based single cell cytometry revealed a size-dependent complex interaction of multiple parameters that promoted tumor and tumor-cell specific NP delivery. Interestingly, the data show that most NPs are sequestered by tumor-associated macrophages and cancer-associated fibroblasts, while only few NPs reach the actual tumor cells. While perfusion is important, leaky blood vessels were found not to promote NP delivery, but rather that delivery efficacy correlated with the maturity level of tumor-associated blood vessels. In line with recent studies, we found that the presence of specialized endothelial cells, expressing high levels of CD276 and Plvap promoted both tumor delivery and tumor cell-specific delivery of NPs. This study identifies several parameters that can be used to determine the suitability of NP delivery to the tumor region or to tumor cells specifically, and enables personalized approaches for maximal delivery of nanoformulations to the targeted tumor.


Sujet(s)
Nanoparticules métalliques , Nanoparticules , Tumeurs , Humains , Microenvironnement tumoral , Taille de particule , Or/métabolisme , Cellules endothéliales/métabolisme , Tumeurs/métabolisme , Systèmes de délivrance de médicaments/méthodes , Lignée cellulaire tumorale , Antigènes B7/métabolisme
8.
Chem Rev ; 121(3): 1746-1803, 2021 02 10.
Article de Anglais | MEDLINE | ID: mdl-33445874

RÉSUMÉ

Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.


Sujet(s)
Systèmes de délivrance de médicaments , Nanoparticules/composition chimique , Tumeurs/composition chimique , Animaux , Humains , Tumeurs/imagerie diagnostique , Peptides/composition chimique , Microenvironnement tumoral
9.
Bioconjug Chem ; 31(4): 1077-1087, 2020 04 15.
Article de Anglais | MEDLINE | ID: mdl-32208650

RÉSUMÉ

Despite the progress in nanotechnology for biomedical applications, great efforts are still being employed in optimizing nanoparticle (NP) design parameters to improve functionality and minimize bionanotoxicity. In this study, we developed CdSe/CdS/ZnS core/shell/shell quantum dots (QDs) that are compact ligand-coated and surface-functionalized with an HIV-1-derived TAT cell-penetrating peptide (CPP) analog to improve both biocompatibility and cellular uptake. Multiparametric studies were performed in different mammalian and murine cell lines to compare the effects of varying QD size and number of surface CPPs on cellular uptake, viability, generation of reactive oxygen species, mitochondrial health, cell area, and autophagy. Our results showed that the number of cell-associated NPs and their respective toxicity are higher for the larger QDs. Meanwhile, increasing the number of surface CPPs also enhanced cellular uptake and induced cytotoxicity through the generation of mitoROS and autophagy. Thus, here we report the optimal size and surface CPP combinations for improved QD cellular uptake.


Sujet(s)
Peptides de pénétration cellulaire/composition chimique , Peptides de pénétration cellulaire/métabolisme , Taille de particule , Boîtes quantiques/composition chimique , Boîtes quantiques/toxicité , Animaux , Autophagie/effets des médicaments et des substances chimiques , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Test de matériaux , Souris , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Transport des protéines , Espèces réactives de l'oxygène/métabolisme , Propriétés de surface , Produits du gène tat du virus de l'immunodéficience humaine/composition chimique
10.
Nanoscale Adv ; 2(9): 3734-3763, 2020 Sep 16.
Article de Anglais | MEDLINE | ID: mdl-36132767

RÉSUMÉ

Nanomaterials are currently widely exploited for their potential in the development of novel cancer therapies, and so far, mainly nanoparticles (NPs) consisting of liposomes and polymers have made their way into the clinic. However, major bottlenecks for the clinical translation of other types of NPs (i.e. inorganic) are the lack of knowledge concerning their long-term distribution in vivo and their potential toxicity. To counter this, various research groups have worked on soluble NPs, such as zinc oxide (ZnO), copper oxide (CuO), and silver (Ag), which tend to dissolve spontaneously into their ionic form, releasing toxic metal ions and leading to reactive oxygen species (ROS) generation when exposed to cellular environments. By fine-tuning the dissolution kinetics of these NPs, it is possible to control the level of ROS production and thus cytotoxicity to selectively destroy tumor tissue. Specifically, cancer cells tend to exhibit a higher basal level of oxidative stress compared to normal cells due to their higher metabolic rates, and therefore, by engineering NPs that generate sufficient ROS that barely exceed toxic thresholds in cancer cells, normal cells will only experience reversible transient damage. This review focuses on the use of these soluble inorganic NPs for selective cancer therapy and on the various in vitro and in vivo studies that have aimed to control the dissolution kinetics of these NPs, either through particle doping or surface modifications.

11.
ACS Appl Mater Interfaces ; 11(18): 16336-16346, 2019 May 08.
Article de Anglais | MEDLINE | ID: mdl-30986026

RÉSUMÉ

Gold nanoparticles (AuNPs) have emerged as promising drug delivery candidates that can be leveraged for cancer therapy. Lung cancer (LC) is a heterogeneous disease that imposes a significant burden on society, with an unmet need for new therapies. Chemotherapeutic drugs such as afatinib (Afb), which is clinically approved for the treatment of epidermal growth factor receptor positive LC, is hydrophobic and has low bioavailability leading to spread around the body, causing severe side effects. Herein, we present a novel afatinib-AuNP formulation termed Afb-AuNPs, with the aim of improving drug efficacy and biocompatibility. This was achieved by synthesis of an alkyne-bearing Afb derivative and reaction with azide-functionalized lipoic acid using copper-catalyzed click chemistry, then conjugation to AuNPs via alkylthiol-gold bond formation. The Afb-AuNPs were found to possess up to 3.7-fold increased potency when administered to LC cells in vitro and were capable of significantly inhibiting cancer cell proliferation, as assessed by MTT assay and electric cell-substrate impedance sensing, respectively. Furthermore, when exposed to Afb-AuNPs, human alveolar epithelial type I-like cells, a model of the healthy lung epithelium, maintained viability and were found to release less proinflammatory cytokines when compared to free drug, demonstrating the biocompatibility of our formulation. This study provides a new platform for the development of nontraditional AuNP conjugates which can be applied to other molecules of therapeutic or diagnostic utility, with potential to be combined with photothermal therapy in other cancers.


Sujet(s)
Afatinib/composition chimique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Nanoconjugués/administration et posologie , Inhibiteurs de protéines kinases/administration et posologie , Afatinib/administration et posologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments , Humains , Test de matériaux , Nanoparticules métalliques/administration et posologie , Nanoparticules métalliques/composition chimique , Nanoconjugués/composition chimique , Polyéthylène glycols/composition chimique , Inhibiteurs de protéines kinases/composition chimique , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/composition chimique
12.
Arch Biochem Biophys ; 613: 31-42, 2017 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-27818203

RÉSUMÉ

Prion diseases are associated with conversion of cellular prion protein (PrPC) into an abnormally folded and infectious scrapie isoform (PrPSc). We previously showed that peptides derived from the unprocessed N-termini of mouse and bovine prion proteins, mPrP1-28 and bPrP1-30, function as cell-penetrating peptides (CPPs), and destabilize model membrane systems, which could explain the infectivity and toxicity of prion diseases. However, subsequent studies revealed that treatment with mPrP1-28 or bPrP1-30 significantly reduce PrPSc levels in prion-infected cells. To explain these seemingly contradictory results, we correlated the aggregation, membrane perturbation and cytotoxicity of the peptides with their cellular uptake and intracellular localization. Although the peptides have a similar primary sequence, mPrP1-28 is amyloidogenic, whereas bPrP1-30 forms smaller oligomeric or non-fibrillar aggregates. Surprisingly, bPrP1-30 induces much higher cytotoxicity than mPrP1-28, indicating that amyloid formation and toxicity are independent. The toxicity is correlated with prolonged residence at the plasma membrane and membrane perturbation. Both ordered aggregation and toxicity of the peptides are inhibited by low pH. Under non-toxic conditions, the peptides are internalized by lipid-raft dependent macropinocytosis and localize to acidic lysosomal compartments. Our results shed light on the antiprion mechanism of the prion protein-derived CPPs and identify a potential site for PrPSc formation.


Sujet(s)
Amyloïde/composition chimique , Peptides de pénétration cellulaire/composition chimique , Protéines prion/composition chimique , Signaux de triage des protéines , Animaux , Sites de fixation , Bovins , Lignée cellulaire tumorale , Membrane cellulaire/composition chimique , Membrane cellulaire/métabolisme , Survie cellulaire , Endosomes/composition chimique , Endosomes/métabolisme , Cellules HeLa , Humains , Concentration en ions d'hydrogène , Cinétique , Liposomes/composition chimique , Lysosomes/composition chimique , Souris , Microscopie électronique à transmission , Peptides/composition chimique , Domaines protéiques , Température
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE