Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Biol Interact ; 356: 109882, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35263611

ABSTRACT

Increasing use of nanomaterials in everyday products such as cosmetics, medicines and food packaging is of grave concern given the lack of understanding with regards the impact such materials have on biological systems. The aim of this study is to examine cell death induced by cationic amorphous silica nanoparticles and determine the involvement of lysosomal cysteine proteases in this process. We report that multiple forms of cell death including apoptosis and pyroptosis are elicited following exposure to amorphous silica nanoparticles and that lysosomal cysteine proteases are involved in both cell death pathways in macrophages. Interestingly, lysosomal cysteine protease mRNA expression and release into the extracellular environment is induced following exposure to amorphous silica nanoparticles. Previously, the determination of nanoparticle-induced toxicity has focused on cytokine readouts, but the work presented here demonstrates that changes to normal protease biology should also be considered when evaluating the molecular mechanisms by which nanoparticulate matter causes cellular inflammation and death.


Subject(s)
Cysteine Proteases , Nanoparticles , Cell Death , Cysteine Proteases/metabolism , Lysosomes , Macrophages/metabolism , Nanoparticles/toxicity , Silicon Dioxide/metabolism , Silicon Dioxide/toxicity
2.
Cancers (Basel) ; 13(24)2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34944794

ABSTRACT

The development of drug resistance remains one of the greatest clinical oncology challenges that can radically dampen the prospect of achieving complete and durable tumour control. Efforts to mitigate drug resistance are therefore of utmost importance, and nanotechnology is rapidly emerging for its potential to overcome such issues. Studies have showcased the ability of nanomedicines to bypass drug efflux pumps, counteract immune suppression, serve as radioenhancers, correct metabolic disturbances and elicit numerous other effects that collectively alleviate various mechanisms of tumour resistance. Much of this progress can be attributed to the remarkable benefits that nanoparticles offer as drug delivery vehicles, such as improvements in pharmacokinetics, protection against degradation and spatiotemporally controlled release kinetics. These attributes provide scope for precision targeting of drugs to tumours that can enhance sensitivity to treatment and have formed the basis for the successful clinical translation of multiple nanoformulations to date. In this review, we focus on the longstanding reputation of pancreatic cancer as one of the most difficult-to-treat malignancies where resistance plays a dominant role in therapy failure. We outline the mechanisms that contribute to the treatment-refractory nature of these tumours, and how they may be effectively addressed by harnessing the unique capabilities of nanomedicines. Moreover, we include a brief perspective on the likely future direction of nanotechnology in pancreatic cancer, discussing how efforts to develop multidrug formulations will guide the field further towards a therapeutic solution for these highly intractable tumours.

3.
J Control Release ; 324: 610-619, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32504778

ABSTRACT

Pancreatic cancer is usually advanced and drug resistant at diagnosis. A potential therapeutic approach outlined here uses nanoparticle (NP)-based drug carriers, which have unique properties that enhance intra-tumor drug exposure and reduce systemic toxicity of encapsulated drugs. Here we report that patients whose pancreatic cancers express elevated levels of Death Receptor 5 (DR5) and its downstream regulators/effectors FLIP, Caspase-8, and FADD had particularly poor prognoses. To take advantage of elevated expression of this pathway, we designed drug-loaded NPs with a surface-conjugated αDR5 antibody (AMG 655). Binding and clustering of the DR5 is a prerequisite for efficient apoptosis initiation, and the αDR5-NPs were indeed found to activate apoptosis in multiple pancreatic cancer models, whereas the free antibody did not. The extent of apoptosis induced by αDR5-NPs was enhanced by down-regulating FLIP, a key modulator of death receptor-mediated activation of caspase-8. Moreover, the DNA topoisomerase-1 inhibitor camptothecin (CPT) down-regulated FLIP in pancreatic cancer models and enhanced apoptosis induced by αDR5-NPs. CPT-loaded αDR5-NPs significantly increased apoptosis and decreased cell viability in vitro in a caspase-8- and FADD-dependent manner consistent with their expected mechanism-of-action. Importantly, CPT-loaded αDR5-NPs markedly reduced tumor growth rates in vivo in established pancreatic tumor models, inducing regressions in one model. These proof-of-concept studies indicate that αDR5-NPs loaded with agents that downregulate or inhibit FLIP are promising candidate agents for the treatment of pancreatic cancer.


Subject(s)
Nanoparticles , Pancreatic Neoplasms , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein , Cell Line, Tumor , Drug Carriers , Humans , Pancreatic Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
4.
Nanoscale ; 11(42): 20261-20273, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31626255

ABSTRACT

The anti-Epidermal Growth Factor Receptor (EGFR) antibody Cetuximab (CTX) has demonstrated limited anti-cancer efficacy in cells overexpressing EGFR due to activating mutations in RAS in solid tumours, such as pancreatic cancer. The utilisation of antibodies as targeting components of antibody-drug conjugates, such as trastuzumab emtansine (Kadcyla), demonstrates that antibodies may be repurposed to direct therapeutic agents to antibody-resistant cancers. Here we investigated the use of CTX as a targeting agent for camptothecin (CPT)-loaded polymeric nanoparticles (NPs) directed against KRAS mutant CTX-resistant cancer cells. CPT was encapsulated within poly(lactic-co-glycolic acid) (PLGA) NPs using the solvent evaporation method. CTX conjugation improved NP binding and delivery of CPT to CTX-resistant cancer cell lines. CTX successfully targeted CPT-loaded NPs to mutant KRAS PANC-1 tumours in vivo and reduced tumour growth. This study highlights that CTX can be repurposed as a targeting agent against CTX-resistant cancers and that antibody repositioning may be applicable to other antibodies restricted by resistance.


Subject(s)
Ado-Trastuzumab Emtansine , Cetuximab , Drug Resistance, Neoplasm/drug effects , Immunoconjugates , Nanoparticles , Neoplasm Proteins/metabolism , Ado-Trastuzumab Emtansine/chemistry , Ado-Trastuzumab Emtansine/pharmacology , Animals , Cetuximab/chemistry , Cetuximab/pharmacology , ErbB Receptors/metabolism , Female , HCT116 Cells , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Mice , Mice, SCID , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
Nanoscale ; 11(2): 742-751, 2019 Jan 03.
Article in English | MEDLINE | ID: mdl-30566168

ABSTRACT

The ability to label active caspase-3 represents a useful pharmacodynamic strategy to determine the efficacy of anti-tumour drugs. Activity-based probes (ABPs) provide a method for the labelling of activated caspases and the recent development of hybrid combinatorial substrate libraries (HyCoSuL) has allowed for the generation of highly selective ABPs to discriminately label these proteases. Here using this approach, a novel caspase-3 selective ABP (CS1) has been developed and validated in apoptotic cells to selectively bind caspase-3 over the closely related caspase-7. However, a critical bottleneck for ABPs is their cell penetrance and therefore this cell-impermeable CS1 probe was subsequently formulated into PLGA-based nanoparticles (CS1-NPs). We demonstrate the ability of these particles to be taken up by the cells and facilitate intracellular delivery of the ABP to effectively label caspase 3 in response to apoptotic stimuli. This work forms the foundation of a novel approach for the labelling of caspase 3 and may have downstream utility to measure real time apoptosis in tumours and other organs.


Subject(s)
Caspase 3/metabolism , Nanoparticles/chemistry , Nanoparticles/metabolism , Peptides/chemistry , Apoptosis , Caspase 3/analysis , Cell Line, Tumor , Combinatorial Chemistry Techniques , Cytoplasm/metabolism , Humans , Molecular Structure , Nanoparticles/ultrastructure , Peptide Library , Peptides/metabolism , Substrate Specificity
6.
Drug Discov Today Technol ; 30: 63-69, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30553522

ABSTRACT

Antibody conjugated nanoparticles (ACNPs) represent a novel strategy for the development of therapies exploiting antibodies to augment the delivery of chemotherapy payloads. Following in the footsteps of the success of antibody drug conjugates (ADCs), ACNPs are only now reaching clinical evaluation. In this review we discuss the success of ADCs and explore the opportunities ACNPs offer, such as broad chemotherapy payload selection, high drug to antibody ratios and the ability to finely tailor drug release in comparison to ADCs. The ability of ACNPs to elicit increased avidity due to multivalent effects and the potential to use these modular platforms in immunotherapeutic approaches is also explored. Through addressing challenges that still remain in bringing these complex formulations to the clinic, ACNPs hold obvious potential for the treatment of a wide range of cancers and other diseases where selective targeting of drug agents is essential.


Subject(s)
Antineoplastic Agents/chemistry , Immunoconjugates/chemistry , Humans , Nanoparticles , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...