Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(21): e2220741120, 2023 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-37186838

RESUMO

Mammalian orthoreoviruses (reoviruses) serve as potential triggers of celiac disease and have oncolytic properties, making these viruses potential cancer therapeutics. Primary attachment of reovirus to host cells is mainly mediated by the trimeric viral protein, σ1, which engages cell-surface glycans, followed by high-affinity binding to junctional adhesion molecule-A (JAM-A). This multistep process is thought to be accompanied by major conformational changes in σ1, but direct evidence is lacking. By combining biophysical, molecular, and simulation approaches, we define how viral capsid protein mechanics influence virus-binding capacity and infectivity. Single-virus force spectroscopy experiments corroborated by in silico simulations show that GM2 increases the affinity of σ1 for JAM-A by providing a more stable contact interface. We demonstrate that conformational changes in σ1 that lead to an extended rigid conformation also significantly increase avidity for JAM-A. Although its associated lower flexibility impairs multivalent cell attachment, our findings suggest that diminished σ1 flexibility enhances infectivity, indicating that fine-tuning of σ1 conformational changes is required to successfully initiate infection. Understanding properties underlying the nanomechanics of viral attachment proteins offers perspectives in the development of antiviral drugs and improved oncolytic vectors.


Assuntos
Orthoreovirus , Reoviridae , Animais , Proteínas do Capsídeo/química , Reoviridae/metabolismo , Orthoreovirus/metabolismo , Proteínas Virais/metabolismo , Ligação Viral , Anticorpos Antivirais , Mamíferos/metabolismo
2.
Biomater Sci ; 11(3): 908-915, 2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36533676

RESUMO

Polymer-drug conjugates are widely investigated to enhance the selectivity of therapeutic drugs to cancer cells, as well as increase circulation lifetime and solubility of poorly soluble drugs. In order to direct these structures selectively to cancer cells, targeting agents are often conjugated to the nanoparticle surface as a strategy to limit drug accumulation in non-cancerous cells and therefore reduce systemic toxicity. Here, we report a simple procedure to generate biodegradable polycarbonate graft copolymer nanoparticles that allows for highly efficient conjugation and intracellular release of S-(+)-camptothecin, a topoisomerase I inhibitor widely used in cancer therapy. The drug-polymer conjugate showed strong efficacy in inhibiting cell proliferation across a range of cancer cell lines over non-cancerous phenotypes, as a consequence of the increased intracellular accumulation and subsequent drug release specifically in cancer cells. The enhanced drug delivery towards cancer cells in vitro demonstrates the potential of this platform for selective treatments without the addition of targeting ligands.


Assuntos
Nanopartículas , Neoplasias , Humanos , Sistemas de Liberação de Medicamentos , Cimento de Policarboxilato , Neoplasias/tratamento farmacológico , Polímeros/química , Nanopartículas/química , Concentração de Íons de Hidrogênio , Linhagem Celular Tumoral
3.
Mol Pharm ; 19(11): 4080-4097, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36069540

RESUMO

Nanomedicines show benefits in overcoming the limitations of conventional drug delivery systems by reducing side effects, toxicity, and exhibiting enhanced pharmacokinetic (PK) profiles to improve the therapeutic window of small-molecule drugs. However, upon administration, many nanoparticles (NPs) prompt induction of host innate immune responses, which in combination with other clearance pathways such as renal and hepatic, eliminate up to 99% of the administered dose. Here, we explore a drug predosing strategy to transiently suppress the mononuclear phagocyte system (MPS), subsequently improving the PK profile and biological behaviors exhibited by a model NP system [hyperbranched polymers (HBPs)] in an immunocompetent mouse model. In vitro assays allowed the identification of five drug candidates that attenuated cellular association. Predosing of lead compounds chloroquine (CQ) and zoledronic acid (ZA) further showed increased HBP retention within the circulatory system of mice, as shown by both fluorescence imaging and positron emission tomography-computed tomography. Flow cytometric evaluation of spleen and liver tissue cells following intravenous administration further demonstrated that CQ and ZA significantly reduced HBP association with myeloid cells by 23 and 16%, respectively. The results of this study support the use of CQ to pharmacologically suppress the MPS to improve NP PKs.


Assuntos
Produtos Biológicos , Nanopartículas , Animais , Camundongos , Nanopartículas/uso terapêutico , Nanomedicina , Sistemas de Liberação de Medicamentos/métodos , Macrófagos , Preparações Farmacêuticas , Cloroquina/farmacologia
4.
Chem Commun (Camb) ; 58(33): 5072-5087, 2022 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-35315846

RESUMO

Understanding biological interactions at a molecular level grants valuable information relevant to improving medical treatments and outcomes. Among the suite of technologies available, Atomic Force Microscopy (AFM) is unique in its ability to quantitatively probe forces and receptor-ligand interactions in real-time. The ability to assess the formation of supramolecular bonds and intermediates in real-time on surfaces and living cells generates important information relevant to understanding biological phenomena. Combining AFM with fluorescence-based techniques allows for an unprecedented level of insight not only concerning the formation and rupture of bonds, but understanding medically relevant interactions at a molecular level. As the ability of AFM to probe cells and more complex models improves, being able to assess binding kinetics, chemical topographies, and garner spectroscopic information will likely become key to developing further improvements in fields such as cancer, nanomaterials, and virology. The rapid response to the COVID-19 crisis, producing information regarding not just receptor affinities, but also strain-dependent efficacy of neutralizing nanobodies, demonstrates just how viable and integral to the pre-clinical development of information AFM techniques are in this era of medicine.


Assuntos
COVID-19 , Nanoestruturas , Humanos , Cinética , Ligantes , Microscopia de Força Atômica/métodos
5.
Antioxidants (Basel) ; 10(12)2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34943005

RESUMO

Human peroxiredoxin-5 (PRDX5) is a unique redox-sensitive protein that plays a dual role in brain ischemia-reperfusion injury. While intracellular PRDX5 has been reported to act as a neuroprotective antioxidative enzyme by scavenging peroxides, once released extracellularly from necrotic brain cells, the protein aggravates neural cell death by inducing expression of proinflammatory cytokines in macrophages through activation of Toll-like receptor (TLR) 2 (TLR2) and 4 (TLR4). Although recent evidence showed that PRDX5 was able to interact directly with TLR4, little is known regarding the role of the cysteine redox state of PRDX5 on its DAMP function. To gain insights into the role of PRDX5 redox-active cysteine residues in the TLR4-dependent proinflammatory activity of the protein, we used a recombinant human PRDX5 in the disulfide (oxidized) form and a mutant version lacking the peroxidatic cysteine, as well as chemically reduced and hyperoxidized PRDX5 proteins. We first analyzed the oxidation state and oligomerization profile by Western blot, mass spectrometry, and SEC-MALS. Using ELISA, we demonstrate that the disulfide bridge between the enzymatic cysteines is required to allow improved TLR4-dependent IL-8 secretion. Moreover, single-molecule force spectroscopy experiments revealed that TLR4 alone is not sufficient to discriminate the different PRDX5 redox forms. Finally, flow cytometry binding assays show that disulfide PRDX5 has a higher propensity to bind to the surface of living TLR4-expressing cells than the mutant protein. Taken together, these results demonstrate the importance of the redox state of PRDX5 cysteine residues on TLR4-induced inflammation.

6.
ACS Appl Bio Mater ; 4(3): 2675-2685, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35014306

RESUMO

Improving our understanding of how design choices in materials synthesis impact biological outcomes is of critical importance in the development of nanomedicines. Here, we show that fluorophore labeling of polymer nanomedicine candidates significantly alters their transport and cell association in multi-cellular tumor spheroids and their penetration in breast cancer xenografts, dependent on the type of the fluorophore and their positioning within the macromolecular structure. These data show the critical importance of the biomaterials structure and architecture in their tissue distribution and intracellular trafficking, which in turn govern their potential therapeutic efficacy. The broader implication of these findings suggests that when developing materials for medical applications, great care should be taken early on in the design process as relatively simple choices may have downstream impacts that could potentially skew preclinical biology data.


Assuntos
Materiais Biocompatíveis/química , Corantes Fluorescentes/química , Polímeros/química , Esferoides Celulares/química , Animais , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/farmacocinética , Células Cultivadas , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/farmacocinética , Humanos , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Teste de Materiais , Camundongos , Estrutura Molecular , Nanomedicina , Tamanho da Partícula , Polímeros/síntese química , Polímeros/farmacocinética , Distribuição Tecidual
7.
Chem Sci ; 11(12): 3268-3280, 2020 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-34122834

RESUMO

There remain several key challenges to existing therapeutic systems for cancer therapy, such as quantitatively determining the true, tissue-specific drug release profile in vivo, as well as reducing side-effects for an increased standard of care. Hence, it is crucial to engineer new materials that allow for a better understanding of the in vivo pharmacokinetic/pharmacodynamic behaviours of therapeutics. We have expanded on recent "click-to-release" bioorthogonal pro-drug activation of antibody-drug conjugates (ADCs) to develop a modular and controlled theranostic system for quantitatively assessing site-specific drug activation and deposition from a nanocarrier molecule, by employing defined chemistries. The exploitation of quantitative imaging using positron emission tomography (PET) together with pre-targeted bioorthogonal chemistries in our system provided an effective means to assess in real-time the exact amount of active drug administered at precise sites in the animal; our methodology introduces flexibility in both the targeting and therapeutic components that is specific to nanomedicines and offers unique advantages over other technologies. In this approach, the in vivo click reaction facilitates pro-drug activation as well as provides a quantitative means to investigate the dynamic behaviour of the therapeutic agent.

8.
Polymers (Basel) ; 11(9)2019 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-31480780

RESUMO

Nanomedicine has generated significant interest as an alternative to conventional cancertherapy due to the ability for nanoparticles to tune cargo release. However, while nanoparticletechnology has promised significant benefit, there are still limited examples of nanoparticles inclinical practice. The low translational success of nanoparticle research is due to the series ofbiological roadblocks that nanoparticles must migrate to be effective, including blood and plasmainteractions, clearance, extravasation, and tumor penetration, through to cellular targeting,internalization, and endosomal escape. It is important to consider these roadblocks holistically inorder to design more effective delivery systems. This perspective will discuss how nanoparticlescan be designed to migrate each of these biological challenges and thus improve nanoparticledelivery systems in the future. In this review, we have limited the literature discussed to studiesinvestigating the impact of polymer nanoparticle structure or composition on therapeutic deliveryand associated advancements. The focus of this review is to highlight the impact of nanoparticlecharacteristics on the interaction with different biological barriers. More specific studies/reviewshave been referenced where possible.

9.
Biomater Sci ; 7(11): 4661-4674, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31469127

RESUMO

As polymeric nanomedicines grow increasingly complex in design, an effective therapeutic release is often inherently tied to localisation to specific intracellular compartments or microenvironments. The inclusion of environmentally-sensitive moieties links the functionality of such materials to the trafficking behaviours exhibited once materials have obtained access to the cellular milieu. In order to perform their designed function, such materials often need to encounter specific biological cues or stimuli. As such, there is an increased need to improve our understanding of how the physicochemical properties of nanomaterials influence post-internalisation behaviours. Amongst the unknown factors that may contribute to the trafficking behaviours and distribution of polymers within the cellular environment, is the influence of the components selected in the development of such materials. To examine whether composition and arrangement of components within small polymeric nanomaterials contribute to their ability to navigate the intracellular space, here we utilise fluorophores to model component selection, varying the fluorescent handle selected and its method of incorporation. We explore the intracellular behaviours of well-characterised hyperbranched polymers in live MDA-MB-468 breast cancer cells in vitro. Changes in distribution as a function of both fluorophore selection and placement are reported, and our data suggest that the individual components used to produce potential nanomedicines are critical to their overall functioning and efficacy. Further to this, through the use of a novel non-conjugated targeting ligand, we demonstrate that there is inherent competition between component-directing factors and cellular influences on the ultimate fate of the polymers. The behaviours reported here suggest that not only does component selection contribute to intracellular processing, but these factors could potentially be harnessed when designing polymers to ensure improved functionality of future materials for therapeutic delivery.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Polietilenoglicóis/farmacocinética , Neoplasias da Mama/diagnóstico por imagem , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Feminino , Citometria de Fluxo , Humanos , Microscopia Confocal , Imagem Óptica , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Distribuição Tecidual , Células Tumorais Cultivadas
10.
Mol Pharm ; 14(12): 4485-4497, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29116801

RESUMO

Nanoscaled polymeric materials are increasingly being investigated as pharmaceutical products, drug/gene delivery vectors, or health-monitoring devices. Surface charge is one of the dominant parameters that regulates nanomaterial behavior in vivo. In this paper, we demonstrated how control over chemical synthesis allowed manipulation of nanoparticle surface charge, which in turn greatly influenced the in vivo behavior. Three methacrylate/methacrylamide-based monomers were used to synthesize well-defined hyperbranched polymers (HBP) by reversible addition-fragmentation chain transfer (RAFT) polymerization. Each HBP had a hydrodynamic diameter of approximately 5 nm as determined by dynamic light scattering (DLS) and transmission electron microscopy (TEM). Incorporation of a fluorescent moiety within the polymeric nanoparticles allowed determination of how charge affected the in vivo pharmacokinetic behavior of the nanomaterials and the biological response to them. A direct correlation between surface charge, cellular uptake, and cytotoxicity was observed, with cationic HBPs exhibiting higher cellular uptake and cytotoxicity than their neutral and anionic counterparts. Evaluation of the distribution of the differently charged HBPs within macrophages showed that all HBPs accumulated in the cytoplasm, but cationic HBPs also trafficked to, and accumulated within, the nucleus. Although cationic HBPs caused slight hemolysis, this was generally below accepted levels for in vivo safety. Analysis of pharmacokinetic behavior showed that cationic and anionic HBPs had short blood half-lives of 1.82 ± 0.51 and 2.34 ± 0.93 h respectively, compared with 5.99 ± 2.30 h for neutral HBPs. This was attributed to the fact that positively charged surfaces are more readily covered with opsonin proteins and thus more visible to phagocytic cells. This was supported by in vitro flow cytometric and qualitative live cell imaging studies, which showed that cationic HBPs tended to be taken up by macrophages more effectively and rapidly than neutral and anionic particles.


Assuntos
Cátions/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Nanopartículas/química , Polímeros/farmacologia , Animais , Cátions/química , Permeabilidade da Membrana Celular , Difusão Dinâmica da Luz , Citometria de Fluxo , Meia-Vida , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Masculino , Metacrilatos/química , Metacrilatos/farmacologia , Camundongos , Microscopia Eletrônica de Transmissão , Modelos Animais , Polimerização , Polímeros/química , Propriedades de Superfície
11.
Mol Pharm ; 14(10): 3539-3549, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28880092

RESUMO

Theranostics is a strategy that combines multiple functions such as targeting, stimulus-responsive drug release, and diagnostic imaging into a single platform, often with the aim of developing personalized medicine.1,2 Based on this concept, several well-established hyperbranched polymeric theranostic nanoparticles were synthesized and characterized as model nanomedicines to investigate how their properties affect the distribution of loaded drugs at both the cell and whole animal levels. An 8-mer peptide aptamer was covalently bound to the periphery of the nanoparticles to achieve both targeting and potential chemosensitization functionality against heat shock protein 70 (Hsp70). Doxorubicin was also bound to the polymeric carrier as a model chemotherapeutic drug through a degradable hydrazone bond, enabling pH-controlled release under the mildly acid conditions that are found in the intracellular compartments of tumor cells. In order to track the nanoparticles, cyanine-5 (Cy5) was incorporated into the polymer as an optical imaging agent. In vitro cellular uptake was assessed for the hyperbranched polymer containing both doxorubicin (DOX) and Hsp70 targeted peptide aptamer in live MDA-MB-468 cells, and was found to be greater than that of either the untargeted, DOX-loaded polymer or polymer alone due to the specific affinity of the peptide aptamer for the breast cancer cells. This was also validated in vivo with the targeted polymers showing much higher accumulation within the tumor 48 h postinjection than the untargeted analogue. More detailed assessment of the nanomedicine distribution was achieved by directly following the polymeric carrier and the doxorubicin at both the in vitro cellular level via compartmental analysis of confocal images of live cells and in whole tumors ex vivo using confocal imaging to visualize the distribution of the drug in tumor tissue as a function of distance from blood vessels. Our results indicate that this polymeric carrier shows promise as a cancer theranostic, demonstrating active targeting to tumor cells with the capability for simultaneous drug release.


Assuntos
Antineoplásicos/farmacocinética , Aptâmeros de Peptídeos/química , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Nanomedicina Teranóstica/métodos , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Feminino , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Químicos , Nanopartículas/química , Polímeros/química , Medicina de Precisão/métodos , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Biomaterials ; 141: 330-339, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28711780

RESUMO

The therapeutic potential of hyperbranched polymers targeted to prostate cancer and loaded with doxorubicin was investigated. Polyethylene glycol hyperbranched polymers were synthesised via RAFT polymerisation to feature glutamate urea targeting ligands for PSMA on the periphery. The chemotherapeutic, doxorubicin, was attached to the hyperbranched polymers through hydrazone formation, which allowed controlled release of the drug from the polymers in vitro endosomal conditions, with 90% release of the drug over 36 h. The polymers were able to target to PSMA-expressing prostate cancer cells in vitro, and demonstrated comparable cytotoxicity to free doxorubicin. The ability of the hyperbranched polymers to specifically facilitate transport of loaded doxorubicin into the cells was confirmed using live cell confocal imaging, which demonstrated that the drug was able to travel with the polymer into cells by receptor mediated internalisation, and subsequently be released into the nucleus following hydrazone degradation. Finally, the ability of the complex to induce a therapeutic effect on prostate cancer cells was investigated through a long term tumour regression study, which confirmed that the DOX-loaded polymers were able to significantly reduce the volume of subcutaneous prostate tumours in vivo in comparison to free doxorubicin and a polymer control, with no adverse toxicity to the animals. This work therefore demonstrates the potential of a hyperbranched polymer system to be utilised for prostate cancer theranostics.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Antígenos de Superfície/metabolismo , Preparações de Ação Retardada/metabolismo , Doxorrubicina/administração & dosagem , Glutamato Carboxipeptidase II/metabolismo , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/tratamento farmacológico , Animais , Antibióticos Antineoplásicos/uso terapêutico , Antígenos de Superfície/análise , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Glutamato Carboxipeptidase II/análise , Humanos , Masculino , Camundongos , Microscopia Confocal/métodos , Imagem Óptica/métodos , Polímeros/metabolismo , Próstata/diagnóstico por imagem , Próstata/efeitos dos fármacos , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Nanomedicina Teranóstica/métodos
13.
Adv Healthc Mater ; 5(16): 2055-68, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27283923

RESUMO

Targeted nanomaterials promise improved therapeutic efficacy, however their application in nanomedicine is limited due to complexities associated with protein conjugations to synthetic nanocarriers. A facile method to generate actively targeted nanomaterials is developed and exemplified using polyethylene glycol (PEG)-functional nanostructures coupled to a bispecific antibody (BsAb) with dual specificity for methoxy PEG (mPEG) epitopes and cancer targets such as epidermal growth factor receptor (EGFR). The EGFR-mPEG BsAb binds with high affinity to recombinant EGFR (KD : 1 × 10(-9) m) and hyperbranched polymer (HBP) consisting of mPEG (KD : 10 × 10(-9) m) and demonstrates higher avidity for HBP compared to linear mPEG. The binding of BsAb-HBP bioconjugate to EGFR on MDA-MB-468 cancer cells is investigated in vitro using a fluorescently labeled polymer, and in in vivo xenograft models by small animal optical imaging. The antibody-targeted nanostructures show improved accumulation in tumor cells compared to non-targeted nanomaterials. This demonstrates a facile approach for tuning targeting ligand density on nanomaterials, by modulating surface functionality. Antibody fragments are tethered to the nanomaterial through simple mixing prior to administration to animals, overcoming the extensive procedures encountered for developing targeted nanomedicines.


Assuntos
Anticorpos Biespecíficos , Anticorpos Antineoplásicos , Sistemas de Liberação de Medicamentos/métodos , Nanoestruturas , Neoplasias/tratamento farmacológico , Polietilenoglicóis , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/farmacologia , Anticorpos Antineoplásicos/química , Anticorpos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Neoplasias/metabolismo , Neoplasias/patologia , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Nanomedicina Teranóstica/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA