Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
Plus de filtres










Gamme d'année
1.
ACS Appl Mater Interfaces ; 15(50): 58241-58250, 2023 Dec 20.
Article de Anglais | MEDLINE | ID: mdl-38059477

RÉSUMÉ

Gold nanomaterials have been shown to augment radiation therapy both in vitro and in vivo. However, studies on these materials are mostly phenomenological due to nanoparticle heterogeneity and the complexity of biological systems. Even accurate quantification of the particle dose still results in bulk average biases; the effect on individual cells is not measured but rather the effect on the overall population. To perform quantitative nanobiology, we coated glass coverslips uniformly at varying densities with Au nanoparticle preparations with different morphologies (45 nm cages, 25 nm spheres, and 30 nm rods). Consequently, the effect of a specific number of particles per unit area in contact with breast cancer cells growing on the coated surfaces was ascertained. Gold nanocages showed the highest degree of radiosensitization on a per particle basis, followed by gold nanospheres and gold nanorods, respectively. All three materials showed little cytotoxic effect at 0 Gy, but clonogenic survival decreased proportionally with the radiation dose and particle coverage density. A similar trend was seen in vivo in the combined treatment antitumor response in 4T1 tumor-bearing animals. The presence of gold affected the type and quantity of reactive oxygen species generated, specifically superoxide and hydroxyl radicals, and the concentration of nanocages correlated with the development of more numerous double-stranded DNA breaks and increased protein oxidation as measured by carbonylation. This work demonstrates the dependence on morphology and concentration of radiation enhancement by gold nanomaterials and may lead to a novel method to differentiate intra- and extracellular functionalities of gold nanomedicine treatment strategies. It further provides insights that can guide the rational development of gold nanomaterial-based radiosensitizers for clinical use.


Sujet(s)
Nanoparticules métalliques , Nanostructures , Radiosensibilisants , Animaux , Or/pharmacologie , Or/métabolisme , Apoptose , Radiosensibilisants/pharmacologie
2.
J Nanobiotechnology ; 21(1): 378, 2023 Oct 17.
Article de Anglais | MEDLINE | ID: mdl-37848956

RÉSUMÉ

BACKGROUND: The combination of drug delivery with immune checkpoint targeting has been extensively studied in cancer therapy. However, the clinical benefit for patients from this strategy is still limited. B7 homolog 3 protein (B7-H3), also known as CD276 (B7-H3/CD276), is a promising therapeutic target for anti-cancer treatment. It is widely overexpressed on the surface of malignant cells and tumor vasculature, and its overexpression is associated with poor prognosis. Herein, we report B7H3 targeting doxorubicin (Dox)-conjugated gold nanocages (B7H3/Dox@GNCs) with pH-responsive drug release as a selective, precise, and synergistic chemotherapy-photothermal therapy agent against non-small-cell lung cancer (NSCLC). RESULTS: In vitro, B7H3/Dox@GNCs exhibited a responsive release of Dox in the tumor acidic microenvironment. We also demonstrated enhanced intracellular uptake, induced cell cycle arrest, and increased apoptosis in B7H3 overexpressing NSCLC cells. In xenograft tumor models, B7H3/Dox@GNCs exhibited tumor tissue targeting and sustained drug release in response to the acidic environment. Wherein they synchronously destroyed B7H3 positive tumor cells, tumor-associated vasculature, and stromal fibroblasts. CONCLUSION: This study presents a dual-compartment targeted B7H3 multifunctional gold conjugate system that can precisely control Dox exposure in a spatio-temporal manner without evident toxicity and suggests a general strategy for synergistic therapy against NSCLC.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Doxorubicine , Tumeurs du poumon , Nanoparticules , Thérapie photothermique , Humains , Antigènes B7 , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Lignée cellulaire tumorale , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Libération de médicament , Or , Concentration en ions d'hydrogène , Hyperthermie provoquée , Tumeurs du poumon/traitement médicamenteux , Photothérapie , Thérapie photothermique/méthodes , Microenvironnement tumoral , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Animaux , Souris , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Periodontol 2000 ; 2023 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-37614160

RÉSUMÉ

Gold nanocages (AuNCs) have been invented and developed over two decades as biomaterial in clinical medicine with great application potential. AuNCs have a characteristic structure of porous walls with hollow interior and a compact size. This makes it possible for them to transport biomolecules or drugs with the advantages of their photothermal effects that could help further destroy germs or tumors while also regulating the release of drugs inside. Furthermore, their bioactivity and application can be broadened by using cell-membrane display technology. AuNCs have shown tremendous potential in antibacterial activity, inflammation modulation, and tissue regeneration, which is required in periodontitis and peri-implantitis treatment. Thus, this article provides an overview of AuNCs synthesis, characteristics, surface modifications, and clinical applications, aiming to serve as a reference for the design and fabrication of AuNCs-based smart materials for periodontal or peri-implant application.

4.
ACS Appl Mater Interfaces ; 14(38): 42931-42939, 2022 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-36099584

RÉSUMÉ

Immune checkpoint blockade (ICB) has been hailed as the hope for conquering cancer as ICB could produce a significant and durable response to tumor cells. However, the high cost and severe side effects of ICB drugs limited their application for further anticancer therapy. Here, we developed a photoactivated immunotherapy nanoplatform (Apt@AuNC). This nanoplatform could target tumor tissues via enhanced penetration retention (EPR) effect and the aptamer (Apt) could be released from Apt@AuNC in tumor sites via illumination. The immune system in the tumor area was then activated after the combination of Apt and PD-1 protein. The heat generated from AuNC was able to continue killing tumor cells. This nanoplatform could not only achieve the precise immunotherapy but also significantly facilitate the anticancer efficacy.


Sujet(s)
Aptamères nucléotidiques , Tumeurs , Aptamères nucléotidiques/pharmacologie , Aptamères nucléotidiques/usage thérapeutique , Lignée cellulaire tumorale , Dimaprit/analogues et dérivés , Or/pharmacologie , Or/usage thérapeutique , Humains , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Nanostructures , Tumeurs/traitement médicamenteux , Récepteur-1 de mort cellulaire programmée
5.
Acta Biomater ; 142: 264-273, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35101580

RÉSUMÉ

Malignant melanoma remains the life-threatening form of skin cancer with high mortality and poor prognosis. Thus, an ideal melanoma therapeutic strategy is of immediate importance which can remove the primary tumor, as well as inhibit the metastasis and recurrence. Here, we report the fabrication of adjuvant monophosphoryl lipid A (MPLA) lipid bilayer-enveloped and photosensitizer indocyanine green (ICG)-loaded gold nanocages (MLI-AuNCs) for immunogenic phototherapy of aggressive melanoma. Hollow porous AuNCs are used as carriers to deliver MPLA and ICG, and protect ICG from photodegradation. Both AuNCs and ICG absorb near infrared (NIR) light and can be applied in controllable NIR-triggered photothermal and photodynamic combination therapy (PTT/PDT) of melanoma. MLI-AuNCs coated by thermosensitive lipid bilayer exhibit uniform size, good biocompatibility and bioavailability with prominent tumor accumulation, which further improve the PTT/PDT efficacy. MLI-AuNCs under NIR irradiation not only destroy the primary tumor by PTT/PDT, but also elicit robust antitumor immune response with melanoma associated antigens and MPLA released in situ. The released antigens and MPLA subsequently enhance the recruitment and maturation of dendritic cells, which further activate the effector T cells to inhibit metastases and recurrence of melanoma. This immunomodulatory-boosted PTT/PDT nanoplatform provides a new opportunity for highly aggressive melanoma treatment. STATEMENT OF SIGNIFICANCE: An ideal tumor therapeutic strategy not only can remove the primary tumor, but also inhibit metastasis and recurrence. Here, we introduced a versatile nanoplatform MLI-AuNCs for immunogenic phototherapy of aggressive melanoma. Adjuvant MPLA and photosensitizer ICG can be protected and co-delivered to the tumors by thermosensitive lipid-enveloped AuNCs. MLI-AuNCs exhibited prominent tumor accumulation ability and produced the potent PTT/PDT effect to destroy the primary tumors with a single dose of NIR irradiation, as well as elicited the strong antitumor immunity to inhibit the metastasis and relapse. This study may provide a potential therapeutic vaccination strategy against advanced melanoma and other difficult-to-treat cancers.


Sujet(s)
Mélanome , Nanoparticules , Photothérapie dynamique , Lignée cellulaire tumorale , Or/pharmacologie , Humains , Vert indocyanine/pharmacologie , Double couche lipidique , Mélanome/thérapie , Photosensibilisants/pharmacologie , Photothérapie
6.
Acta Biomater ; 134: 621-632, 2021 10 15.
Article de Anglais | MEDLINE | ID: mdl-34329782

RÉSUMÉ

Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. The primary treatment for CRC is surgical resection, along with chemotherapy in more advanced or inoperable cases. There is a growing interest to complement both curative and palliative treatment with immunotherapies such as the programmed cell death-1 (PD-1) and PD-ligand 1 (PDL1) checkpoint inhibitors and transforming growth factor (TGF) ß inhibitors. However, the clinical outcomes of current immunotherapeutic strategies are limited by tumor heterogeneity and drug resistance. Nanomedicine-based photothermal therapy (PTT) has shown encouraging results for solid tumor ablation. Herein, we designed and synthesized gold nanocages functionalized with primary macrophage membrane and surface anti-PDL1 antibody, and loaded with a TGFß inhibitor, galunisertib. The GNC-Gal@CMaP nanocomposites achieved low-temperature PTT and immunogenic cell death, which subsequently enhanced the anti-tumor efficacy of anti-PDL1 antibody and galunisertib via activation of antigen-presenting cells that primed tumor-specific effector T cells. This study provides experimental proof for a combination of immunotherapy and PTT against CRC. STATEMENT OF SIGNIFICANCE: The combination of photothermal therapy (PTT) with immunotherapy can achieve an inherently synergistic anti-tumor effect. Here we integrated low-temperature PTT, PDL1 antibody and TGF-ß inhibitor in hollow gold nanocage nanocomposites (GNC-Gal@CMaP) that selectively targeted colon cancer cells and accumulated in the tumor microenvironment. The GNC-Gal@CMaP nanocomposites achieved low-temperature PTT and immunogenic cell death, which subsequently enhanced the anti-tumor efficacy of anti-PDL1 antibody and galunisertib via activation of antigen-presenting cells that primed tumor-specific effector T cells. This study provides experimental proof for a combination of immunotherapy and PTT against CRC.


Sujet(s)
Tumeurs colorectales , Or , Anticorps monoclonaux humanisés , Tumeurs colorectales/traitement médicamenteux , Humains , Immunothérapie , Photothérapie , Thérapie photothermique , Pyrazoles , Quinoléines , Microenvironnement tumoral
7.
J Nanobiotechnology ; 19(1): 54, 2021 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-33627152

RÉSUMÉ

BACKGROUND: Gene and chemical therapy has become one of the rising stars in the field of molecular medicine during the last two decades. However, there are still numerous challenges in the development of efficient, targeted, and safe delivery systems that can avoid siRNA degradation and reduce the toxicity and adverse effects of chemotherapy medicine. RESULTS: In this paper, a highly efficient AS1411 aptamer modified, dsDNA and MMP-2 cleavable peptide-fabricated gold nanocage vehicle, which could load doxorubicin hydrochloride (DOX) and siRNAs to achieve a combination of tumor responsive genetic therapy, chemotherapy, and photothermal treatment is presented. Our results show that this combined treatment achieved targeted gene silencing and tumor inhibition. After nearly one month of treatment with DOX-loaded Au-siRNA-PAA-AS1411 nanoparticles with one dose every three days in mice, a synergistic effect promoting the eradication of long-lived tumors was observed along with an increased survival rate of mice. The combined genetic, chemotherapeutic, and photothermal treatment group exhibited more than 90% tumor inhibition ratio (tumor signal) and a ~ 67% survival rate compared with a 30% tumor inhibition ratio and a 0% survival rate in the passive genetic treatment group. CONCLUSIONS: The development of nanocarriers with double-stranded DNA and MMP-2 cleavable peptides provides a new strategy for the combined delivery of gene and chemotherapy medicine. Au-siRNA-PAA-AS1411 exerts high anticancer activities on lung cancer, indicating immense potentials for clinical application.


Sujet(s)
Techniques de transfert de gènes , Or/composition chimique , Or/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Nanoparticules métalliques/composition chimique , Petit ARN interférent/pharmacologie , Animaux , Aptamères nucléotidiques , Lignée cellulaire tumorale , Doxorubicine/administration et posologie , Vecteurs de médicaments , Systèmes de délivrance de médicaments/méthodes , Poumon , Tumeurs du poumon/anatomopathologie , Souris , Souris de lignée BALB C , Souris nude , Oligodésoxyribonucléotides , Taille de particule , Taux de survie
8.
Int J Pharm ; 593: 120162, 2021 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-33307159

RÉSUMÉ

Due to the lack of effective therapeutic targets and the passive delivery of a limited quantity of nanoparticles to the tumors, the photothermal conversion agents used in photothermal therapy (PTT) have not been effective in treating triple-negative breast cancer (TNBC). As a result, there is a need to improve the tumor-targeting ability of these photothermal conversion agents. To address this, a microwave-triggered heat shock protein (HSP)-targeted gold nano-system (cmHSP-AuNC), with a gold nanocage (AuNC) as a photothermal conversion agent and anti-HSP monoclonal antibody (cmHSP) as a targeting ligand, was fabricated. cmHSP-AuNC was characterized based on morphology, particle size, zeta potentials, absorption spectrum, and photothermal conversion ability. The expression of HSP70 in 4T1 cells after microwave irradiation was verified by western blotting, and the optimal treatment conditions to achieve the highest expression were determined. Both in vitro and in vivo results indicated that the induction through microwave irradiation could effectively activate the HSP70 overexpression in TNBC, thereby significantly improving the targeting ability, tumor accumulation and anti-tumor efficacy of cmHSP-AuNC. This study proposes a promising strategy for improving the targeting ability and therapeutic efficacy of PTT.


Sujet(s)
Tumeurs du sein , Tumeurs du sein triple-négatives , Lignée cellulaire tumorale , Femelle , Or , Protéines du choc thermique , Humains , Micro-ondes , Photothérapie , Thérapie photothermique , Tumeurs du sein triple-négatives/thérapie
9.
Int J Nanomedicine ; 15: 10271-10284, 2020.
Article de Anglais | MEDLINE | ID: mdl-33364758

RÉSUMÉ

INTRODUCTION: Cancer theragnosis involving cancer diagnosis and targeted therapy simultaneously in one integrated system would be a promising solution of cancer treatment. Herein, a convenient and practical cancer theragnosis agent was constructed by combining gold nanocages (AuNCs) covered with selenium and a chitosan (CS) shell (AuNCs/Se) to incorporate the anti-cancer drug doxorubicin (DOX) as a multifunctional targeting nanocomposite (AuNCs/DOX@Se-iRGD) for photoacoustic imaging (PAI)-guided chemo-photothermal synergistic therapy that contributes to enhanced anti-cancer efficacy. The novel design of AuNCs/DOX@Se-iRGD gives the nanocomposite two outstanding properties: (1) AuNCs, with excellent LSPR property in the NIR region, act as a contrast agent for enhanced PAI and photothermal therapy (PTT); (2) Se acts as an anti-cancer nanoagent and drug delivery cargo. METHODS: The photothermal performance of these nanocomposites was evaluated in different concentrations with laser powder densities. These nanocomposites were also incubated in pH 5.3, 6.5, 7.4 PBS and NIR laser to study their drug release ability. The cellular uptake was studied by measuring the Se and Au concentrations inside the cells using inductively coupled plasma-mass spectrometry (ICP-MS). Besides, in vitro and in vivo anti-tumor activity were carried out by cytotoxicity assay MTT and tumor model nude mice, respectively. As for imaging, the PA value and images of these nanocomposites accumulated in the tumor site were sequentially collected at specific time points for 48 h. RESULTS AND DISCUSSION: The prepared AuNCs/DOX@Se-iRGD showed excellent biocompatibility and physiological stability in different media. In vivo results indicated that the targeting nanocomposite presented the strongest contrast-enhanced PAI signals, which could provide contour and location information of tumor, 24 h after intravenous injection. Likewise, the combined treatment of chemo- and photothermal synergistic therapy significantly inhibited tumor growth when compared with the two treatments carried out separately and showed negligible acute toxicity to the major organs. CONCLUSION: This study demonstrates that AuNCs/DOX@Se-iRGD has great prospect to become a multifunctional anti-tumor nanosystem for PAI-guided chemo- and photothermal synergistic therapy.


Sujet(s)
Vecteurs de médicaments/composition chimique , Or/composition chimique , Techniques photoacoustiques , Thérapie photothermique/méthodes , Sélénium/composition chimique , Nanomédecine théranostique/méthodes , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Association thérapeutique , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Libération de médicament , Humains , Souris , Souris nude
10.
Acta Biomater ; 104: 188-197, 2020 03 01.
Article de Anglais | MEDLINE | ID: mdl-31945508

RÉSUMÉ

Advanced colorectal cancer has a high mortality rate since conventional treatments have limited therapeutic effects and poor prognosis with high risks of metastasis and recurrence. Photodynamic therapy (PDT) is a promising treatment modality for the eradication of colorectal cancer, but its curative efficacy is severely affected by tumor hypoxia. Herein, we developed a core-shell gold nanocage coated with manganese dioxide and hyaluronic acid (AMH) for targeted delivery to colorectal tumors and oxygenation-boosted immunogenic phototherapy in situ. The AMH nanoparticles can generate abundant oxygen from mild acidic/H2O2 medium, which can further enhance the PDT efficacy of AMH itself under near infrared (NIR) irradiation. Meanwhile, AMH-based PDT induced immunogenic cell death (ICD) of tumor cells with damage-associated molecular patterns (DAMPs) release and facilitated the dendritic cells (DCs) maturation to further potentiate the systematic antitumor immunity against advanced tumors. In vivo experiment results exhibited that AMH nanoparticles not only had the ability of targeting tumor but also in situ produced sufficient oxygen to relieve the tumor hypoxia. Furthermore, AMH-mediated oxygen-boosted immunogenic PDT effectively inhibited the tumor growth and recurrence. Thus, this work provides a potent targeted delivery nanoplatform for enhanced immunogenic PDT against advanced cancers. STATEMENT OF SIGNIFICANCE: Local hypoxic tumor microenvironment not only greatly limits the photodynamic therapy (PDT) efficacy, but also has an association with tumor invasiveness and metastasis. This study provides an AMH nanoparticle for targeted delivery to colorectal tumors and oxygenation-boosted immunogenic PDT in situ. AMH nanoparticle exhibits a good tumor-targeted ability to in situ produce abundant oxygen to relieve the tumor hypoxia, and initiates the potent oxygen-boosted immunogenic PDT effect under NIR irradiation to effectively inhibit the growth and recurrence of colorectal tumor. This oxygen-boosted immunogenic PDT nanosystem can be a promising candidate for advanced tumor treatment.


Sujet(s)
Tumeurs colorectales/immunologie , Tumeurs colorectales/thérapie , Nanoparticules/composition chimique , Oxygène/pharmacologie , Photothérapie , Animaux , Lignée cellulaire tumorale , Tumeurs colorectales/anatomopathologie , Cellules dendritiques/métabolisme , Femelle , Or/composition chimique , Acide hyaluronique/composition chimique , Composés du manganèse/composition chimique , Souris de lignée BALB C , Nanoparticules/ultrastructure , Oxydes/composition chimique , Photothérapie dynamique , Distribution tissulaire
11.
Nanomicro Lett ; 13(1): 28, 2020 Nov 21.
Article de Anglais | MEDLINE | ID: mdl-34138183

RÉSUMÉ

The immune response of a biomaterial determines its osteoinductive effect. Although the mechanisms by which some immune cells promote regeneration have been revealed, the biomaterial-induced immune response is a dynamic process involving multiple cells. Currently, it is challenging to accurately regulate the innate and adaptive immune responses to promote osteoinduction in biomaterials. Herein, we investigated the roles of macrophages and dendritic cells (DCs) during the osteoinduction of biphasic calcium phosphate (BCP) scaffolds. We found that osteoinductive BCP directed M2 macrophage polarization and inhibited DC maturation, resulting in low T cell response and efficient osteogenesis. Accordingly, a dual-targeting nano-in-micro scaffold (BCP loaded with gold nanocage, BCP-GNC) was designed to regulate the immune responses of macrophages and DCs. Through a dual-wavelength photosensitive switch, BCP-GNC releases interleukin-4 in the early stage of osteoinduction to target M2 macrophages and then releases dexamethasone in the later stage to target immature DCs, creating a desirable inflammatory environment for osteogenesis. This study demonstrates that biomaterials developed to have specific regulatory capacities for immune cells can be used to control the early inflammatory responses of implanted materials and induce osteogenesis.

12.
Acta Biomater ; 102: 416-426, 2020 01 15.
Article de Anglais | MEDLINE | ID: mdl-31760223

RÉSUMÉ

Controlling of pro-inflammation induced by pro-inflammatory cytokines and anti-inflammatory response induced by M2 macrophages is important for osteogenesis in the process of bone tissue repair. Thus, we fabricated biomimetic anti-inflammatory nano-capsule (BANC) that can block cytokines and promote M2 macrophage polarization, presenting a positive role for bone tissue repair. The BANC is a biomimic nanosystem, coated with lipopolysaccharide-treated macrophage cell membranes with cytokine receptors enveloping gold nanocage (AuNC) as "cytokine blocker", and loaded with resolvin D1 inside into AuNC as "M2 polarization inducer" whose controlled-release could be triggered under near-infrared laser irradiation in sequence, and these chronological events were consistent with the healing process of bone tissue repair. Moreover, in vivo application of femoral bone defects revealed that the BANC composite boron-containing mesoporous bioactive glass scaffolds improved the final effects of bone tissue repair through preventing inflammatory response, promoting M2 polarization in sequence in accord with the in vitro investigation. Hence, cytokine neutralization and M2 macrophage polarization enables the BANC to enhance the bone tissue repair as a biomimetic anti-inflammation effector. Therefore, this study provides potential therapeutic strategies for trauma-mediated or inflammation-related bone defects based on a biomimetic nanomaterial with weakened pro-inflammatory and enhanced anti-inflammatory effects. STATEMENT OF SIGNIFICANCE: Cell membrane-mimic nanomaterials have been popular for blocking natural cell responses for some infection diseases, yet their role in biological process of bone repair is unknown. Here, we fabricated Biomimetic Anti-inflammatory Nano-Capsule (BANC), coated with cell membrane with cytokines receptors on the surface which could neutralize the pro-inflammatory cytokine receptor to block activated pro-inflammation, loaded with Resolvin D1 inside which could be controllably released by NIR irradiation to promote M2 macrophage polarization for the following bone formation during the process of bone repair. Administration of BANC as cytokines blocker and M2 polarization inducer to enhance the bone regeneration, thus presenting a promising potential for the treatment of bone repair and regeneration.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Régénération osseuse/effets des médicaments et des substances chimiques , Cytokines/antagonistes et inhibiteurs , Inflammation/prévention et contrôle , Macrophages/effets des médicaments et des substances chimiques , Nanocapsules/usage thérapeutique , Animaux , Matériaux biomimétiques/composition chimique , Membrane cellulaire/composition chimique , Acide docosahexaénoïque/usage thérapeutique , Vecteurs de médicaments/composition chimique , Vecteurs de médicaments/usage thérapeutique , Femelle , Fémur/effets des médicaments et des substances chimiques , Lipopolysaccharides/composition chimique , Lipopolysaccharides/usage thérapeutique , Souris , Souris de lignée C57BL , Nanocapsules/composition chimique , Cellules RAW 264.7 , Récepteurs aux cytokines/composition chimique , Récepteurs aux cytokines/usage thérapeutique
13.
Biomaterials ; 217: 119327, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31299626

RÉSUMÉ

Photochemotherapy is currently an effective anticancer therapy. Recently, it has been reported that cancer cells pretreated with epidermal growth factor receptor (EGFR) inhibitor erlotinib (Erl) can significantly synergize its apoptosis against the DNA damaging agent doxorubicin (Dox). As a result, we designed two gold nanocages (Au NCs) microcontainers covered with different smart polymer shell-PAA (pH responsive) and p (NIPAM-co-AM) (temperature responsive) containing Erl and Dox respectively. The acidic tumor microenvironment and NIR light irradiation can selectively activate the release of Erl and Dox. Time staggered release of Erl and Dox and photothermal therapy enhance the apoptotic signaling pathways, resulting in improved tumor cell killing in both MCF-7 (low EGFR expression) and A431 (very high EGFR expression) tumor cells, but more efficient in the latter. The photochemotherapy strategy controls the order and duration of drug exposure precisely in spatial and temporal, and significantly improves the therapeutic efficacy against high EGFR expressed tumors.


Sujet(s)
Doxorubicine/pharmacologie , Systèmes de délivrance de médicaments , Chlorhydrate d'erlotinib/pharmacologie , Or/composition chimique , Hyperthermie provoquée , Nanoparticules métalliques/composition chimique , Photothérapie , Polymères/composition chimique , Animaux , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Mort cellulaire/effets des médicaments et des substances chimiques , Libération de médicament , Endocytose/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Femelle , Humains , Concentration en ions d'hydrogène , Rayons infrarouges , Cellules MCF-7 , Nanoparticules métalliques/ultrastructure , Souris de lignée BALB C , Souris nude , Température , Facteurs temps
14.
Pharmaceutics ; 11(3)2019 Mar 25.
Article de Anglais | MEDLINE | ID: mdl-30934538

RÉSUMÉ

Gold nanocages (AuNCs) are biocompatible and porous nanogold particles that have been widely used in biomedical fields. In this study, hyaluronic acid (HA) and peptide- modified gold nanocages (HA-AuNCs/T/P) loaded with 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA-1) were prepared to investigate their potential for combating inflammation. TPCA-1 was released from AuNCs, intracellularly when HA was hydrolyzed by hyaluronidase. HA-AuNCs/T/P show a much higher intracellular uptake than AuNCs/T/P, and exhibit a much higher efficacy on the suppression of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) than free TPCA-1, suggesting great improvement to the anti-inflammatory efficacy of TPCA-1 through the application of AuNCs. HA-AuNCs/T/P can also reduce the production of reactive oxygen species in inflammatory cells. This study suggests that HA-AuNCs/T/P may be potential agents for anti-inflammatory treatment, and are worthy of further investigation.

15.
Biomaterials ; 197: 380-392, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30703743

RÉSUMÉ

Systemic lupus erythematosus (SLE) constitutes an autoimmune disease characterized by the breakdown of tolerance to self-antigens, sustained production of pathogenic autoantibodies, and damage to multiple organs and tissues. Nanoparticle (NP)-based therapeutics have demonstrated efficacy in attenuating the progression of SLE. However, investigations of nano-drugs that address the crucial initiating factor in the pathogenesis of SLE; e.g., inefficient clearance of apoptotic cells by phagocytes and consequent accumulation of self-antigens, have seldom been reported. Here, an apoptotic cell-mimicking gold nanocage (AuNC)-based nano drug carrier capable of correcting the impaired clearance of apoptotic cells in SLE was rationally designed and generated by conjugating phosphatidylserine (PS) on the surface of liposome-coated AuNCs for liver X receptor (LXR) agonist T0901317 delivery. Notably, PS-lipos-AuNC@T0901317 could efficiently enhance apoptotic cell clearance by elevating the expression of Mer, one of the pivotal phagocytosis-associated receptors on macrophages, resulting in decreased production of anti-dsDNA autoantibodies, reduced inflammatory response, and alleviation of kidney damage in lupus model mice. Additionally, PS-lipos-AuNC could be tracked by photoacoustic imaging for nano drug carrier biodistribution. By addressing the crucial pathogenic factor of SLE, the NP-based delivery system in this study is envisioned to provide a promising strategy to treat this complex and challenging disease.


Sujet(s)
Apoptose , Systèmes de délivrance de médicaments , Or/administration et posologie , Hydrocarbures fluorés/administration et posologie , Récepteurs hépatiques X/agonistes , Lupus érythémateux disséminé/traitement médicamenteux , Nanocapsules/administration et posologie , Sulfonamides/administration et posologie , Animaux , Autoanticorps/analyse , Cytokines/métabolisme , Évolution de la maladie , Évaluation préclinique de médicament , Femelle , Or/pharmacocinétique , Hydrocarbures fluorés/usage thérapeutique , Hydrocarbures fluorés/toxicité , Liposomes/administration et posologie , Glomérulonéphrite lupique/traitement médicamenteux , Glomérulonéphrite lupique/immunologie , Souris , Souris de lignée C57BL , Souris de lignée MRL lpr , Phosphatidylsérine , Sulfonamides/usage thérapeutique , Sulfonamides/toxicité , Distribution tissulaire , c-Mer Tyrosine kinase/biosynthèse , c-Mer Tyrosine kinase/génétique
16.
Nanomedicine ; 18: 315-325, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30391484

RÉSUMÉ

Photodynamic therapy is a clinically approved treatment approach for cancer. However, it has limited applications owing to poor water solubility and the short wavelength absorption of the photosensitizer (PS). We selected a near-infrared photosensitizer, SiNC, and encapsulated into a gold nanocage (AuNC) in the presence of phase-changing material. Then, the PS-encapsulated nanocage was coated with glycol chitosan (GC) with a cleavable peptide linkage or stable cysteine linkage to protect the PS from premature release and to improve the biocompatibility of the nanocage. We obtained particles of GC-coated SiNC-encapsulated AuNC with a neutral surface charge and approximately 160 nm in size. The enzyme-cleavable peptide-linked GC formulation (GC-pep@SiNC-AuNC) showed stronger phototoxicity and tumor suppression efficacy in a glioblastoma model compared with free NIR-PS and stable cysteine-linked GC-AuNC (GC-cys@SiNC-AuNC). This polymer-coated SiNC-AuNC may be a promising agent for brain cancer phototherapy.


Sujet(s)
Chitosane/composition chimique , Glioblastome/thérapie , Or/composition chimique , Rayons infrarouges , Nanoparticules métalliques/composition chimique , Photosensibilisants/usage thérapeutique , Photothérapie , Animaux , Lignée cellulaire tumorale , Chitosane/synthèse chimique , Endocytose , Humains , Nanoparticules métalliques/ultrastructure , Souris de lignée BALB C , Souris nude , Peptides/synthèse chimique , Peptides/composition chimique , Spectroscopie par résonance magnétique du proton , Distribution tissulaire
17.
Mol Imaging ; 17: 1536012118775827, 2018.
Article de Anglais | MEDLINE | ID: mdl-29873290

RÉSUMÉ

PURPOSE: Melanoma is a lethal skin cancer with unmet clinical needs for targeted imaging and therapy. Nanoscale materials conjugated with targeting components have shown great potential to improve tumor delivery efficiency while minimizing undesirable side effects in vivo. Herein, we proposed to develop targeted nanoparticles for melanoma theranostics. METHOD: In this work, gold nanocages (AuNCs) were conjugated with α-melanocyte-stimulating hormone (α-MSH) peptide and radiolabeled with 64Cu for melanocortin 1 receptor-(MC1R) targeted positron emission tomography (PET) in a mouse B16/F10 melanoma model. RESULTS: Their controlled synthesis and surface chemistry enabled well-defined structure and radiolabeling efficiency. In vivo pharmacokinetic evaluation demonstrated comparable organ distribution between the targeted and nontargeted AuNCs. However, micro-PET/computed tomography (CT) imaging demonstrated specific and improved tumor accumulation via MC1R-mediated delivery. By increasing the coverage density of α-MSH peptide on AuNCs, the tumor delivery efficiency was improved. CONCLUSION: The controlled synthesis, sensitive PET imaging, and optimal tumor targeting suggested the potential of targeted AuNCs for melanoma theranostics.


Sujet(s)
Or/composition chimique , Mélanome expérimental/imagerie diagnostique , Mélanome expérimental/anatomopathologie , Nanoparticules métalliques/composition chimique , Imagerie moléculaire/méthodes , Tomographie par émission de positons , Récepteur de la mélanocortine de type 1/métabolisme , Animaux , Nanoparticules métalliques/ultrastructure , Souris de lignée C57BL , Polyéthylène glycols/composition chimique , Distribution tissulaire , Tomodensitométrie , Hormone mélanotrope alpha/composition chimique
18.
Mol Imaging Biol ; 20(6): 919-929, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-29736563

RÉSUMÉ

PURPOSE: Tumor proteases have been recognized as significant regulators in the tumor microenvironment, but the current strategies for in vivo protease imaging have tended to focus on the development of a probe design rather than the investigation of a novel imaging strategy by leveraging the imaging technique and probe. Herein, it is the first report to investigate the ability of multispectral photoacoustic imaging (PAI) to estimate the distribution of protease cleavage sites inside living tumor tissue by using an activatable photoacoustic (PA) probe. PROCEDURES: The protease MMP-2 is selected as the target. In this probe, gold nanocages (GNCs) with an absorption peak at ~ 800 nm and fluorescent dye molecules with an absorption peak at ~ 680 nm are conjugated via a specific enzymatic peptide substrate. Upon enzymatic activation by MMP-2, the peptide substrate is cleaved and the chromophores are released. Due to the different retention speeds of large GNCs and small dye molecules, the probe alters its intrinsic absorption profile and produces a distinct change in the PA signal. A multispectral PAI technique that can distinguish different chromophores based on intrinsic PA spectral signatures is applied to estimate the signal composition changes and indicate the cleavage interaction sites. Finally, the multispectral PAI technique with the activatable probe is tested in solution, cultured cells, and a subcutaneous tumor model in vivo. RESULTS: Our experiment in solution with enzyme ± inhibitor, cell culture ± inhibitor, and in vivo tumor model with administration of the developed probe ± inhibitor demonstrated the probe was cleaved by the targeted enzyme. Particularly, the in vivo estimation of the cleavage site distribution was validated with the result of ex vivo immunohistochemistry analysis. CONCLUSIONS: This novel synergy of the multispectral PAI technique and the activatable probe is a potential strategy for the distribution estimation of tumor protease activity in vivo.


Sujet(s)
Or/composition chimique , Imagerie moléculaire/méthodes , Sondes moléculaires/composition chimique , Tumeurs/diagnostic , Peptide hydrolases/métabolisme , Techniques photoacoustiques/méthodes , Animaux , Humains , Souris , Souris nude , Nanocapsules/composition chimique , Tumeurs/enzymologie , Tumeurs/anatomopathologie , Peptide hydrolases/analyse , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Int J Hyperthermia ; 34(1): 19-29, 2018 02.
Article de Anglais | MEDLINE | ID: mdl-28540812

RÉSUMÉ

PURPOSE: To demonstrate delivery of Au nanocages to cells using the galectin-1 binding peptide anginex (Ax) and to demonstrate the value of this targeting for selective in vitro photothermal cell killing. MATERIALS AND METHODS: Au nanocages were synthesised, coated with polydopamine (PDA), and conjugated with Ax. Tumour and endothelial cell viability was measured with and without laser irradiation. Photoacoustic (PA) mapping and PA flow cytometry were used to confirm cell targeting in vitro and in tissue slices ex vivo. RESULTS: Cell viability was maintained at ≥50% at 100 pM suggesting low toxicity of the nanocage alone. Combining the targeted construct (25 pM) with low power 808 nm laser irradiation for 10-20 min (a duration previously shown to induce rapid and sustained heating of Au nanocages [AuNC] in solution), resulted in over 50% killing of endothelial and tumour cells. In contrast, the untargeted construct combined with laser irradiation resulted in negligible cell killing. We estimate approximately 6 × 104 peptides were conjugated to each nanocage, which also resulted in inhibition of cell migration. Binding of the targeted nanocage reached a plateau after three hours, and cell association was 20-fold higher than non-targeted nanocages both in vitro and ex vivo on tumour tissue slices. A threefold increase in tumour accumulation was observed in preliminary in vivo studies. CONCLUSIONS: These studies demonstrate Ax's potential as an effective targeting agent for Au-based theranostics to tumour and endothelial cells, enabling photothermal killing. This platform further suggests potential for multimodal in vivo therapy via next-generation drug-loaded nanocages.


Sujet(s)
Galectine 1/métabolisme , Or/métabolisme , Nanostructures/composition chimique , Photothérapie/méthodes , Animaux , Souris , Souris de lignée BALB C
20.
Acta Biomater ; 64: 223-236, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-29030300

RÉSUMÉ

A pH-sensitive copolymer PAsp(DIP)-b-PAsp(MEA) (PDPM) was synthesized and self-assembled to micelle loading chemotherapeutic drug doxorubicin (DOX) and introducing a gold nanocage structure for photothermo-chemotherapy and photoacoustic imaging. After further surface modification with polyethylene glycol (PEG), the DOX-loaded pH-sensitive gold nanocage (D-PGNC) around 100 nm possessed a uniform spherical structure with a pH-sensitive core of PAsp(DIP) incorporating DOX, an interlayer crosslinked via disulfide bonds and decorated with discontinuous gold shell, and a PEG corona. The release of DOX from D-PGNC was turned off in bloodstream due to the cross-linking and gold decoration of interlayer but turned on inside tumor tissue by multiple stimulations including the low pH value of tumor tissue (≈6.8), the low lysosomal pH value of cancer cells (≈5.0) and near-infrared (NIR) irradiation. The gold nanocage receiving NIR irradiation could generate hyperthermia to ablate tumor cells. Moreover, the photoacoustic (PA) imaging and analysis of DOX fluorescence inside tumor tissue demonstrated that photothermal therapy based on the gold nanocage effectively drove DOX penetration inside tumor. Owing to the rapid intratumor release and deep tissue penetration of drug favorable for killing cancer cells survived the photothermal therapy, the combined therapy based on D-PGNC via NIR irradiation exhibited a synergistic treatment effect superior to either chemotherapy or NIR-induced photothermal therapy alone. STATEMENT OF SIGNIFICANCE: The novelty of the manuscript is its multifunctional system which incorporates anticancer drug DOX in its pH-sensitive core and acts as a template to introduce a gold nanocage. This nanomedicine presents potentials of sequestrating drug molecules in blood circulation but releasing them inside tumor upon responding to the acidic microenvironment therein. Exposure to NIR laser further expedited the pH-sensitive DOX release and promoted DOX penetration into cancer tissues far away from the vasculature. Consequently, the combined photothermo-chemotherapy showed synergistic effects to inhibit tumor growth and prolong animal survival in nude mice bearing human SKOV-3 ovarian tumor. Moreover, owing to the decoration with gold nanocage, the tumor accumulation and intratumor diffusion of the micelles were easily trackable using photoacoustic imaging.


Sujet(s)
Or/composition chimique , Hyperthermie provoquée/méthodes , Nanoparticules métalliques/composition chimique , Micelles , Tumeurs , Techniques photoacoustiques/méthodes , Photothérapie dynamique/méthodes , Lignée cellulaire tumorale , Humains , Concentration en ions d'hydrogène , Tumeurs/imagerie diagnostique , Tumeurs/métabolisme , Tumeurs/thérapie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...