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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Korean Med Sci ; 34(44): e272, 2019 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-31726492

RESUMO

BACKGROUND: Nanoparticle-mediated photothermal therapy (PTT) has been well studied as a treatment for cancer. However, the therapeutic outcome of PTT is often hindered by the penetration depth of laser light. In the tumor margin beyond the laser penetration limit, tumor recurrence often occurs, bypassing the immune response of the host. Accumulating evidence suggests the prominent role of tumor microenvironment (TME) and its interactions with the immune components contribute to an immunosuppressive milieu during the post-therapy period. Here, we explored the immunosuppressive cascade generated after PTT, which is responsible for tumor recurrence, and identified the potential targets to achieve an effective PTT period. METHODS: Here, we investigated the immunosuppressive cascade generated after PTT in a CT26 tumor bearing mouse. The liposomal system loaded with the indocyanine green (ICG) was utilized for the generation of PTT with high efficiency. Immunological factors such as cytokines and protein expressions post-therapy were investigated through enzyme-linked immunosorbent assay, flow cytometry and western blot analysis. RESULTS: Our results suggested that PTT with ICG-loaded liposomes (Lipo-ICG) was effective for the first 5 days after treatment, resulting in tumor suppression. However, an immunosuppressive and pro-inflammatory environment developed thereafter, causing the recruitment and upregulation of the immune evasion factors of heat shock protein 70, programmed death ligand 1, indoleamine-dioxygenase, interleukin-6, transforming growth factor-ß, regulatory T-cells, and myeloid-derived suppressor cells, to develop immunotolerance. CONCLUSION: Collectively, these findings have determined potential therapeutic targets to modulate the TME during PTT and achieve tumor ablation without remission.


Assuntos
Tolerância Imunológica , Neoplasias/imunologia , Microambiente Tumoral , Animais , Linhagem Celular Tumoral , Citocinas/sangue , Feminino , Humanos , Verde de Indocianina/química , Verde de Indocianina/metabolismo , Verde de Indocianina/uso terapêutico , Raios Infravermelhos , Lipossomos/química , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Recidiva Local de Neoplasia , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/metabolismo , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Distribuição Tecidual
2.
Molecules ; 24(5)2019 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-30832383

RESUMO

Biodegradable polymers have been developed for the targeted delivery of therapeutics to tumors. However, tumor targeting and imaging are usually limited by systemic clearance and non-specific adsorption. In this study, we used poly(amino acid) derivatives, such as poly(succinimide), to synthesize a nanomicelle-forming poly(hydroxyethylaspartamide) (PHEA, P) modified sequentially with octadecylamine, polyethylene glycol (PEG, P), and glycine (G) to design PHEA-PEG-glycine (PPG) nanoparticles (NPs). These PPG NPs were further tethered to cyclic Arg-Gly-Asp (cRGD) sequences for formulating tumor-targeting PPG-cRGD NPs, and then loaded with IR-780 dye (PPG-cRGD-IR-780) for visualizing tumor homing. cRGD cloaked in PPG NPs could bind specifically to both tumor endothelium and cancer cells overexpressing αvß3 integrins. PPG-cRGD NPs exhibited enhanced physiological stability, cellular viability, and targeted intracellular uptake in cancer cells. In addition, PPG-cRGD NPs offered enhanced systemic circulation, leading to preferential tumor targeting and prolonged fluorescence tumor imaging for nearly 30 days. Nevertheless, non-targeted formulations demonstrated premature systemic clearance with short-term tumor imaging. Histochemical analysis showed no damage to normal organs, reaffirming the biocompatibility of PHEA polymers. Overall, our results indicated that PPG-cRGD NPs, which were manipulated to obtain optimal particle size and surface charge, and were complemented with tumor targeting, could improve the targeted and theranostic potential of therapeutic delivery.


Assuntos
Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Peptídeos/administração & dosagem , Polietilenoglicóis/administração & dosagem , Aminas/síntese química , Aminas/química , Animais , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicina/química , Humanos , Indóis/química , Integrinas/genética , Camundongos , Nanopartículas/química , Neoplasias/patologia , Oligopeptídeos/administração & dosagem , Oligopeptídeos/síntese química , Oligopeptídeos/química , Peptídeos/síntese química , Peptídeos/química , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Polímeros/administração & dosagem , Polímeros/química , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Biomater Sci ; 11(7): 2581-2589, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36794531

RESUMO

CrkII, a member of the adaptor protein family, is known to participate in bone homeostasis via the regulation of osteoclasts and osteoblasts. Therefore, silencing CrkII would beneficially impact the bone microenvironment. In this study, CrkII siRNA encapsulated by a bone-targeting peptide (AspSerSer)6-liposome was evaluated for its therapeutic applications using a receptor activator of nuclear factor kappa-B ligand (RANKL)-induced bone loss model. (AspSerSer)6-liposome-siCrkII maintained its gene-silencing ability in both osteoclasts and osteoblasts in vitro and significantly reduced osteoclast formation while increasing osteoblast differentiation in vitro. Fluorescence image analyses showed that the (AspSerSer)6-liposome-siCrkII was present largely in bone, where it remained present for up to 24 hours and was cleared by 48 hours, even when systemically administrated. Importantly, microcomputed-tomography revealed that bone loss induced by RANKL administration was recovered by systemic administration of (AspSerSer)6-liposome-siCrkII. Collectively, the findings of this study suggest that (AspSerSer)6-liposome-siCrkII is a promising therapeutic strategy for the development of treatments for bone diseases, as it overcomes the adverse effects derived from ubiquitous expression via bone-specific delivery of siRNA.


Assuntos
Doenças Ósseas , Reabsorção Óssea , Humanos , Osteogênese , RNA Interferente Pequeno/metabolismo , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Lipossomos/metabolismo , Osteoclastos , Osteoblastos , Doenças Ósseas/metabolismo , Diferenciação Celular
4.
Adv Mater ; 35(13): e2209615, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36649533

RESUMO

Pulmonary exposure to some engineered nanomaterials can cause chronic lesions as a result of unresolved inflammation. Among 2D nanomaterials and graphene, MoS2 has received tremendous attention in optoelectronics and nanomedicine. Here an integrated approach is proposed to follow up the transformation of MoS2 nanosheets at the nanoscale and assesss their impact on lung inflammation status over 1 month after a single inhalation in mice. Analysis of immune cells, alveolar macrophages, extracellular vesicles, and cytokine profiling in bronchoalveolar lavage fluid (BALF) shows that MoS2 nanosheets induced initiation of lung inflammation. However, the inflammation is rapidly resolved despite the persistence of various biotransformed molybdenum-based nanostructures in the alveolar macrophages and the extracellular vesicles for up to 1 month. Using in situ liquid phase transmission electron microscopy experiments, the dynamics of MoS2 nanosheets transformation triggered by reactive oxygen species could be evidenced. Three main transformation mechanisms are observed directly at the nanoscale level: 1) scrolling of the dispersed sheets leading to the formation of nanoscrolls and folded patches, 2) etching releasing soluble MoO4 - , and 3) oxidation generating oxidized sheet fragments. Extracellular vesicles released in BALF are also identified as a potential shuttle of MoS2 nanostructures and their degradation products and more importantly as mediators of inflammation resolution.


Assuntos
Vesículas Extracelulares , Pneumonia , Animais , Camundongos , Molibdênio/química , Dissulfetos/química , Inflamação/induzido quimicamente
5.
Biomater Sci ; 11(5): 1853-1866, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36655902

RESUMO

Human papilloma virus (HPV), one of the most common cancer-causing viruses, accounts for more than 90% of human anal and cervical cancers. Clinical studies have focused on adjuvant therapy with vaccines to improve therapeutic outcomes in patients with late-stage HPV-related cancers. In the present study, a mannose receptor (CD206) targeting a lithocholic acid-modified polyethylenimine (PEI) nano-adjuvant delivering the toll-like receptor 7/8 agonist, resiquimod (R848) (mLAPMi-R848), in a HPV E6- and E7-expressing TC-1 tumor murine model was developed. Peritumoral administration of mLAPMi resulted in enhanced accumulation in tumor/tumor-draining lymph nodes and significantly targeted antigen presenting cells like macrophage and dendritic cells. PEI-based nanocarriers can exploit the adjuvant potency of R848 and improve the antitumor immunity. Hence, co-administration of mLAPMi-R848 along with an E6E7 peptide in TC-1 tumor mice eradicated tumor burden and elicited splenocyte-induced cytotoxicity in TC-1 cancer cells. In a bilateral TC-1 tumor model, administration of mLAPMi-R848 and E6E7 peptide significantly suppressed both primary and secondary tumor burdens and improved the overall survival rate. Immune cell profiling revealed elevated levels of mature DCs and CD8+ T cells but reduced levels of tumor-associated immunosuppressive cells (TAICs) like myeloid derived suppressor cells (MDSCs) and regulatory T (Treg) cells in distal tumors. Overall, this study demonstrated that mLAPMi-R848 has improved the antitumor immunity of the peptide antigen against HPV-induced cancers by targeted immunodulation of antigen presenting cells (APCs) and reducing TAICs. Furthermore, this nano-adjuvant has the potential to offer a new treatment option for patients with cervical cancer and can be applied for the treatment of other HPV induced cancers.


Assuntos
Infecções por Papillomavirus , Vacinas contra Papillomavirus , Neoplasias do Colo do Útero , Feminino , Humanos , Animais , Camundongos , Neoplasias do Colo do Útero/tratamento farmacológico , Açúcares , Vacinas contra Papillomavirus/uso terapêutico , Infecções por Papillomavirus/tratamento farmacológico , Infecções por Papillomavirus/prevenção & controle , Proteínas E7 de Papillomavirus/uso terapêutico , Adjuvantes Imunológicos/uso terapêutico , Peptídeos/uso terapêutico , Vacinas de Subunidades Antigênicas , Camundongos Endogâmicos C57BL
6.
Int J Nanomedicine ; 18: 4055-4066, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37520301

RESUMO

Purpose: Current antiretroviral therapies (ART) for human immunodeficiency virus (HIV) are not curative, as the virus persists in latent reservoirs, requiring lifelong adherence to ART and increasing the risk of co-morbidities. "Shock and kill" approaches to reactivate HIV from latent reservoirs followed by administration of anti-HIV drugs represent a promising strategy for eradicating latent HIV. To achieve effective shock and kill, we describe a strategy to eradicate the HIV reservoir that combines latency reversing agents (LRAs), broadly neutralizing antibodies (bnAbs), and natural killer (NK) cells. This strategy utilizes a polymer nanodepot (ND) that co-encapsulates the LRA and bnAb to reactivate latent infection and elicit enhanced cytotoxicity from co-administered NK cells. Methods: Poly(lactic-co-glycolic acid) (PLGA) NDs were synthesized using the nanoprecipitation method to co-encapsulate an LRA (TNF-α) and a bnAb (3BNC117) (TNF-α-3BNC117-NDs). ACH-2 cells were used as a cellular model of latent HIV infection. An NK92 subline, genetically modified to constitutively express the Fc receptor CD16, was administered to ACH-2 cells in combination with TNF-α-3BNC117-NDs. ACH-2 cell death and extracellular p24 were measured via flow cytometry and ELISA, respectively. Results: Stable PLGA NDs co-encapsulated TNF-α and 3BNC117 with high efficiencies and released these agents in physiological conditions. NK92 phenotype remained similar in the presence of TNF-α-3BNC117-NDs. TNF-α released from NDs efficiently reactivated HIV in ACH-2 cells, as measured by a 3.0-fold increase in the frequency of intracellular p24 positive cells. Released 3BNC117 neutralized and bound reactivated virus, targeting 57.5% of total ACH-2 cells. Critically, TNF-α-3BNC117-NDs significantly enhanced NK92 cell-mediated killing of ACH-2 cells (1.9-fold) and reduced extracellular levels of p24 to baseline. Conclusion: These findings suggest the therapeutic potential of our novel ND-based tripartite strategy to reactivate HIV from latently infected cells, generate an HIV-specific site for bnAb binding, and enhance the killing of reactivated HIV-infected target cells by NK92 cells.


Assuntos
Infecções por HIV , HIV-1 , Humanos , Infecções por HIV/tratamento farmacológico , Anticorpos Amplamente Neutralizantes/farmacologia , Anticorpos Amplamente Neutralizantes/uso terapêutico , Latência Viral , Fator de Necrose Tumoral alfa , Células Matadoras Naturais , Linfócitos T CD4-Positivos
7.
Adv Drug Deliv Rev ; 178: 113972, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34509573

RESUMO

Extracellular vesicles (EVs) have emerged as new drug delivery systems as well as a regenerative cell-free effectors going beyond academic research to reach industrial research and development (R&D). Many proof-of-concept studies are now published describing the delivery of drugs, nanoparticles or biologics among which nucleic acids, proteins, viruses, etc. Their main interests rely on their intrinsic biocompatibility, targeting capabilities and biological activities. The possibility of loading EVs with exogenous therapeutic drug/nanoparticles or imaging tracers opens up the perspectives to extend EV therapeutic properties and enable EV tracking. Clinical translation is still hampered by the difficulty to produce and load EVs with large scale, efficient and cGMP methods. In this review, we critically discuss important notions related to EV engineering and the methods available with a particular focus on technologies fitted for clinical translation. Besides, we provide a tentative data reporting frame in order to support comparability and standardization in the field.


Assuntos
Engenharia Celular , Vesículas Extracelulares/metabolismo , Sistemas de Liberação de Medicamentos , Vesículas Extracelulares/química , Humanos , Projetos de Pesquisa
8.
Adv Drug Deliv Rev ; 179: 114001, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34673131

RESUMO

Extracellular vesicles (EV) are emergent therapeutic effectors that have reached clinical trial investigation. To translate EV-based therapeutic to clinic, the challenge is to demonstrate quality, safety, and efficacy, as required for any medicinal product. EV research translation into medicinal products is an exciting and challenging perspective. Recent papers, provide important guidance on regulatory aspects of pharmaceutical development, defining EVs for therapeutic applications and critical considerations for the development of potency tests. In addition, the ISEV Task Force on Regulatory Affairs and Clinical Use of EV-based Therapeutics as well as the Exosomes Committee from the ISCT are expected to contribute in an active way to the development of EV-based medicinal products by providing update on the scientific progress in EVs field, information to patients and expert resource network for regulatory bodies. The contribution of our work group "Extracellular Vesicle translatiOn to clinicaL perspectiVEs - EVOLVE France", created in 2020, can be positioned in complement to all these important initiatives. Based on complementary scientific, technical, and medical expertise, we provide EV-specific recommendations for manufacturing, quality control, analytics, non-clinical development, and clinical trials, according to current European legislation. We especially focus on early phase clinical trials concerning immediate needs in the field. The main contents of the investigational medicinal product dossier, marketing authorization applications, and critical guideline information are outlined for the transition from research to clinical development and ultimate market authorization.


Assuntos
Desenvolvimento de Medicamentos/organização & administração , Drogas em Investigação/farmacologia , Vesículas Extracelulares/fisiologia , Técnicas de Química Analítica/métodos , Ensaios Clínicos como Assunto/organização & administração , Vias de Administração de Medicamentos , Composição de Medicamentos , Estabilidade de Medicamentos , Europa (Continente) , Humanos , Controle de Qualidade , Secretoma/fisiologia
9.
Cells ; 9(11)2020 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-33171765

RESUMO

Immune checkpoint inhibitors become a standard therapy for malignant melanoma. As immune checkpoint inhibitor monotherapies proved to have limited efficacy in significant portion of patients, it is envisaged that combination with other therapeutic modalities may improve clinical outcomes. We investigated the effect of combining photodynamic therapy (PDT) and TLR5 agonist flagellin-adjuvanted tumor-specific peptide vaccination (FlaB-Vax) on the promotion of PD-1 blockade-mediated melanoma suppression using a mouse B16-F10 implantation model. Using a bilateral mouse melanoma cancer model, we evaluated the potentiation of PD-1 blockade by the combination of peritumoral FlaB-Vax delivery and PDT tumor ablation. A photosensitizing agent, pheophorbide A (PhA), was used for laser-triggered photodynamic destruction of the primary tumor. The effect of combination therapy in conjunction with PD-1 blockade was evaluated for tumor growth and survival. The effector cytokines that promote the activation of CD8+ T cells and antigen-presenting cells in tumor tissue and tumor-draining lymph nodes (TDLNs) were also assayed. PDT and FlaB-Vax combination therapy induced efficacious systemic antitumor immune responses for local and abscopal tumor control, with a significant increase in tumor-infiltrating effector memory CD8+ T cells and systemic IFNγ secretion. The combination of PDT and FlaB-Vax also enhanced the infiltration of tumor antigen-reactive CD8+ T cells and the accumulation of migratory CXCL10-secreting CD103+ dendritic cells (DCs) presumably contributing to tumor antigen cross-presentation in the tumor microenvironment (TME). The CD8+ T-cell-dependent therapeutic benefits of PDT combined with FlaB-Vax was significantly enhanced by a PD-1-targeting checkpoint inhibitor therapy. Conclusively, the combination of FlaB-Vax with PDT-mediated tumor ablation would serve a safe and feasible combinatorial therapy for enhancing PD-1 blockade treatment of malignant melanoma.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Anticâncer/imunologia , Flagelina/farmacologia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Fotoquimioterapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Clorofila/análogos & derivados , Clorofila/farmacologia , Clorofila/uso terapêutico , Terapia Combinada , Apresentação Cruzada/efeitos dos fármacos , Humanos , Memória Imunológica , Interferon gama/metabolismo , Lipossomos , Melanoma Experimental/tratamento farmacológico , Camundongos Endogâmicos C57BL , Nanopartículas/química , Receptor de Morte Celular Programada 1/metabolismo
10.
Theranostics ; 9(9): 2505-2525, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31131050

RESUMO

The delivery of therapeutics into tumors remains a challenge in nanoparticle-mediated drug delivery. However, effective therapies such as photothermal therapy (PTT) are limited by quick systemic clearance and non-specific biodistribution. Anti-tumor strategies tailored to accommodate both tumor accumulation/retention and cellular internalization under a single platform would be a promising strategy. This work demonstrates a hierarchical activating strategy that would exhibit enhanced circulation and rapid tumor-tropism as well as facilitate tumor penetration, followed by tumor-specific drug release to realize trackable photothermal/chemotherapy. Methods: We engineered a lithocholic acid-conjugated disulfide-linked polyethyleneimine micelle (LAPMi) loaded with paclitaxel (LAPMi-PTX, L), followed by the electrostatic adsorption of indocyanine green (ICG, I) on LAPMI-PTX and subsequently coated them with thermosensitive DPPC and DSPE-PEG-NH2 lipids (L), producing Lipid/ICG/LAPMi-PTX (LIL-PTX) nanoparticles (NPs). The characteristics of NPs, including physicochemical characterization, photothermal & pH responsiveness, cell uptake, tumor spheroid penetration, anti-tumor efficacy and hierarchical activation of LIL-PTX NPs were investigated in vitro and in vivo by using CT26 cell line. The anti-metastatic potential of LIL-PTX NPs were demonstrated using 4T1 orthotopic tumor model. Results: The NPs synthesized possessed charge switchability in the mildly acidic pH, and were laser- and pH-responsive. Dual stimuli-responsive nature of LIL-PTX NPs improved the disposition of therapeutics to the tumor, reflected by enhanced intracellular uptake, tumor spheroid penetration and in vitro cytotoxicity studies. LIL-PTX NPs readily switched its surface charge from neutral to positive upon reaching the tumor milieu, thus resulting in rapid tumor tropism and accumulation. Under near-infrared laser irradiation, the thermosensitive lipids on LIL-PTX NPs were deshielded, and the tumor-penetrating LAPMi-PTX was subsequently exposed to the tumor milieu, thus resulting in enhanced intracellular internalization. Next, LAPMi-PTX evaded the endo-lysosomes, thereby releasing the PTX through the degradation of LAPMi mediated by intracellular GSH in the tumor. LIL-PTX NPs significantly improved the therapy by eradicating primary tumors completely and suppressing their subsequent lung metastasis. Conclusion: The improved therapeutic index is due to enhanced passive targeting by rapid tumor-tropic accumulation and tumor penetration by laser-driven exposure of LAPMi, thereby improving the therapeutic delivery for image-guided photothermal/chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Tratamento Farmacológico/métodos , Hipertermia Induzida/métodos , Neoplasias Experimentais/terapia , Fototerapia/métodos , Nanomedicina Teranóstica/métodos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Camundongos Endogâmicos BALB C , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Paclitaxel/farmacologia , Radioterapia Guiada por Imagem/métodos , Resultado do Tratamento
11.
ACS Appl Mater Interfaces ; 11(22): 19782-19792, 2019 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-31088067

RESUMO

Tumor adaption to hypoxic stress not only plays a crucial role in tumor malignancy but also can protect cancer cells from therapeutic interventions. Hence, therapeutic strategies attenuating tumor hypoxia in conjunction with conventional therapies may be an ideal approach. Here, we report the application of in situ oxygenic carbon nano-onion (CNO)/manganese oxide nanopods (iOCOMs) as novel theranostic photothermal transducers to neutralize the oncogenic influence of the hypoxic tumor microenvironment (TME). The developed onion-ring-shaped carbon nanoparticles or carbon nano-onions (CNOs) and iOCOM nanopods (CNO embedded in MnO2 nanosheets) were biologically stable and nontoxic and showed photothermal activity under near-infrared laser irradiation (808 nm). In addition, iOCOM assisted in the dismutation of hydrogen peroxide (H2O2), a potentially toxic reactive oxygen species that is secreted excessively by cancer cells in the hypoxic TME, resulting in in situ oxygenation and repolarization of the hypoxic TME to normoxia. The manganese ions released from iOCOM during the catalysis of H2O2 assisted in TME-responsive T1 magnetic resonance imaging (MRI). The in situ oxygenation by iOCOM in the hypoxic TME downregulated the secretion of hypoxia-inducible factor 1-α, which subsequently interfered with the cancer cell proliferation, favored tumor angiogenesis, and most importantly prevented metastatic epithelial-to-mesenchymal transition of tumor cells. Collectively, this work presents a new paradigm for antitumor strategies by targeting the tumor adaption to hypoxia in combination with photothermal therapy.


Assuntos
Hipóxia/patologia , Fototerapia/métodos , Animais , Feminino , Peróxido de Hidrogênio/química , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Compostos de Manganês/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Óxidos/química
12.
J Biomater Sci Polym Ed ; 29(7-9): 734-749, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28994338

RESUMO

Despite the introduction of many efficient post-consolidation therapies for complete relapse in leukemia patients, many patients suffer from relapse. Reactive oxygen species (ROS) are considered an important parameter in the immunosuppression of acute myeloid leukemia, where they suppress the cytotoxic action of immune cells such as NK cells and T cells. This study demonstrates a way to achieve effective inhibition of immunosuppression by loading the drug histamine dihydrochloride (HDC) onto graphene quantum dots (GQDs) using hyaluronic acid as a targeting moiety for K-562 cells. The prepared GQD-based nanoplatform was stable and achieved high drug loading on the surface, which resulted in a sustained drug release profile over a period of time. Additionally, the drug-loaded graphene nanoplatform proved to be non-toxic at higher concentrations to K-562 cells and could be effectively taken up into cells due to the targeting moiety. In vitro ROS detection assays proved that the HDC loaded graphene nanoplatform could effectively inhibit ROS and thus prevent the immunosuppression caused by leukemic cells.


Assuntos
Portadores de Fármacos/química , Grafite/química , Histamina/química , Histamina/farmacologia , Tolerância Imunológica/efeitos dos fármacos , Leucemia/imunologia , Pontos Quânticos/química , Transporte Biológico , Sobrevivência Celular/efeitos dos fármacos , Di-Hidroxifenilalanina/química , Portadores de Fármacos/metabolismo , Liberação Controlada de Fármacos , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/química , Células K562 , Leucemia/patologia , Modelos Moleculares , Conformação Molecular , Espécies Reativas de Oxigênio/metabolismo
13.
Biomaterials ; 169: 45-60, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29631167

RESUMO

Peritoneal carcinomatosis (PC) is a fatal condition arising in the gastrointestinal tract. PC patients administered drugs locally in the tumor region, such as in intraperitoneal chemotherapy (IPCh), suffer from low drug retention time and tumor penetration. Herein, we synthesized a lithocholic acid (LCA)-conjugated disulfide-linked polyethyleneimine (ssPEI) micelle (LAPMi) nanoconstruct by covalently conjugating ssPEI and LCA, thereby forming positive charged nanomicellar structures loaded with paclitaxel (PTX) (LAPMi-PTX) for IPCh. The incorporation of a positive surface charge aided in prolonging the peritoneal retention time, presumably via ascites-induced protein corona formation, and the subsequent size expansion caused resistance against undesired clearance through lymphatic openings. Furthermore, preferential tumor penetration by LAPMi-PTX is attributable to the permeation-enhancing properties of LCA, and the subsequent tumor activatable drug release was induced by the presence of disulfide linkages. By integrating these properties, LAPMi exhibited prolonged peritoneal residence time, enhanced tumor permeation and chemotherapeutic effect evidenced by in vitro, tumor spheroid and in vivo studies. Importantly, our strategy enabled significant PC inhibition and increased the overall survival rate of tumor-bearing mice. In conclusion, we provided a new paradigm of intractable PC treatment by enabling the prolonged residence time of the nanoconstruct, thereby enhancing tumor penetration and anti-tumor therapy.


Assuntos
Antineoplásicos Fitogênicos , Carcinoma/tratamento farmacológico , Portadores de Fármacos , Paclitaxel , Neoplasias Peritoneais/tratamento farmacológico , Peritônio/metabolismo , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacocinética , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética
14.
Biomaterials ; 180: 240-252, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30055399

RESUMO

Multifunctional nanoparticles integrating cancer cell imaging and treatment modalities into a single platform are recognized as a promising approach; however, their development currently remains a challenge. In this study, we synthesized magnetic field-inducible drug-eluting nanoparticles (MIDENs) by embedding superparamagnetic iron oxide nanoparticles (Fe3O4; SPIONs) and cancer therapeutic drugs (doxorubicin; DOX) in a temperature-responsive poly (lactic-co-glycolic acid) (PLGA) nanomatrix. Application of an external alternating magnetic field (AMF) generated heat above 42 °C and subsequent transition of the PLGA polymer matrix (Tg = 42-45 °C) from the glassy to the rubbery state, facilitating the controlled release of the loaded DOX, ultimately allowing for simultaneous hyperthermia and local heat-triggered chemotherapy for efficient dual cancer treatment. The average size of the synthesized MIDENs was 172.1 ±â€¯3.20 nm in diameter. In vitro studies showed that the MIDENs were cytocompatible and especially effective in destroying CT26 colon cancer cells with AMF application. In vivo studies revealed that the MIDENs enabled enhanced T2 contrast magnetic resonance imaging and a significant suppression of malignant tumor growth under an AMF. Our multifunctional MIDENs, composed of biocompatible substances and therapeutic/imaging modalities, will be greatly beneficial for cancer image-guided thermo-chemotherapy applications.


Assuntos
Doxorrubicina/química , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Campos Magnéticos , Nanopartículas de Magnetita/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Hipertermia Induzida
15.
J Control Release ; 280: 20-30, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29723613

RESUMO

In this study, we developed novel hyaluronic acid cross-linked zein nanogels (HA-Zein NGs) to deliver the potential anticancer agent curcumin (CRC), a naturally occurring phytochemical drug in cancer cells. In vitro studies showed that they are highly compatible with the tested cell lines. They showed CD44 specific uptake in CT26 cell line more than by the CD44 receptor pre-inhibited CT26 cells. The CRC encapsulated HA-Zein NGs (HA-Zein-CRC NGs) found to exert a specific toxicity against CT26 sparing healthy normal fibroblast cells in vitro. The apoptotic effects were further confirmed with flow cytometry showing that the HA-Zein-CRC NGs exhibited high anticancer activity against the CT26 cells. The in vivo bio-distribution with a CT26 tumor model showed their high tumor accumulation thereby improved antitumor efficacy with a low dosage of CRC, compared to the previous reports. Thus, the preclinical studies clearly showed that these novel HA-Zein NGs would be highly beneficial in encapsulating hydrophobic drugs with improved pharmacokinetics thereby enhancing the therapeutic outcomes.


Assuntos
Curcumina/química , Portadores de Fármacos/química , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/química , Nanopartículas/química , Zeína/química , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Materiais Biocompatíveis/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Reagentes de Ligações Cruzadas/química , Curcumina/uso terapêutico , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Camundongos Endogâmicos BALB C , Terapia de Alvo Molecular/métodos , Polimerização , Distribuição Tecidual
16.
J Control Release ; 276: 72-83, 2018 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-29499218

RESUMO

Stimuli-responsive polymeric nanoparticles are useful for overcoming challenges such as transfection efficiency and the specific and safe delivery of genes to cancer cells. Transfection outcomes can be improved through spatially and temporally controlled gene release. We formulated a nanoassembly comprising a disulfide-crosslinked polyethylenimine (ssPEI) conjugated with a tumor-specific cell-penetrating peptide (DS 4-3) (SPD) polyplex and bovine serum albumin (BSA)-loaded IR780 (BI) nanoparticle, thereby forming a dual-stimulus-triggered, tumor-penetrating and gene-carrying nanoassembly (BI-SPD) via electrostatic complexing. BI-SPD nanoassembly were composed of highly stable nanosized complexes with an average size of 457 ±â€¯27.5 nm, exhibiting an up to two-fold enhanced transfection efficiency with no sign of potential cytotoxicity in breast cancer cells. Moreover, upon laser irradiation, a four-fold increase in transfection efficiency was achieved due to the rapid endosomal escape of polyplexes triggered by the local heat induced by the BI-SPD nanoassembly. Additionally, the high redox environment in tumor cells facilitated the disassembly of the SPD polyplex for efficient plasmid release in the cytosol. The BI-SPD nanoassembly also exhibited high penetration and enhanced photothermally triggered gene expression in the 4T1 spheroid model. This BI-SPD nanoassembly has the potential to enhance the expression of therapeutic genes in tumor models without causing significant toxicity to surrounding healthy tissues, since it has shown higher tumor targeting and accumulation in the 4T1 tumor in mice model.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , DNA/administração & dosagem , Nanopartículas/administração & dosagem , Polietilenoimina/administração & dosagem , Soroalbumina Bovina/administração & dosagem , Animais , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/farmacocinética , Corantes/administração & dosagem , Corantes/farmacocinética , DNA/farmacocinética , Dissulfetos , Técnicas de Transferência de Genes , Indóis/administração & dosagem , Indóis/farmacocinética , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos Endogâmicos BALB C , Plasmídeos , Polietilenoimina/farmacocinética , Soroalbumina Bovina/farmacocinética
18.
Nanomaterials (Basel) ; 6(4)2016 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28335204

RESUMO

Nanotechnology has enabled the development of many alternative anti-cancer approaches, such as thermal therapies, which cause minimal damage to healthy cells. Current challenges in cancer treatment are the identification of the diseased area and its efficient treatment without generating many side effects. Image-guided therapies can be a useful tool to diagnose and treat the diseased tissue and they offer therapy and imaging using a single nanostructure. The present review mainly focuses on recent advances in the field of thermal therapy and imaging integrated with multifunctional inorganic nanoparticles. The main heating sources for heat-induced therapies are the surface plasmon resonance (SPR) in the near infrared region and alternating magnetic fields (AMFs). The different families of inorganic nanoparticles employed for SPR- and AMF-based thermal therapies and imaging are described. Furthermore, inorganic nanomaterials developed for multimodal therapies with different and multi-imaging modalities are presented in detail. Finally, relevant clinical perspectives and the future scope of inorganic nanoparticles in image-guided therapies are discussed.

19.
Biomed Res Int ; 2015: 959175, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26078971

RESUMO

Today, nanotechnology plays a vital role in biomedical applications, especially for the diagnosis and treatment of various diseases. Among the many different types of fabricated nanoparticles, magnetic metal oxide nanoparticles stand out as unique and useful tools for biomedical applications, because of their imaging characteristics and therapeutic properties such as drug and gene carriers. Polymer-coated magnetic particles are currently of particular interest to investigators in the fields of nanobiomedicine and fundamental biomaterials. Theranostic magnetic nanoparticles that are encapsulated or coated with polymers not only exhibit imaging properties in response to stimuli, but also can efficiently deliver various drugs and therapeutic genes. Even though a large number of polymer-coated magnetic nanoparticles have been fabricated over the last decade, most of these have only been used for imaging purposes. The focus of this review is on polysaccharide-coated magnetic nanoparticles used for imaging and gene delivery.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Nanopartículas de Magnetita/uso terapêutico , Nanotecnologia , Diagnóstico por Imagem/métodos , Humanos , Polímeros/química , Polímeros/uso terapêutico , Polissacarídeos/química , Polissacarídeos/uso terapêutico
20.
Curr Pharm Des ; 21(29): 4285-309, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26323422

RESUMO

Chitosan on its own is a well-established natural polymer and is widely regarded as a biodegradable, biocompatible and nontoxic material for drug delivery applications. Although unmodified chitosan has some mucoadhesive properties on its own, its bioavailability is limited due to its short retention time in the body. Moreover, the high solubility of chitosan at acidic pH levels limits its use for mucosal drug delivery (especially through the oral route). Chemically-modified mucoadhesive chitosan, especially thiolated chitosan, has arisen as an alternative to create novel mucosal drug delivery systems. The mucoadhesive properties that are conferred to the thiolated chitosan certainly set this novel class of second or third-generation thiomers apart. To understand the significance of mucoadhesive chitosan, we first present the mechanism of mucoadhesion and provide comprehensive coverage of description of a variety of chemical modifications to prepare mucoadhesive thiolated chitosan derivatives. We then present the plethora of applications of these modified chitosan variants in a wide range of drug delivery fields, including the delivery of antigens, proteins and genes through a variety of routes, including oral, nasal, pulmonary, vaginal and others. By presenting the range of applications for mucoadhesive chitosan drug carriers we herein demonstrate that chemically-modified thiolated chitosan is a versatile and effective material for a new class of drug delivery vehicles.


Assuntos
Quitosana/farmacocinética , Portadores de Fármacos/farmacocinética , Mucosa/metabolismo , Adesividade , Animais , Quitosana/análogos & derivados , Sistemas de Liberação de Medicamentos/métodos , Humanos , Mucinas/metabolismo , Mucosa/efeitos dos fármacos , Nanoestruturas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA