Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Small ; 18(27): e2201585, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35644863

RESUMO

To overcome the autophagy compromised mechanism of protective cellular processes by "eating"/"digesting" damaged organelles or potentially toxic materials with autolysosomes in tumor cells, lysosomal impairment can be utilized as a traditional autophagy dysfunction route for tumor therapy; however, this conventional one-way autophagy dysfunction approach is always limited by the therapeutic efficacy. Herein, an innovative pharmacological strategy that can excessively provoke autophagy via endoplasmic reticulum (ER) stress is implemented along with lysosomal impairment to enhance autophagy dysfunction. In this work, the prepared tellurium double-headed nanobullets (TeDNBs) with controllable morphology are modified with human serum albumin (HSA) which facilitates internalization by tumor cells. On the one hand, ER stress can be stimulated by upregulating the phosphorylation eukaryotic translation initiation factor 2 (P-eIF2α) owing to the production of tellurite (TeO32- ) in the specifical hydrogen peroxide-rich tumor environment; thus, autophagy overstimulation occurs. On the other hand, OME can deacidify and impair lysosomes by downregulating lysosomal-associated membrane protein 1 (LAMP1), therefore blocking autolysosome formation. Both in vitro and in vivo results demonstrate that the synthesized TeDNBs-HSA/OME (TeDNBs-HO) exhibit excellent therapeutic efficacy by autophagy dysfunction through ER stress induction and lysosomal damnification. Thus, TeDNBs-HO is verified to be a promising theranostic nanoagent for effective tumor therapy.


Assuntos
Lisossomos , Telúrio , Autofagia , Estresse do Retículo Endoplasmático , Humanos , Lisossomos/metabolismo , Fosforilação
2.
Small ; 18(45): e2203942, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36156383

RESUMO

As a promising therapeutic strategy against cancer, immunotherapy faces critical challenges, especially in solid tumors. Immune checkpoint blockade therapy, particularly blocking the interaction of the programmed cell death 1 (PD1)-PD1 ligand 1 (PD-L1) axis, can reverse the suppression of T cells so as to destroy tumor cells and exert antitumor effects. Here, a strategy of multiple activation of immune pathways is developed, to provide supporting evidence for potential antitumor therapies. Briefly, a pH/glutathione responsive drug-loading hollow-manganese dioxide (H-MnO2 )-based chlorine6 (Ce6)-modified DNAzyme therapeutic nanosystem for the combination of gene therapy and immunotherapy is established. The H-MnO2 nanoparticles could efficiently deliver the DNAzyme and glycyrrhizic acid (GA) to enhance the tumor target effects. In the tumor microenvironments, the biodegradation of H-MnO2 via pH-induced hydrolyzation allows the release of guest DNAzyme payloads and host Mn2+ ions, which serve as PD-L1 mRNA-targeting reagent and require DNAzyme cofactors for activating gene therapy. In addition, Mn2+ is also associated with the immune activation of thcGAS-STING pathway. Auxiliary photosensitizers Ce6 and GA could produce reactive oxygen species, resulting in immunogenic cell death. Overall, this study provides a general strategy for targeted gene inhibition and GA release, which is valuable for the development of potential tumor immunotherapies.


Assuntos
DNA Catalítico , Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Compostos de Manganês , Antígeno B7-H1 , DNA Catalítico/metabolismo , Óxidos , Fotoquimioterapia/métodos , Sistemas de Liberação de Fármacos por Nanopartículas , Neoplasias/terapia , Imunoterapia/métodos , Microambiente Tumoral , Linhagem Celular Tumoral
3.
Mol Pharm ; 16(8): 3489-3501, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31246475

RESUMO

Recently, multifunctional clearable inorganic theranostic nanoparticles have been attracting more and more attention. Protein-based nanoparticles can be cleared by the hepatobiliary system efficiently. In this work, ultrasmall gadolinium oxide (Gd2O3) nanoparticles, which possess the advantage of high longitudinal relaxation rate, were coated with bovine serum albumin (BSA). After the Gd2O3/BSA nanoparticles were linked with two-dimensional photothermal MoS2 nanomaterials, the nanoparticles were also modified with hyaluronic acid (HA) through the disulfide bonds for tumor-targeting effect. As indicated by in vitro and in vivo studies, these Gd2O3/BSA@MoS2-HA nanoparticles could be rapidly degraded and excreted after reacting with glutathione (GSH) by the redox response, thus avoiding long-term toxicity. In addition, the cellular uptake study and in vivo multispectral optoacoustic tomography (MSOT), X-ray computed tomography (CT), and magnetic resonance (MR) triple-modal images demonstrated that Gd2O3/BSA@MoS2-HA nanoparticles exhibited a high tumor uptake effect after intravenous injection. Consequently, such clearable theranostic nanoparticles with multiple functions, which are applicable in multimodal imaging-guided cancer therapy, might show promise for applications in nanomedical science.


Assuntos
Gadolínio/administração & dosagem , Imagem Multimodal/métodos , Nanopartículas/administração & dosagem , Neoplasias/terapia , Nanomedicina Teranóstica/métodos , Animais , Linhagem Celular Tumoral/transplante , Terapia Combinada/métodos , Modelos Animais de Doenças , Feminino , Gadolínio/química , Gadolínio/farmacocinética , Glutationa/metabolismo , Humanos , Hipertermia Induzida/métodos , Injeções Intravenosas , Imageamento por Ressonância Magnética/métodos , Taxa de Depuração Metabólica , Camundongos , Nanopartículas/química , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Tamanho da Partícula , Técnicas Fotoacústicas/métodos , Fototerapia/métodos , Soroalbumina Bovina/química , Distribuição Tecidual , Tomografia Computadorizada por Raios X/métodos
4.
Adv Healthc Mater ; 13(14): e2303683, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38386961

RESUMO

Employing tumor whole cells for tumor immunotherapy is a promising tumor therapy proposed in the early stage, but its therapeutic efficacy is weakened by the methods of eliminating pathogenicity and the mass ratio of the effective antigen carried by itself. Here, by adding gold ion to live cancer cells in the microfluidic droplets, this work obtains dead tumor whole cells with NIR-controlled catalytic ability whose pathogenicity is removed while plenary tumor antigens, major structure, and homing ability are reserved. The engineered tumor cell (Cell-Au) with the addition of prodrug provides 1O2 in an O2-free Russell mechanism, which serves better in a hypoxic tumor microenvironment. This tumor whole-cell catalytic vaccine (TWCV) promotes the activation of dendritic cells and the transformation of macrophages into tumor suppressor phenotype. In 4T1 tumor-bearing mice, the Cell-Au-based vaccine supports the polarization of cytotoxicity T cells, resulting in tumor eradication and long-term animal survival. Compared with antigen vaccines or adoptive cell therapy which takes months to obtain, this TWCV can be prepared in just a few days with satisfactory immune activation and tumor therapeutic efficacy, which provides an alternative way for the preparation of personalized tumor vaccines across tumor types and gives immunotherapy a new path.


Assuntos
Vacinas Anticâncer , Ouro , Imunoterapia , Animais , Ouro/química , Imunoterapia/métodos , Camundongos , Linhagem Celular Tumoral , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/química , Camundongos Endogâmicos BALB C , Catálise , Feminino , Microambiente Tumoral/imunologia , Nanopartículas Metálicas/química , Células Dendríticas/imunologia , Humanos , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/patologia
5.
ACS Nano ; 18(12): 9031-9042, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38470458

RESUMO

Cuproptosis has drawn enormous attention in antitumor material fields; however, the responsive activation of cuproptosis against tumors using nanomaterials with high atom utilization is still challenging. Herein, a copper-based nanoplatform consisting of acid-degradable copper hydride (CuH) nanoparticles was developed via a microfluidic synthesis. After coating with tumor-targeting hyaluronic acid (HA), the nanoplatform denoted as HA-CuH-PVP (HCP) shows conspicuous damage toward tumor cells by generating Cu+ and hydrogen (H2) simultaneously. Cu+ can induce apoptosis by relying on Fenton-like reactions and lead to cuproptosis by causing mitochondrial protein aggregation. Besides, the existence of H2 can enhance both cell death types by causing mitochondrial dysfunction and intracellular redox homeostatic disorders. In vivo experimental results further exhibit the desirable potential of HCP for killing tumor cells and inhibiting lung metastases, which will broaden the horizons of designing copper-based materials triggering apoptosis and cuproptosis for better antitumor efficacy.


Assuntos
Cobre , Nanopartículas , Microfluídica , Apoptose , Ácido Hialurônico , Hidrogênio
6.
Acta Pharm Sin B ; 14(2): 795-807, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38322334

RESUMO

Recent innovations in nanomaterials inspire abundant novel tumor-targeting CRISPR-based gene therapies. However, the therapeutic efficiency of traditional targeted nanotherapeutic strategies is limited by that the biomarkers vary in a spatiotemporal-dependent manner with tumor progression. Here, we propose a self-amplifying logic-gated gene editing strategy for gene/H2O2-mediated/starvation multimodal cancer therapy. In this approach, a hypoxia-degradable covalent-organic framework (COF) is synthesized to coat a-ZIF-8 in which glucose oxidase (GOx) and CRISPR system are packaged. To intensify intracellular redox dyshomeostasis, DNAzymes which can cleave catalase mRNA are loaded as well. When the nanosystem gets into the tumor, the weakly acidic and hypoxic microenvironment degrades the ZIF-8@COF to activate GOx, which amplifies intracellular H+ and hypoxia, accelerating the nanocarrier degradation to guarantee available CRISPR plasmid and GOx release in target cells. These tandem reactions deplete glucose and oxygen, leading to logic-gated-triggered gene editing as well as synergistic gene/H2O2-mediated/starvation therapy. Overall, this approach highlights the biocomputing-based CRISPR delivery and underscores the great potential of precise cancer therapy.

7.
ACS Nano ; 18(11): 7923-7936, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38445625

RESUMO

Tumor whole cell, carrying a complete set of tumor-associated antigens and tumor-specific antigens, has shown great potential in the construction of tumor vaccines but is hindered by the complex engineering means and limited efficacy to cause immunity. Herein, we provided a strategy for the self-mineralization of autologous tumor cells with palladium ions in microfluidic droplets, which endowed the engineered cells with both immune and catalytic functions, to establish a bioorthogonally catalytic tumor whole-cell vaccine. This vaccine showed strong inhibition both in the occurrence and recurrence of tumor by invoking the immediate antitumor immunity and building a long-term immunity.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Microfluídica , Imunoterapia , Neoplasias/terapia , Antígenos de Neoplasias
8.
Biomaterials ; 291: 121871, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36323073

RESUMO

Effective activation of cGAS-STING pathway combined with immune checkpoint blockade (ICB) within the immunosuppressive tumor microenvironment to induce stronger immune responsiveness yet remains challenging. CRISPR-Cas9 gene editing technology, which offers the benefits of permanence and irreversibility, could recognize the target genome sequence with sgRNA (Guide RNA) and guide the Cas9 protease to knock down the target gene. Herein, a nanoplatform (HMnMPH) for dual activation of cGAS-STING pathway in combination with CRISPR-Cas9 gene editing to silence programmed death ligand 1 (PD-L1) to trigger long-term immunotherapy was reported. The HMnMPH consists of hollow manganese dioxide (HMn) loaded with STING agonist (MSA-2) and CRISPR-Cas9/sg-PD-L1 plasmid with further modification of hyaluronic acid (HA). In acidic and GSH overexpressed tumor environment, HMnPMH was degraded to release large amounts of Mn ions and STING agonists, strongly and persistently activating the cGAS-STING pathway to promote the release of type I interferon and pro-inflammatory factors. Meanwhile, the released CRISPR-Cas9 plasmid could knockdown the PD-L1 immune checkpoint and restart immunosuppressive T cells to differentiate into cytotoxic T lymphocytes significantly, which reduced the activity of primary and distal tumors and demonstrated a long-term immune memory effect on distal tumors.


Assuntos
Edição de Genes , Neoplasias , Humanos , Antígeno B7-H1/metabolismo , Sistemas CRISPR-Cas/genética , Imunoterapia , Neoplasias/genética , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Microambiente Tumoral
9.
Biomaterials ; 257: 120236, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32738655

RESUMO

Recent years have witnessed the blooming of gas therapy nanoplatforms, which emerged as a promising area for cancer therapy. However, uncontrolled or inadequate generation of gas and unclear therapeutic mechanisms, which were still regarded as big challenges to apply gas therapy into clinical. Here in, a gas treatment based on sulfur dioxide (SO2) prodrug doped nanorattles was explored, which could not only inhibit superficial tumor but also deep tumor. A Benzothiazole sulfinate (BTS, a water-soluble SO2 prodrug) doped rattle-structured rough nanocapsule with high drug payload (~80%) composed of gold nanorods cores and polydopamine (PDA) shell (GNRs@PDA-BTS, GPBRs) has been prepared. Taking advantages of excellent photothermal conversion ability as well as acidic condition in the tumor sites, SO2 gas release could be precisely controlled by both photothermal and pH, thus realizing "collusion inside" gas therapy and "outside" photothermal therapy. In addition, the cytotoxic SO2 was found to induce cell apoptosis accompanied by the upregulation of intracellular reactive oxygen species (ROS) levels and modulation of apoptosis-relative proteins such as p53, bcl-2, Bax and caspase-3. Such photothermal/pH triggered SO2 gas therapy may provide an effective strategy to stimulate further development of deep tumor therapy.


Assuntos
Nanotubos , Pró-Fármacos , Ouro , Concentração de Íons de Hidrogênio , Fototerapia , Polímeros
10.
Biomaterials ; 250: 120076, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32361390

RESUMO

Nowadays, limited deep tumor penetration and lower therapeutic effect are still the major obstacle in nanomedicines for cancer therapy. Here, we developed high-efficiency nanocomposites, SO2 prodrug (BTS) loaded Au-Ag hollow nanotriangles (Au-Ag-BTS HTNs), which could not only synergistically upregulate Bax expression in mitochondria, but also be triggered by acidic tumor microenvironment to generate SO2 for deep tumor therapy. Upon NIR laser irradiation, Au-Ag hollow nanotriangles (Au-Ag HTNs) produced plenty of heat for photothermal therapy (PTT), while the acidic condition in tumor cells induced on-demand SO2 release from BTS for deep tumor therapy. More importantly, the combined therapy could simultaneously upregulate the expression of apoptosis factor Bax as well as downregulate Bcl-2 in mitochondria, which would induce an increase of Caspase-3 expression to accelerate the apoptosis of tumor cells, thereby achieving a win-win cooperation. The results indicated that enhanced deep tumor therapeutic effect based on Au-Ag-BTS HTNs was realized in vitro and in vivo. Such pH-triggered SO2 therapy offered a novel strategy for responding tumor microenvironment and improving penetration and heterogeneity distribution of nanotherapeutics in tumor.


Assuntos
Neoplasias , Pró-Fármacos , Ouro , Humanos , Mitocôndrias , Neoplasias/tratamento farmacológico , Prata , Microambiente Tumoral , Proteína X Associada a bcl-2
11.
Biomater Sci ; 8(7): 1981-1995, 2020 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-32068203

RESUMO

Improving the generation of reactive oxygen species (ROS) while consuming glutathione (GSH) is the main method for amplifying intracellular oxidative stress. However, in previous studies, it was normally necessary to combine two or more materials to achieve the effect of destroying the intracellular redox homeostasis. This made the preparation process relatively complicated. Herein, we designed ultra-small bismuth sulfide quantum dot (Bi2S3 QD)-doped hollow mesoporous Prussian blue (HMPB) (HMPB/Bi2S3) nanocubes for amplified tumor oxidative stress to augment photo-/radiotherapy. In addition to being photothermal materials, Prussian blue can be used as both a catalyst for the Fenton reaction and a consumer of GSH due to the multivalent state of iron. Ferrous ions (Fe(ii)) can produce toxic ROS-hydroxyl radicals (˙OH) with abundant hydrogen peroxide in the tumor cells by the Fenton reaction. Meanwhile, ferric ions (Fe(iii)) can oxidize the intracellular GSH to GSSG, thus depleting the concentration of GSH inside tumors. As a result, oxidative stress imbalance could be induced by the reversible redox property of Fe(ii/iii), thereby causing DNA damage and increasing the cell membrane permeability to realize enhanced photo-/radiotherapy. As a sensitizer for radiotherapy, ultra-small Bi2S3 QDs (3-5 nm) are doped in HMPB, thus improving the therapeutic effect by prolonging blood circulation and reducing systemic toxicity via kidney metabolism. Therefore, such a reversible HMPB/Bi2S3 nanocube is a promising therapeutic agent for amplified tumor oxidative stress to augment photo-/radiotherapy, which might show further applications in nanomedical science.


Assuntos
Bismuto/química , Neoplasias da Mama/terapia , Ferrocianetos/química , Ferro/química , Fototerapia/métodos , Radioterapia/métodos , Sulfetos/química , Animais , Neoplasias da Mama/metabolismo , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Células MCF-7 , Camundongos , Nanoestruturas , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Pontos Quânticos , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Biomaterials ; 254: 120140, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32473481

RESUMO

Due to the highly complex biological formation procedure, tumor is still difficult to be treated efficiently and always associated with proliferation, migration and inflammation during treatment. Herein, a novel strategy of boosted photocatalytic activity induced NAMPT-regulating therapy is used for tumors inhibition based on FK866 loaded bismuth-humic acids heterojunction (Bi-HA/FK866). With the reduction function of HA, Bi (Ⅲ) can be reduced to elemental Bi, which can be excited by NIR laser to form electron-hole pair due to the narrow bandgap. Moreover, the coated HA and Bi could form a heterojunction structure, which could decrease the electron-hole recombination, and further boost the photocatalytic activity, leading to highly efficient ROS generation and GSH depletion. The resulted ROS could induce DNA damage of the tumor cells, thus enhancing the sensitivity to the inhibitor of NAMPT (FK866) to downregulate NAD/ERK/NF-κB signal pathways, and eventually simultaneously prevent cancer progression. Moreover, the decreased NAD could downregulate NADPH and further suppress the innate antioxidant defense system by inhibiting reduction of GSSG. The boosted photocatalytic activity induced NAMPT-regulating therapy offers a promising way to address the important issue of penetration depth limitation induced cancer relapse and migration, providing more possibilities toward successful clinical application.


Assuntos
Bismuto , Neoplasias , Acrilamidas , Proliferação de Células , Citocinas , Humanos , Substâncias Húmicas , Inflamação , Neoplasias/tratamento farmacológico , Piperidinas
13.
Theranostics ; 9(14): 3992-4005, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281527

RESUMO

Rod-shape nanoplatform have received tremendous attention owing to their enhanced ability for cell internalization and high capacity for drug loading. MoS2, widely used in electronic devices, electrocatalysis, sensor and energy-storage, has been studied as photothermal agents over the years. However, the efficacy of rod-shape MoS2 based photothermal agents for photothermal therapy has not been studied before. Here, a near-infrared (NIR) light-absorbing MoS2 nanosheets coated mesoporous silica nanorods with human serum albumin (HSA) modifying and Ce6 loading (MSNR@MoS2-HSA/Ce6) were constructed for combined photothermal and photodynamic therapy. Methods: The near-infrared (NIR) light was used to trigger the synergistic anti-tumor therapy. In addition, breast cancer cell line was applied to evaluate the in vitro anti-tumor activity. The multi-modal imaging capacity and tumor-killing efficiency of the designed nanocomposites in vivo was also demonstrated with the 4T1 tumor-bearing mouse model. Results: These nanocomposites could not only perform NIR light triggered photodynamic therapy (PDT) and photothermal therapy (PTT), but also achieve in vivo fluorescence (FL) /multispectral optical tomography (MSOT)/X-ray computed tomography (CT) triple-model bioimaging. What's more, the rod-shape nanoplatform could be endowed with better anti-tumor ability based on the EPR effect and HSA-mediated active tumor targeting. At the same time, the hyperthermia generated by MoS2 could synergistically improve the PDT effect with the acceleration of the blood flow, leading to the increase of the oxygen level in tumor tissue. Conclusion: MSNR@MoS2-HSA/Ce6 proves to be a promising multi-functional nanoplatform for effective treatment of tumor.


Assuntos
Molibdênio/química , Nanotubos/química , Fotoquimioterapia/métodos , Fototerapia/métodos , Dióxido de Silício/química , Tomografia Computadorizada por Raios X/métodos , Animais , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Feminino , Fluorescência , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Espécies Reativas de Oxigênio/metabolismo , Oxigênio Singlete/metabolismo
14.
ACS Appl Mater Interfaces ; 11(7): 6777-6788, 2019 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-30668088

RESUMO

Recently, rodlike nanomaterials with specific aspect ratio for efficient cellular uptake have received enormous attention. For functional nanomaterials, such as photothermal agents, large surface areas for their rod-shaped exterior that increase the amount of light absorbed would lead to a higher absorption coefficient as well as drug-loading property. In this project, we coated rodlike mesoporous silica with gold nanoshells (MSNR@Au hybrid), modifying them with ultrasmall gadolinium (Gd)-chelated supramolecular photosensitizers, TPPS4 (MSNR@Au-TPPS4(Gd)), which could be applied to near-infrared fluorescence/multispectral optoacoustic tomography/computed tomography/magnetic resonance imaging and imaging-guided remotely controlled photothermal (PTT)/photodynamic (PDT) combined antitumor therapy. Gold nanoshells, as a perfect PTT agent, were used to assemble the rodlike mesoporous silica nanoparticles with larger superficial area and higher drug loading, thus obtaining the MSNR@Au hybrid. HS-ß-CD, which was used as the host, was adsorbed on the gold nanoshell (MSNR@Au-ß-CD) to link TPPS4(Gd) through the host-guest reaction, thus forming CD-TPPS4 supramolecular photosensitizers (supraPSs). Compared with conventional PSs, supraPSs have host screens, which could reduce the self-aggregation of TPPS4, and consequently generate 1O2 with high efficiency. The in vivo quadmodal imaging of MSNR@Au-TPPS4(Gd) nanoparticles revealed an intensive tumor uptake effect after injection. The in vivo antitumor efficacy further testified that the synergistic therapy, which was more efficient than any other monotherapy, exhibited an excellent tumor inhibition therapeutic effect. As a result, this encourages to further explore multifunctional theranostic nanoparticles based on gold shells for combined cancer therapy.


Assuntos
Meios de Contraste , Ouro , Hipertermia Induzida/métodos , Nanoconchas , Neoplasias Experimentais , Fotoquimioterapia , Fármacos Fotossensibilizantes , Dióxido de Silício , Tomografia Computadorizada por Raios X , Animais , Linhagem Celular Tumoral , Meios de Contraste/química , Meios de Contraste/farmacocinética , Meios de Contraste/farmacologia , Ouro/química , Ouro/farmacocinética , Ouro/farmacologia , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanoconchas/química , Nanoconchas/uso terapêutico , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/terapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacocinética , Fármacos Fotossensibilizantes/farmacologia , Dióxido de Silício/química , Dióxido de Silício/farmacocinética , Dióxido de Silício/farmacologia , Nanomedicina Teranóstica/métodos
15.
Nanoscale ; 11(13): 6270-6284, 2019 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-30882830

RESUMO

Radiotherapy (RT) is one of the most widely applied treatments for cancer therapy in clinics. Herein, we constructed innovative multifunctional nanotheranostic MnO2-mSiO2@Au-HA nanoparticles (MAHNPs) based on one-pot MnO2-mSiO2 nanohybrids (MNHs) and gold nanoparticles (AuNPs) for multispectral optoacoustic tomography (MSOT)/computed tomography (CT) and magnetic resonance (MR) imaging-guided hypoxia-maneuvered radiotherapy. The MNHs were prepared via a facile one-pot approach, which avoided the leakage of MnO2 nanoparticles and increased the synthetic efficiency. The Mn2+ ions triggered the breakdown of endogenous H2O2 to generate O2 to convert the hypoxic tumor micro-environment (TME), thus enhancing radiotherapy by self-supply oxygen. In addition, hyaluronic acid (HA) was employed to modify the surface of the MnO2-mSiO2@Au nanoparticles to improve their biocompatibility and cellular uptake. The well-designed nanoparticles could perform remarkable photothermal therapy (PTT) and hypoxia-maneuvered radiotherapy (RT) simultaneously and MSOT/CT/MR imaging. The in vivo studies showed that the MAHNPs achieved almost total suppression of tumor growth without observable recurrence, which raises new possibilities for clinical nanotheranostics with multimodal diagnostic and therapeutic coalescent design.


Assuntos
Hipóxia Celular , Ouro/química , Compostos de Manganês/química , Nanopartículas/química , Óxidos/química , Dióxido de Silício/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Feminino , Hemólise/efeitos dos fármacos , Ácido Hialurônico/química , Peróxido de Hidrogênio/química , Raios Infravermelhos , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Nanopartículas/metabolismo , Nanopartículas/uso terapêutico , Nanopartículas/toxicidade , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/terapia , Fototerapia , Distribuição Tecidual , Tomografia Computadorizada por Raios X
16.
J Mater Chem B ; 7(15): 2499-2511, 2019 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32255127

RESUMO

Photothermal therapy (PTT) has shown promising potential and bright prospects in damaging primary tumors; however, it is limited to metastatic and recrudescent tumors as PTT requires straightforward light irradiation. Moreover, metastatic and recrudescent tumor immunosuppression due to host T-cell antitumor activity is dramatically impeded because of programmed cell death 1 ligand (PD-L1) and programmed cell death receptor 1 (PD-1) pathways and immune checkpoint blockade (ICB) therapy. In this work, we demonstrate that PTT combined with ICB could not only eliminate primary tumors, but also prevent tumor metastasis to the lungs/liver. In particular, we have designed immunoadjuvant nanomedicine carriers on the basis of polydopamine (PDA) simultaneously loaded with resiquimod (R848)-a kind of toll-like receptor 7 (TLR7) agonist-and carbon dots (CDs)-a fluorescent agent. This nanomedicine is defined as PDA-PEG-R848-CD nanoparticle (NP). The multitasking PDA-PEG-R848-CD NPs can destroy 4T1 breast tumors by PTT under near-infrared laser irradiation in addition to generating tumor-associated antigens. Moreover, the PTT effect triggered the release of R848, thereby inducing a strong antitumor immune response. Meanwhile, this synergistic therapy also shows the abscopal effects by completely inhibiting the growth of untreated distant tumors by effectively triggering the tumors infiltrated by CD3/CD8. Such findings suggest that PDA-PEG-R848-CD NPs could significantly potentiate the systemic therapeutic efficiency of PD-L1 checkpoint blockade therapy by activating both innate and adaptive immune systems in the body.


Assuntos
Antígeno B7-H1/metabolismo , Portadores de Fármacos/química , Imunoterapia , Indóis/química , Nanomedicina/métodos , Fototerapia , Polímeros/química , Temperatura , Animais , Antígeno B7-H1/imunologia , Carbono/química , Linhagem Celular Tumoral , Imidazóis/química , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Camundongos , Nanopartículas/química , Metástase Neoplásica , Polietilenoglicóis/química
17.
ACS Appl Mater Interfaces ; 11(6): 5771-5781, 2019 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-30653297

RESUMO

Recently, we developed ultrasmall molybdenum disulfide (MoS2) quantum dots for computed tomography (CT) and multispectral optoacoustic tomography (MSOT) imaging-guided photothermal therapy (PTT). But, due to rapid body elimination and limited blood circulation time, the tumor uptake of the dots is low. In our study, this problem was solved via designing an amino-modified biodegradable nanomaterial based on MoS2 quantum-dots-doped disulfide-based SiO2 nanoparticles (denoted MoS2@ss-SiO2) for multimodal application. By integrating the MoS2 quantum dots into clearable SiO2 nanoparticles, this nanoplatform with an appropriate particle size can not only degrade and excrete in a reasonable period induced by redox responsiveness of glutathione but also exhibit a high tumor uptake due to the longer blood circulation time. Moreover, hyaluronic acid and chlorin e6 (Ce6) were adsorbed on the outer shell for tumor-targeting effect and photodynamic therapy, respectively. So, this biodegradable and clearable theranostic nanocomposite, which is applicable in integrated fluorescence/CT/MSOT imaging-guided combined photothermal therapy (PTT) and photodynamic therapy, is very promising in biomedical applications in the future.


Assuntos
Dissulfetos/química , Molibdênio/química , Nanocompostos/química , Dióxido de Silício/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Clorofilídeos , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/uso terapêutico , Camundongos , Microscopia Confocal , Imagem Multimodal , Nanocompostos/toxicidade , Nanopartículas/química , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia , Porfirinas/química , Porfirinas/uso terapêutico , Pontos Quânticos/química , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA