Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Small ; 18(41): e2201704, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36071027

RESUMO

Ferroptosis is a form of regulated cell death which can not only kill tumor cells but also enhance immunogenicity of tumor cells, and it is evidenced to be involved in a variety of tumor treatments, especially in cancer immunotherapy. Tumor cell-derived exosomes are reported to influence the progression and metastasis process of tumors. In the process of ferroptosis, exosomes are also demonstrated as mediators to export iron under high intracellular iron concentration and resist ferroptosis. Under this regard, the combined application of ferroptosis inducer and the inhibitor of iron-containing exosomes may enhance the ferroptosis. Herein, biocompatible hybrid nanoparticles composed of the iron oxide nanoparticles, polymers with oxaliplatin attached, and siProminin2 are constructed. The siProminin2 mediated exosomal inhibition can restore the intracellular iron concentration, which can also inhibit the secretion of tumor cell-derived exosomes. The combination of immunotherapy with oxaliplatin, ferroptosis-based cancer therapy and inhibition of tumor cell-derived exosomes can enhance the immune activation effects. The nanoparticles represent an excellent triple therapeutic strategy for enhancing ferroptosis-based cancer therapy and immunotherapy.


Assuntos
Ferroptose , Neoplasias , Linhagem Celular Tumoral , Humanos , Imunoterapia , Ferro/metabolismo , Neoplasias/metabolismo , Oxaliplatina/farmacologia , Polímeros
2.
Small ; 18(18): e2107712, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35285149

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is on of the most lethal malignant tumors with relatively poor prognosis, characterized with insufficient drug penetration, low immune response and obvious drug resistances. The therapeutic inefficiency is multifactorially related to its specific tumor microenvironment (TME), which is representatively featured as rich stroma and immunosuppression. In this work, a versatile drug delivery system is developed that can coencapsulate two prodrugs modified from gemcitabine (GEM) and a signal transducer and activator of transcription 3 (STAT3) inhibitor (HJC0152), and the gradient pH variation is further sensed in the TME of PDAC to achieve a higher penetration by reversing its surficial charges. The escorted prodrugs can release GEM intracellularly, and respond to the hypoxic condition to yield the parental STAT3 inhibitor HJC0152, respectively. By inhibiting STAT3, the tumor immunosuppression microenvironment can be re-educated through the reversion of M2-like tumor associated macrophages (M2-TAMs), recruitment of cytotoxic T lymphocytes and downregulation of regulatory T cells (Treg s). Furthermore, cytidine deaminase (CDA) and α-smooth muscle actin (α-SMA) expression can be downregulated, plus the lipid modification of GEM, the drug resistance of GEM can be greatly relieved. Based on the above design, a synergetic therapeutic efficacy in PDAC treatment can be achieved to provide more opportunity for clinical applications.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Pró-Fármacos , Carcinoma Ductal Pancreático/tratamento farmacológico , Linhagem Celular Tumoral , Resistência a Medicamentos , Humanos , Terapia de Imunossupressão , Micelas , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Pró-Fármacos/uso terapêutico , Microambiente Tumoral , Neoplasias Pancreáticas
3.
Nano Lett ; 20(7): 5275-5283, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32421336

RESUMO

In this work, we proposed a carry-on nitric-oxide (NO) luggage strategy for enhanced chemotherapeutic efficacy. A stimuli-responsive NO-releasing polypeptide was prepared as the building block to assemble into a micelle as a chemodrug-carrier. The micelle was anchored with cRGD peptide with the aim of targeting to tumors' neoangiogenesis. In situ generation of NO at the tumor site can promote the neovascularization to recruit more chemotherapeutics. Besides, the introduced exogenous NO can directly induce apoptosis, synergistically with the chemotherapeutics. A specific near-infrared-region (NIR) NO-probe was also developed to be coloaded to the micelle to report the in situ NO-release. In vitro and in vivo experiments were performed to demonstrate the targeting capability, increased accumulation, real-time NO-release reporting phenomenon, improved antitumor efficacy, and favorable biosafety. Embedding NO into drug cargo as carry-on luggage for enhanced chemotherapeutic efficacy, hopefully, can cast new lights and build a basic principle in the future clinical translation of nanomedicines.


Assuntos
Neoplasias , Óxido Nítrico , Linhagem Celular Tumoral , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Humanos , Micelas , Nanomedicina , Neoplasias/tratamento farmacológico
4.
Nano Lett ; 19(6): 3527-3534, 2019 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31058513

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is considered as one of the most aggressive malignancies due to its unique microenvironment of which the cardinal histopathological feature is the remarkable desmoplasia of the stroma, taking up about 80% of the tumor mass. The desmoplastic stroma negatively affects drug diffusion and the infiltration of T cells, leading to an immunosuppressive microenvironment. However, this unique microenvironment can limit the physical spread of pancreatic cancer via a neighbor suppression effect. Here, a tumor central stroma targeting and microenvironment responsive strategy was applied to generate a nanoparticle coloading paclitaxel and phosphorylated gemcitabine. The designed nanoparticle disrupted the central stroma while preserving the external stroma, thereby promoting the antitumor effectiveness of chemotherapeutics. Additionally, the resulting nanoparticle can modulate the tumor immunosuppressive microenvironment by augmenting the number of cytotoxic T cells and restraining the percentage of T regulatory cells. The relatively intact external stroma can effectively maintain the neighbor suppression effect and prevent tumor metastasis. Combining stroma targeting with the delivery of stimuli-responsive polymeric nanoparticles embodies an effective tumor-tailored drug delivery system.


Assuntos
Antineoplásicos/administração & dosagem , Carcinoma Ductal Pancreático/tratamento farmacológico , Desoxicitidina/análogos & derivados , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Desoxicitidina/administração & dosagem , Desoxicitidina/uso terapêutico , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Humanos , Camundongos , Nanopartículas/química , Paclitaxel/uso terapêutico , Neoplasias Pancreáticas/patologia , Microambiente Tumoral/efeitos dos fármacos , Gencitabina
5.
Phys Chem Chem Phys ; 21(41): 22939-22946, 2019 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-31598612

RESUMO

Developing low-cost and metal-free electrocatalysts for the oxygen reduction reaction (ORR) and oxygen evolution reaction (OER) is desirable for renewable energy technologies. Recent experiments show that tellurium (Te) atoms can be effectively doped into black phosphorus (BP) nanosheets, and they greatly improve its OER catalytic performance. However, the specific active sites and microscopic configurations in the atomic-scale are still ambiguous. Here, we show that the doped Te atoms prefer to bond with each other to form clusters in phosphorene and they can be further stabilized by various intrinsic defects (Stone-Wales, single vacancy defects and zigzag nanoribbon). Benefiting from the reduced binding strength of O*, Te dopants and intrinsic defects synergistically boost the catalytic activity of phosphorene. The best OER catalytic activity could be realized in the cluster SW2-Te1p (Stone-Wales defect decorated by one Te atom). For ORR, the cluster Pri-Te3p (pristine phosphorene decorated by three Te atoms) exhibits optimal catalytic activity. Calculated ORR/OER potential gaps indicate that the SW2-Te3p cluster most likely acts as the efficient bifunctional catalytic site for both ORR and OER.

6.
J Chem Phys ; 151(14): 144710, 2019 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-31615251

RESUMO

We systematically explored the catalytic performance of a novel two dimensional material PtTe sheet for nitrogen reduction reaction (NRR) and hydrogen evolution reaction (HER) by using first-principles calculation. Although pristine PtTe shows poor NRR and HER activity, doping transition metal (TM) atoms into the lattice could effectively enhance the catalytic performance. Calculations show that four TM doped structures, including W-Pt18Te17, Ru-Pt18Te17, Mo-Pt18Te17, and Cr-Pt18Te17, are promising NRR catalysts on the prerequisite of whose HER activities are effectively suppressed. Moreover, the HER performance of the PtTe sheet could also be significantly improved with introduced TM atoms. In particular, Cu, Cr, Co, Ni, Mo, Rh, Ru, and Tc atoms supported by the PtTe sheet with Te-vacancy are promising HER electrocatalysts. The improved HER performance is attributed to the reduced adsorption free energy of the H atom. Both the doped TM atoms and the adjacent Pt atoms are effective active sites. Among all considered structures, Mo-, Cr-, and Ru-Pt18Te17 sheets boost catalytic activity for both NRR and HER. This study provides new design strategies to enhance the catalytic performance of the PtTe sheet for the NRR/HER.

7.
Nano Lett ; 18(3): 1908-1915, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-29473753

RESUMO

Various delivery vectors have been integrated within biologically derived membrane systems to extend their residential time and reduce their reticuloendothelial system (RES) clearance during systemic circulation. However, rational design is still needed to further improve the in situ penetration efficiency of chemo-drug-loaded membrane delivery-system formulations and their release profiles at the tumor site. Here, a macrophage-membrane-coated nanoparticle is developed for tumor-targeted chemotherapy delivery with a controlled release profile in response to tumor microenvironment stimuli. Upon fulfilling its mission of tumor homing and RES evasion, the macrophage-membrane coating can be shed via morphological changes driven by extracellular microenvironment stimuli. The nanoparticles discharged from the outer membrane coating show penetration efficiency enhanced by their size advantage and surface modifications. After internalization by the tumor cells, the loaded drug is quickly released from the nanoparticles in response to the endosome pH. The designed macrophage-membrane-coated nanoparticle (cskc-PPiP/PTX@Ma) exhibits an enhanced therapeutic effect inherited from both membrane-derived tumor homing and step-by-step controlled drug release. Thus, the combination of a biomimetic cell membrane and a cascade-responsive polymeric nanoparticle embodies an effective drug delivery system tailored to the tumor microenvironment.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Membrana Celular/metabolismo , Preparações de Ação Retardada/metabolismo , Macrófagos/metabolismo , Nanopartículas/metabolismo , Paclitaxel/administração & dosagem , Animais , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/uso terapêutico , Materiais Biomiméticos/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Camundongos , Nanopartículas/ultraestrutura , Paclitaxel/farmacocinética , Paclitaxel/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos
8.
Mol Pharm ; 14(10): 3409-3421, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28832164

RESUMO

Most small molecular chemotherapeutics have poor water solubility and unexpected pharmacokinetics and toxicity to normal tissues. A series of nano drug delivery systems have been developed to solve the problems, among which a micelle based on linear-dendritic polymer-drug conjugates (LDPDCs) is a promising strategy to deliver hydrophobic chemotherapeutics due to its small size, fine stability in blood circulation, and high drug loading capacity. In this work we synthesized a novel amphiphilic linear-dendritic PEG-PTX8 conjugate which can also encapsulate extra free PTX and self-assemble into uniform ultrasmall micelles with a hydrated diameter of 25.50 ± 0.27 nm. To realize efficient drug delivery to tumor sites, a cyclic tumor homing and penetrating peptide iNGR was linked to the PEG-PTX8 conjugate. The biological evaluation was performed on a human triple negative breast cancer model. PTX accumulation in tumor at 24 h of the TNBC-bearing mice treated with iNGR-PEG-PTX8/PTX micelles was significantly enhanced (P < 0.001, two-way ANOVA) compared to that of Taxol and untargeted MeO-PEG-PTX8/PTX micelles. Furthermore, iNGR-PEG-PTX8/PTX micelles showed an obvious strong antitumor effect, and the median survival time of TNBC bearing mice treated with iNGR-modified micelles was significantly extended compared to Taxol. Therefore, this smart micelle system may be a favorable platform for effective TNBC therapy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Dendrímeros/química , Portadores de Fármacos/química , Paclitaxel/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Humanos , Camundongos , Camundongos Nus , Micelas , Oligopeptídeos/química , Paclitaxel/uso terapêutico , Polietilenoglicóis/química , Solubilidade , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Environ Sci Pollut Res Int ; 31(11): 16473-16484, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38321280

RESUMO

g-C3N4/ZnxCd(1-x)S composites were synthesized by a simple hydrothermal method. The composites were characterized by X-ray diffraction, UV-vis diffuse reflectance spectroscopy, infrared spectroscopy, and electron micro-projective microscopy. According to the performance of ZnxCd(1-x)S for the photocatalytic degradation of norfloxacin under visible light in water, the best stoichiometric number of x was 0.5. The best photolytic norfloxacin degradation rate of g-C3N4/ZnxCd(1-x)S composites was 89.8%, which was obtained when the dosage ratio of g-C3N4 to ZnxCd(1-x)S was 1:1. The experiment was conducted to investigate the effect of pH on the catalyst to obtain the optimal NORF degradation environment pH in the range of 7 ± 0.3; by simulating the anions that may be contained in the actual environmental water, the results showed that the catalyst has a certain effect on the degradation of NORF when the water contains NO3-, Cl- and HCO3-. In addition, this study also obtained that the main active substances produced by the catalyst during degradation were electron-hole pairs by adding different trapping agents in the NORF removal experiments; and the catalyst was able to achieve a degradation rate of 86.1% after four cycles of the experiments, which proved that it had good stability.


Assuntos
Antibacterianos , Norfloxacino , Antibacterianos/química , Cádmio , Fotólise , Luz , Zinco
10.
Adv Mater ; 36(26): e2312219, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38608672

RESUMO

Targeting the competitive-cooperative relationships among tumor cells and various immune cells can efficiently reverse the immune-dysfunction microenvironment to boost the immunotherapies for the triple-negative breast cancer treatment. Hence, a bacterial outer membrane vesicle-based nanocomplex is designed for specifically targeting malignant cells and immune cells to reconcile the relationships based on metabolic-immune crosstalk. By uniquely utilizing the property of charge-reversal polymers to realize function separation, the nanocomplexes could synergistically regulate tumor cells and immune cells. This approach could reshape the immunosuppressive competition-cooperation pattern into one that is immune-responsive, showcasing significant potential for inducing tumor remission in TNBC models.


Assuntos
Neoplasias de Mama Triplo Negativas , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Humanos , Animais , Linhagem Celular Tumoral , Camundongos , Microambiente Tumoral/efeitos dos fármacos , Feminino , Imunoterapia , Nanopartículas/química
11.
J Control Release ; 358: 382-397, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37100207

RESUMO

Precisely reporting the inflammatory microenvironment, which can provide important basis for disease diagnosis and progression determination, is always challenging. In this work, we develop a targeting peptide-conjugated chemiluminescent reporter (OFF), which can be recognized by in-situ circulated neutrophils once injected, and then transported to the inflamed tissues characterized with overexpressed superoxide anion (O2-), assisted by the chemotaxis natural of neutrophils. Subsequently, the chemiluminescent probe can particularly respond to O2- to release the caged photons (ON) for visualizing inflammatory diseases such as subcutaneous tumor, colorectal cancer peritoneal metastasis (CCPM), ear swelling and kidney failure. The chemiluminescent probe provides a reliable tool to early detect inflammation and precisely excise micrometastatic lesions under optical guidance. This study provides a potential approach for improving the performance of luminophore in advanced bioimaging applications.


Assuntos
Medições Luminescentes , Neutrófilos , Medições Luminescentes/métodos , Superóxidos , Indicadores e Reagentes , Diagnóstico por Imagem
12.
ACS Nano ; 17(8): 7847-7864, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37039779

RESUMO

Medicinal treatment against epilepsy is faced with intractable problems, especially epileptogenesis that cannot be blocked by clinical antiepileptic drugs (AEDs) during the latency of epilepsy. Abnormal circuits of neurons interact with the inflammatory microenvironment of glial cells in epileptic foci, resulting in recurrent seizures and refractory epilepsy. Herein, we have selected phenytoin (PHT) as a model drug to derive a ROS-responsive and consuming prodrug, which is combined with an electro-responsive group (sulfonate sodium, SS) and an epileptic focus-recognizing group (α-methyl-l-tryptophan, AMT) to form hydrogel nanoparticles (i.e., a nanogel). The nanogel will target epileptic foci, release PHT in response to a high concentration of reactive oxygen species (ROS) in the microenvironment, and inhibit overexcited circuits. Meanwhile, with the clearance of ROS, the nanogel can also reduce oxidative stress and alleviate microenvironment inflammation. Thus, a synergistic regulation of epileptic lesions will be achieved. Our nanogel is expected to provide a more comprehensive strategy for antiepileptic treatment.


Assuntos
Epilepsia , Humanos , Espécies Reativas de Oxigênio/uso terapêutico , Nanogéis , Epilepsia/tratamento farmacológico , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Fenitoína
13.
ACS Nano ; 17(18): 18164-18177, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37703316

RESUMO

Reperfusion injury presents a significant obstacle to neuronal survival following successful recanalization in ischemic stroke, which is characterized by intricate pathophysiological processes comprising numerous interconnected pathways. Oxidative stress-induced neuronal ferroptosis and the overactivation of glial cells play important roles in this phenomenon. In this study, we developed a targeted cross-linked micelle loaded with idebenone to rescue the ischemic penumbra by inhibiting neuronal ferroptosis and glial overactivation. In rat models, the CREKA peptide-modified micelles accumulate in the damaged brain via binding to microthrombi in the ipsilateral microvessels. Upon reactive oxygen species (ROS) stimulation, diselenide bonds within the micelles are transformed to hydrophilic seleninic acids, enabling synchronized ROS consumption and responsive drug release. The released idebenone scavenges ROS, prevents oxidative stress-induced neuronal ferroptosis, attenuates glial overactivation, and suppresses pro-inflammatory factors secretion, thereby modulating the inflammatory microenvironment. Finally, this micelle significantly reinforces neuronal survival, reduces infarct volume, and improves behavioral function compared to the control groups. This pleiotropic therapeutic micelle provides a proof-of-concept of remodeling the lesion microenvironment by inhibiting neuronal ferroptosis and glial overactivation to treat cerebral ischemia-reperfusion injury.


Assuntos
Ferroptose , Traumatismo por Reperfusão , Animais , Ratos , Micelas , Espécies Reativas de Oxigênio , Neuroglia , Traumatismo por Reperfusão/tratamento farmacológico
14.
Bioact Mater ; 27: 474-487, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37159612

RESUMO

A long-standing paucity of effective therapies results in the poor outcomes of triple-negative breast cancer brain metastases. Immunotherapy has made progress in the treatment of tumors, but limited by the non-immunogenicity of tumors and strong immunosuppressive environment, patients with TNBC brain metastases have not yet benefited from immunotherapy. Dual immunoregulatory strategies with enhanced immune activation and reversal of the immunosuppressive microenvironment provide new therapeutic options for patients. Here, we propose a cocktail-like therapeutic strategy of microenvironment regulation-chemotherapy-immune synergistic sensitization and construct reduction-sensitive immune microenvironment regulation nanomaterials (SIL@T). SIL@T modified with targeting peptide penetrates the BBB and is subsequently internalized into metastatic breast cancer cells, releasing silybin and oxaliplatin responsively in the cells. SIL@T preferentially accumulates at the metastatic site and can significantly prolong the survival period of model animals. Mechanistic studies have shown that SIL@T can effectively induce immunogenic cell death of metastatic cells, activate immune responses and increase infiltration of CD8+ T cells. Meanwhile, the activation of STAT3 in the metastatic foci is attenuated and the immunosuppressive microenvironment is reversed. This study demonstrates that SIL@T with dual immunomodulatory functions provides a promising immune synergistic therapy strategy for breast cancer brain metastases.

15.
Acta Biomater ; 167: 387-400, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37276955

RESUMO

The tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC) is the main block for the penetration of chemotherapy. In the tumor microenvironment, a dense matrix composed of fibrin is formed on the exterior, while the interior is featured by high reduction, hypoxia and low pH. How to match the special microenvironment to on-demand drug release is the key to improve chemotherapeutic efficacy. Herein, a microenvironment-responsive micellar system is developed to deepen tumoral penetration. Briefly, the conjugation of a fibrin-targeting peptide to PEG-poly amino acid has been utilized to achieve accumulation of micelles in the tumor stroma. By modification of micelles with hypoxia-reducible nitroimidazole which becomes protonated under acidic conditions, their surface charge is more positive, facilitating deeper penetration into tumors. Paclitaxel was loaded onto the micelles via a disulfide bond to enable glutathione (GSH)-responsive release. Therefore, the immunosuppressive microenvironment is relived through the alleviation of hypoxia and depletion of GSH. Hopefully, this work could establish paradigms by designing sophisticated drug-delivery systems to tactfully employ and retroact the tamed tumoral microenvironment to improve the therapeutic efficacy based on understanding the multiple hallmarks and learning the mutual regulation. STATEMENT OF SIGNIFICANCE: Tumor microenvironment(TME) is an unique pathological feature of pancreatic cancer and an inherent barrier to chemotherapy. Numerous studies regard TME as the targets for drug delivery. In this work, we propose a hypoxia-responsive nanomicellar drug delivery system that aiming hypoxia TME of pancreatic cancer. The nanodrug delivery system could respond to the hypoxic microenvironment and enhance the penetration of the inner tumor at the same time preserving the outer tumor stroma, thus achieving targeted treatment of PDAC by preserving the integrity of the outer stroma. Simultaneously, the responsive group can reverse the degree of hypoxia in TME by disrupting the redox balance in the tumor region, thus achieving precise treatment of PDAC by matching the pathological characteristics of TME. We believe our article would provide new design ideas for the future treatments for pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Micelas , Microambiente Tumoral , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Hipóxia , Glutationa , Terapia de Imunossupressão , Linhagem Celular Tumoral , Neoplasias Pancreáticas
16.
Acta Pharm Sin B ; 12(5): 2506-2521, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35646523

RESUMO

Retinal pigment epithelial (RPE) is primarily impaired in age-related macular degeneration (AMD), leading to progressive loss of photoreceptors and sometimes choroidal neovascularization (CNV). mTOR has been proposed as a promising therapeutic target, while the usage of its specific inhibitor, rapamycin, was greatly limited. To mediate the mTOR pathway in the retina by a noninvasive approach, we developed novel biomimetic nanocomplexes where rapamycin-loaded nanoparticles were coated with cell membrane derived from macrophages (termed as MRaNPs). Taking advantage of the macrophage-inherited property, intravenous injection of MRaNPs exhibited significantly enhanced accumulation in the CNV lesions, thereby increasing the local concentration of rapamycin. Consequently, MRaNPs effectively downregulated the mTOR pathway and attenuate angiogenesis in the eye. Particularly, MRaNPs also efficiently activated autophagy in the RPE, which was acknowledged to rescue RPE in response to deleterious stimuli. Overall, we design and prepare macrophage-disguised rapamycin nanocarriers and demonstrate the therapeutic advantages of employing biomimetic cell membrane materials for treatment of AMD.

17.
Adv Healthc Mater ; 11(3): e2101578, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34800085

RESUMO

The rapid postoperative recurrence and short survival time of glioblastoma (GBM) patients necessitate immediate and effective postoperative treatment. Herein, an immediate and mild postoperative local treatment strategy is developed that regulates the postoperative microenvironment and delays GBM recurrence. Briefly, an injectable hydrogel system (imGEL) loaded with Zn(II)2 -AMD3100 (AMD-Zn) and CpG oligonucleotide nanoparticles (CpG NPs) is injected into the operation cavity, with long-term function to block the recruitment of microglia/ macrophages and activate cytotoxic T cells. The finding indicated that the imGEL can regulate the immune microenvironment, inhibit GBM recurrence, and gain valuable time for subsequent adjuvant clinical chemotherapy.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/cirurgia , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Glioblastoma/cirurgia , Humanos , Hidrogéis/uso terapêutico , Nanopartículas/uso terapêutico , Microambiente Tumoral
18.
Biomaterials ; 280: 121306, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34952381

RESUMO

Despite tremendous progress achieved in immunotherapy, many critical challenges in treating pancreatic ductal adenocarcinoma (PDAC) persist. Considering the poor vascularization of PDAC, after intramuscular administration exosomes can targeted deliver "cargos" to pancreatic tumors and bypass obstructions of the intrinsic overexpressed stroma through lymphatics. Herein, we propose a strategy to derive exosomes from immunogenically dying tumor cells and exploit their properties for several purposes, including antigen presentation, adjuvant supply, and "cargo" delivery of vaccines against pancreatic cancer via intramuscular injection. To enhance the immunostimulatory effects, the MART-1 peptide is modified to the exosomes to expand T-cell-related responses. Furthermore, CCL22 siRNA is electroporated into the exosomes (referred to as spMEXO) to hinder the CCR4/CCL22 axis between DCs and Tregs, thereby suppressing Treg expansion. Both in vitro and in vivo studies demonstrate that spMEXO can serve as an effective prophylactic vaccine to delay tumor growth, whereas combining spMEXO with PDAC first-line chemotherapeutics (co-administration of gemcitabine with albumin-paclitaxel) demonstrated significantly enhanced therapeutic effects in established PANC-02 tumors. Therefore, the present work provides an effective strategy to employ cancer vaccines through intramuscular injection in PDAC and highlights the potential of exosomes derived from immunogenically dying tumor cells as a versatile tool to develop nanovaccines for immunotherapy.


Assuntos
Carcinoma Ductal Pancreático , Exossomos , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/terapia , Linhagem Celular Tumoral , Exossomos/genética , Humanos , Imunoterapia , Neoplasias Pancreáticas/patologia , Vacinação
19.
Nanomicro Lett ; 13(1): 92, 2021 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-34138315

RESUMO

Although current anticancer immunotherapies using immune checkpoint inhibitors (ICIs) have been reported with a high clinical success rate, numerous patients still bear 'cold' tumors with insufficient T cell infiltration and low immunogenicity, responding poorly to ICI therapy. Considering the advancements in precision medicine, in-depth mechanism studies on the tumor immune microenvironment (TIME) among cold tumors are required to improve the treatment for these patients. Nanomedicine has emerged as a promising drug delivery system in anticancer immunotherapy, activates immune function, modulates the TIME, and has been applied in combination with other anticancer therapeutic strategies. This review initially summarizes the mechanisms underlying immunosuppressive TIME in cold tumors and addresses the recent advancements in nanotechnology for cold TIME reversal-based therapies, as well as a brief talk about the feasibility of clinical translation.

20.
Adv Sci (Weinh) ; 8(20): e2102256, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34398516

RESUMO

A versatile tumor-targeting stimuli-responsive theranostic platform for peritoneal metastases of colorectal cancer is proposed in this work for tumor tracking and photothermal-enhanced chemotherapy. A quenched photosensitizer ("off" state) is developed and escorted into a tumor-targeting oxaliplatin-embedded micelle. Once reaching the tumor cell, the micelle is clasped to release free oxaliplatin, as well as the "off" photosensitizer, which is further activated ("turned-on") in the tumor reducing microenvironment to provide optical imaging and photothermal effect. The combined results from hyperthermia-enhanced chemotherapy, deep penetration, perfused O2 , and the leveraged GSH-ROS imbalance in tumor cells are achieved for improved antitumor efficacy and reduced systematic toxicity.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Tratamento Farmacológico , Oxaliplatina/farmacologia , Neoplasias Peritoneais/tratamento farmacológico , Terapia Fototérmica , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Humanos , Camundongos , Metástase Neoplásica , Oxaliplatina/química , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/secundário , Medicina de Precisão , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA