Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
J Mater Chem B ; 12(27): 6690-6702, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38895854

RESUMO

Peripheral nerve injuries (PNIs) caused by mechanical contusion are frequently encountered in clinical practice, using nerve guidance conduits (NGCs) is now a promising therapy. An NGC creates a microenvironment for cell growth and differentiation, thus understanding physical and biochemical cues that can affect nerve-cell fate is a prerequisite for rationally designing NGCs. However, most of the previous works were focused on some static cues, the dynamic nature of the nerve microenvironment has not yet been well captured. Herein, we develop a micropatterned shape-memory polymer as a programmable substrate for providing a dynamic cue for nerve-cell growth. The shape-memory properties enable temporal programming of the substrate, and a dynamic microenvironment is created during standard cell culturing at 37 °C. Unlike most of the biomedical shape-memory polymers that recover rapidly at 37 °C, the proposed substrate shows a slow recovery process lasting 3-4 days and creates a long-term dynamic microenvironment. Results demonstrate that the vertically programmed substrates provide the most suitable dynamic microenvironment for PC12 cells as both the differentiation and maturity are promoted. Overall, this work provides a strategy for creating a long-term dynamic microenvironment for regulating nerve-cell fate and will inspire the rational design of NGCs for the treatment of PNIs.


Assuntos
Diferenciação Celular , Células PC12 , Ratos , Animais , Polímeros/química , Proliferação de Células/efeitos dos fármacos , Propriedades de Superfície , Microambiente Celular , Neurônios/citologia , Materiais Inteligentes/química
2.
Medicine (Baltimore) ; 103(18): e38038, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38701277

RESUMO

The present study aimed to establish an effective prognostic nomogram model based on the Naples prognostic score (NPS) for resectable thoracic esophageal squamous cell carcinoma (ESCC). A total of 277 patients with ESCC, who underwent standard curative esophagectomy and designated as study cohort, were retrospectively analyzed. The patients were divided into different groups, including NPS 0, NPS 1, NPS 2, and NPS 3 or 4 groups, for further analysis, and the results were validated in an external cohort of 122 ESCC patients, who underwent surgery at another cancer center. In our multivariate analysis of the study cohort showed that the tumor-node-metastasis (TNM) stage, systemic inflammation score, and NPS were the independent prognostic factors for the overall survival (OS) and progression-free survival (PFS) durations. In addition, the differential grade was also an independent prognostic factor for the OS in the patients with ESCC after surgery (all P < .05). The area under the curve of receiver operator characteristics for the PFS and OS prediction with systemic inflammation score and NPS were 0.735 (95% confidence interval [CI] 0.676-0.795, P < .001) and 0.835 (95% CI 0.786-0.884, P < .001), and 0.734 (95% CI 0.675-0.793, P < .001) and 0.851 (95% CI 0.805-0.896, P < .001), respectively. The above independent predictors for OS or PFS were all selected in the nomogram model. The concordance indices (C-indices) of the nomogram models for predicting OS and PFS were 0.718 (95% CI 0.681-0.755) and 0.669 (95% CI 0.633-0.705), respectively, which were higher than that of the 7th edition of American Joint Committee on Cancer TNM staging system [C-index 0.598 (95% CI 0.558-0.638) for OS and 0.586 (95% CI 0.546-0.626) for PFS]. The calibration curves for predicting the 5-year OS or PFS showed a good agreement between the prediction by nomogram and actual observation. In the external validation cohort, the nomogram discrimination for OS was better than that of the 7th edition of TNM staging systems [C-index: 0.697 (95% CI 0.639-0.755) vs 0.644 (95% CI 0.589-0.699)]. The calibration curves showed good consistency in predicting the 5-year survival between the actual observation and nomogram predictions. The decision curve also showed a higher potential of the clinical application of predicting the 5-years OS of the proposed nomogram model as compared to that of the 7th edition of TNM staging systems. The preoperative NPS-based nomogram model had a certain potential role for predicting the prognosis of ESCC patients.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Esofagectomia , Nomogramas , Humanos , Masculino , Feminino , Estudos Retrospectivos , Carcinoma de Células Escamosas do Esôfago/cirurgia , Carcinoma de Células Escamosas do Esôfago/mortalidade , Carcinoma de Células Escamosas do Esôfago/patologia , Pessoa de Meia-Idade , Neoplasias Esofágicas/cirurgia , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Prognóstico , Esofagectomia/métodos , Idoso , Estadiamento de Neoplasias , Adulto
3.
Adv Healthc Mater ; 13(18): e2304536, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38519046

RESUMO

Intense and persistent oxidative stress, excessive inflammation, and impaired angiogenesis severely hinder diabetic wound healing. Bioactive hydrogel dressings with immunoregulatory and proangiogenic properties have great promise in treating diabetic wounds. However, the therapeutic effects of dressings always depend on drugs with side effects, expensive cytokines, and cell therapies. Herein, a novel dynamic borate-bonds crosslinked hybrid multifunctional hydrogel dressings with photothermal properties are developed to regulate the microenvironment of diabetic wound sites and accelerate the whole process of its healing without additional medication. The hydrogel is composed of phenylboronic acid-modified chitosan and hyaluronic acid (HA) crosslinked by tannic acid (TA) through borate bonds and Prussian blue nanoparticles (PBNPs) with photothermal response characteristics are embedded in the polymer networks. The results indicate hydrogels show inherent broad-spectrum antioxidative activities through the integrated interaction of borate bonds, TA, and PBNPs. Meanwhile, combined with the regulation of macrophage phenotype by HA, the inflammatory microenvironment of diabetic wounds is transformed. Moreover, the angiogenesis is then enhanced by the mild photothermal effect of PBNPs, followed by promoted epithelialization and collagen deposition. In summary, this hybrid hydrogel system accelerates all stages of wound repair through antioxidative stress, immunomodulation, and proangiogenesis, showing great potential applications in diabetic wound management.


Assuntos
Quitosana , Ácido Hialurônico , Hidrogéis , Taninos , Cicatrização , Cicatrização/efeitos dos fármacos , Hidrogéis/química , Hidrogéis/farmacologia , Animais , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Camundongos , Quitosana/química , Taninos/química , Taninos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Diabetes Mellitus Experimental/terapia , Nanopartículas/química , Células RAW 264.7 , Antioxidantes/química , Antioxidantes/farmacologia , Ácidos Borônicos/química , Ácidos Borônicos/farmacologia , Masculino , Humanos , Temperatura Alta , Ferrocianetos/química , Ferrocianetos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Ratos
4.
Nat Commun ; 15(1): 814, 2024 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-38280861

RESUMO

Surgery is the mainstay of treatment modality for malignant melanoma. However, the deteriorative hypoxic microenvironment after surgery is recognized as a stemming cause for tumor recurrence/metastasis and delayed wound healing. Here we design and construct a sprayable therapeutic hydrogel (HIL@Z/P/H) encapsulating tumor-targeted nanodrug and photosynthetic cyanobacteria (PCC 7942) to prevent tumor recurrence/metastasis while promote wound healing. In a postsurgical B16F10 melanoma model in female mice, the nanodrug can disrupt cellular redox homeostasis via the photodynamic therapy-induced cascade reactions within tumor cells. Besides, the photosynthetically generated O2 by PCC 7942 can not only potentiate the oxidative stress-triggered cell death to prevent local recurrence of residual tumor cells, but also block the signaling pathway of hypoxia-inducible factor 1α to inhibit their distant metastasis. Furthermore, the long-lasting O2 supply and PCC 7942-secreted extracellular vesicles can jointly promote angiogenesis and accelerate the wound healing process. Taken together, the developed HIL@Z/P/H capable of preventing tumor recurrence/metastasis while promoting wound healing shows great application potential for postsurgical cancer therapy.


Assuntos
Hidrogéis , Oxigênio , Camundongos , Animais , Feminino , Hidrogéis/farmacologia , Recidiva Local de Neoplasia/prevenção & controle , Cicatrização/fisiologia , Hipóxia , Microambiente Tumoral
5.
ACS Biomater Sci Eng ; 9(8): 5039-5050, 2023 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-37535675

RESUMO

The convenience and availability are of great significance for the early screening of cancer. Herein, a magnetic nanoreporter with renal clearable capability and activatable catalytic activity was developed for colorimetric urinalysis of tumors. The magnetic nanoreporters were prepared by loading 3.2 nm Fe3O4 nanoparticles (NPs) and glucose oxidase (GOD) into macrophage cell-derived microvesicles (MVs) through electroporation, and these compositions serve as renal clearable catalytic reporters, synergistic catalysts, and targeted delivery carriers, respectively. The magnetic nanoreporters can convert the H2O2 in the mildly acidic tumor microenvironment into hydroxyl radicals through the synergistic catalysis of Fe3O4 NPs and GOD. Then the MVs can be disintegrated by the radicals, and ultrasmall Fe3O4 NPs will be released from the MVs at the tumor site, enabling rapid clearance of the Fe3O4 NPs into urine and a direct colorimetric urinalysis of the tumor within 4 h. The magnetic nanoreporters had good biocompatibility, and the released Fe3O4 NPs were rapidly excreted from the body, avoiding the potential toxicity. We envision that the magnetic nanoreporters can be used for convenient and rapid cancer screening.


Assuntos
Nanopartículas de Magnetita , Neoplasias/química , Humanos , Animais , Camundongos , Linhagem Celular Tumoral , Colorimetria , Urinálise/métodos , Nanopartículas de Magnetita/química , Nanopartículas Metálicas/química
6.
Acta Biomater ; 168: 497-514, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37507035

RESUMO

The persistent transformation of quiescent hepatic stellate cells (HSCs) into myofibroblasts (MFs) and the excessive proliferation of MF-HSCs in the liver contribute to the pathogenesis of liver fibrosis, cirrhosis, and liver cancer. Glycolysis inhibition of MF-HSCs can reverse their MF phenotype and suppress their abnormal expansion. Here, we have developed vitamin A-derivative (VA) decorated PEG-PCL polymeric micelles to encapsulate the labile and hydrophobic camptothecin (CPT) and direct its active attack on HSCs, selectively inhibiting of HIF-1α and cellular glycolysis, ultimately repressing hepatic fibrogenesis. The obtained micelles exhibited a good stability, biocompatibility, pH sensitivity, and exceptional HSC-targetability, allowing an efficient accumulation of their carried CPT in acutely and chronically injured livers. On their intracellular release of CPT specifically in MF-HSCs, these CPT micelles nicely inhibited the HIF-1α and a series of glycolytic players in MF-HSCs and prominently suppressed their proliferation and MF phenotypic characteristics. Accordingly, on in vitro administration to the mice challenged by CCl4 or subjected to bile duct ligation, these VA-decorated CPT micelles ameliorated the pathological symptoms of the livers, as evidenced by the significant reduction in serum levels of ALT and AST, infiltration of inflammatory cells, and collagen accumulation, the drastic down-regulation of multiple fibrotic genes, and the good recovery of attenuated hepatocyte CYP2E1 and lipogenesis regulator PPARγ. Overall, the CPT carried by VA-decorated PEG-PCL polymeric micelles can selectively inhibit the glycolysis and expansion of HSCs and thus suppress fibrogenesis, providing an original and effective approach for anti-fibrotic therapy. STATEMENT OF SIGNIFICANCE: Our work introduces an innovative antifibrotic drug system that is developed upon the active targeting of CPT and aims for the fate reversal of HSCs. Through HSC-targeted delivery achieved by PEG-PCL polymeric micelles decorated with vitamin A-derivatives, CPT significantly suppressed the expressions of HIF-1α and glycolytic enzymes in MF-HSCs, as well as their pathologic expansion in mouse livers. It effectively ameliorated chronic liver fibrosis in mice induced by CCl4 injection or BDL and restored the damaged liver structure and function. These compelling findings demonstrate the therapeutic potential of glycolytic HSC-targeting in combating fibrosis and related disorders and thus provide new promise for future clinical management of such prevalent and life-threatening conditions.


Assuntos
Células Estreladas do Fígado , Vitamina A , Camundongos , Animais , Vitamina A/efeitos adversos , Vitamina A/metabolismo , Células Estreladas do Fígado/metabolismo , Micelas , Células Cultivadas , Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Glicólise , Camptotecina/farmacologia
7.
Adv Healthc Mater ; 12(22): e2202871, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37276021

RESUMO

In situ vaccines have revolutionized immunotherapy as they can stimulate tumor-specific immune responses, with the cancer being the antigen source. However, the heterogeneity of tumor antigens and insufficient dendritic cells (DCs) activation result in low cancer immunogenicity and hence poor vaccine response. Herein, a new in situ vaccine composed of acid-responsive liposome-coated polydopamine (PDA) nanoparticles modified with mannose and loaded with resiquimod (R848) is designed to promote the efficacy of immunotherapy. The in situ vaccine can actively target the tumor site based on the decomposition of the liposome, while the PDA nanoparticles promote photothermal therapy and capture the immunogenic cell-death-induced tumor-associated antigens based on the adsorption effect of dopamine-mimetic mussels. The PDA nanoparticles, which are modified with a mannose ligand, target the DCs and release R848 for activated antigen presentation. As a result, the in situ vaccine not only effectively activates the maturation of the DCs but also significantly enhances their effect on cytotoxic T lymphocyte cells. Furthermore, the vaccine effectively inhibits the distant recurrence and metastasis of tumors via long-term immune memory effects. Therefore, the in situ vaccine provides a potential strategy for improving the efficacy of cancer immunotherapy.


Assuntos
Vacinas Anticâncer , Nanopartículas , Lipossomos , Terapia Fototérmica , Manose , Imunoterapia , Apresentação de Antígeno , Antígenos de Neoplasias , Vacinação , Vacinas Anticâncer/farmacologia , Células Dendríticas
8.
Biosensors (Basel) ; 13(5)2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-37232858

RESUMO

The effective detection and release of circulating tumor cells (CTCs) are of great significance for cancer diagnosis and monitoring. The microfluidic technique has proved to be a promising method for CTCs isolation and subsequent analysis. However, complex micro-geometries or nanostructures were often constructed and functionalized to improve the capture efficiency, which limited the scale-up for high-throughput production and larger-scale clinical applications. Thus, we designed a simple conductive nanofiber chip (CNF-Chip)-embedded microfluidic device with a herringbone microchannel to achieve the efficient and specific capture and electrical stimulation-triggered rapid release of CTCs. Here, the most used epithelial cell adhesion molecule (EpCAM) was selected as the representative biomarker, and the EpCAM-positive cancer cells were mainly studied. Under the effects of the nanointerface formed by the nanofibers with a rough surface and the herringbone-based high-throughput microfluidic mixing, the local topographic interaction between target cells and nanofibrous substrate in the microfluidic was synergistically enhanced, and the capture efficiency for CTCs was further improved (more than 85%). After capture, the sensitive and rapid release of CTCs (release efficiency above 97%) could be conveniently achieved through the cleavage of the gold-sulfur bond by applying a low voltage (-1.2 V). The device was successfully used for the effective isolation of CTCs in clinical blood samples from cancer patients, indicating the great potential of this CNF-Chip-embedded microfluidic device in clinical applications.


Assuntos
Técnicas Analíticas Microfluídicas , Nanofibras , Células Neoplásicas Circulantes , Humanos , Nanofibras/química , Molécula de Adesão da Célula Epitelial , Microfluídica , Dispositivos Lab-On-A-Chip , Linhagem Celular Tumoral
9.
ACS Appl Bio Mater ; 6(6): 2404-2414, 2023 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-37192493

RESUMO

Immunogenic cell death (ICD) is a promising cancer immunotherapy by inducing antigen-presenting cell maturation. Many inorganic nanomodulators have been developed for cancer therapy via ion overload, and their ICD-inducing properties have also been explored for immunotherapy. Here, we report a potassium chloride nanoparticle (PCNP)-loaded poly(lactic-co-glycolic acid) nanoparticle coated with cancer cell membrane (PC@P-CCM) for cancer therapy. Through cancer cell membrane (CCM)-achieved surface functionalization, the homotypic targeting behaviors of PC@P-CCM are dramatically enhanced. Once internalized by cancer cells, the PC@P-CCM could be degraded in acidic lysosomes, thus releasing K+ and Cl- ions. These ions can change the osmotic pressure of cancer cells, causing a hypertonic state in the cancer cells in a short time and leading to the rupture and death of cancer cells. Furthermore, these ions can stimulate cancer cells to secrete adenosine triphosphate (ATP) and high mobility group box 1 (HMGB-1); meanwhile, calreticulin (CRT) showed increased presentation on the surface of cancer cells, which can further induce dendritic cell maturation and promote the immunotherapy. This work provides a new perspective on KCl nanoparticle-based cancer immunotherapy.


Assuntos
Nanopartículas , Neoplasias , Humanos , Morte Celular Imunogênica , Cloreto de Potássio/farmacologia , Neoplasias/terapia , Imunoterapia , Nanopartículas/uso terapêutico
10.
ACS Appl Mater Interfaces ; 15(20): 24013-24022, 2023 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-37178127

RESUMO

Detection of circulating tumor cells (CTCs) is important for early cancer diagnosis, prediction of postoperative recurrence, and individualized treatment. However, it is still challenging to achieve efficient capture and gentle release of CTCs from the complex peripheral blood due to their rarity and fragility. Herein, inspired by the three-dimensional (3D) network structure and high glutathione (GSH) level of the tumor microenvironment (TME), a 3D stereo (3D-G@FTP) fibrous network is developed by combining the liquid-assisted electrospinning method, gas foaming technique, and metal-polyphenol coordination interactions to achieve efficient trapping and gentle release of CTCs. Compared with the traditional 2D@FTP fibrous scaffold, the 3D-G@FTP fibrous network could achieve higher capture efficiency (90.4% vs 78.5%) toward cancer cells in a shorter time (30 min vs 90 min). This platform showed superior capture performance toward heterogeneous cancer cells (HepG2, HCT116, HeLa, and A549) in an epithelial cell adhesion molecule (EpCAM)-independent manner. In addition, the captured cells with high cell viability (>90.0%) could be gently released under biologically friendly GSH stimulus. More importantly, the 3D-G@FTP fibrous network could sensitively detect 4-19 CTCs from six kinds of cancer patients' blood samples. We expect this TME-inspired 3D stereo fibrous network integrating efficient trapping, broad-spectrum recognition, and gentle release will promote the development of biomimetic devices for rare cell analysis.


Assuntos
Células Neoplásicas Circulantes , Humanos , Células Neoplásicas Circulantes/metabolismo , Glutationa , Células HeLa , Molécula de Adesão da Célula Epitelial/metabolismo , Linhagem Celular Tumoral , Separação Celular/métodos , Microambiente Tumoral
11.
Small Methods ; 7(5): e2201327, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37075716

RESUMO

In the treatment of solid tumors, the complex barriers composed of cancer-associated fibroblasts (CAFs) prevent drug delivery and T cells infiltration into tumor tissues. Although nanocarriers hold great prospects in drug delivery, fibrosis causes the biological barrier and immunosuppressive tumor microenvironment (ITM) that impairs the anti-tumor efficacy of nanocarriers. Here, a small dendritic macromolecule loaded with doxorubicin (PAMAM-ss-DOX) (DP) is synthesized and encapsulated into pH-responsive nanoliposome, together with adjuvant toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) and losartan (LOS). The pH-responsive liposome facilitates the simultaneous and effective delivery of DP, R848, and LOS, which can decompose and release these drugs under the acidic tumor microenvironment. The small sized DP (≈25 nm) with the ability to penetrate into tumor tissue and immunogenic cell death (ICD) can reverse the ITM and elicit immune response, which is equivalent to the effect of an in situ vaccine. Moreover, LOS reduces the activity of CAFs effectively, which can contribute to the infiltration of T cells. Therefore, this nano-platform provides a new therapeutic strategy for enhanced chemo-immunotherapy.


Assuntos
Lipossomos , Neoplasias , Humanos , Lipossomos/farmacologia , Microambiente Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Imunoterapia , Neoplasias/tratamento farmacológico
12.
Adv Mater ; 35(21): e2300216, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36912443

RESUMO

The acidic microenvironment of tumors significantly reduces the anti-tumor effect of immunotherapy. Herein, a hierarchically structured fiber device is developed as a local drug delivery system for remodeling the acidic tumor microenvironment (TME) to improve the therapeutic effect of immunotherapy. Proton pump inhibitors in the fiber matrix can be sustainedly released to inhibit the efflux of intracellular H+ from tumor cells, resulting in the remodeling of the acidic TME. The targeted micelles and M1 macrophage membrane-coated nanoparticles in internal cavities of fiber can induce immunogenic cell death (ICD) of tumor cells and phenotypic transformation of tumor-associated macrophages (TAMs), respectively. The relief of the acidity in the TME further promotes ICD and the polarization of TAMs, alleviating the immunosuppressive microenvironment and synergistically enhancing the antitumor immune response. In vivo results reveal this local drug delivery system restores the pH value of TME from 6.8 to 7.2 and exhibit an excellent immunotherapeutic effect.


Assuntos
Neoplasias , Microambiente Tumoral , Humanos , Imunoterapia/métodos , Neoplasias/patologia , Sistemas de Liberação de Medicamentos , Macrófagos/metabolismo
13.
Small ; 19(24): e2207252, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36922734

RESUMO

The abnormal pressure in tumor tissue is a significant limitation on the drug delivery efficiency of tumor therapy. This work reports a gradient-driven nanomotor as drug nanocarrier with the pressure-counterworking function. The dual-fuel nanomotors are formed by co-electrospinning of the photosensitive polymers with calcium peroxide (CaO2 ) and catalase (CAT), followed by ultraviolet (UV) irradiation and bovine serum albumin (BSA) incubation. The UV-responsive cleavage nanomotors can effectively release O2 molecules at the fractures as a driving force to increase the delivery speed and escape the phagocytosis of macrophage system in normal tissues. Furthermore, CAT catalyzes H2 O2 produced by CaO2 and the tumor interstitial fluids to provide stronger power for the nanomotors. Additionally, according to the analysis of directional motions of the nanomotors, the functional relationship between the rotational diffusion coefficient (DR ) and the physiological viscosity is constructed. The dual-fuel nanocarriers enable up to 13.25% of the injected dose (ID)/per gram tissue and significantly improve the penetration in deep tumor. It is of vital importance to design and obtain the adaptive pressure-gradient counterworking nanomotors, which can effectively improve the drug delivery efficiency in vitro and in vivo.


Assuntos
Sistemas de Liberação de Medicamentos , Líquido Extracelular , Preparações Farmacêuticas , Polímeros
14.
J Control Release ; 353: 535-548, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36481693

RESUMO

Immunotherapy has achieved remarkable research outcomes and shows the potential to cure cancer. However, its therapeutic response is limited in terms of the immunosuppressive tumor microenvironment induced by hypoxia, in which the adenosinergic A2A receptor (A2AR) pathway is mainly participated. Here, we developed a novel core/shell structured nanoplatform composed of macrophage membrane-coated mesoporous silica nanoparticles which loaded catalase, doxorubicin (Dox), and resiquimod (R848), to promote the efficacy of immunotherapy. The nanoplatform is able to actively target the tumor site via ligand binding, and the A2AR of T regulatory (Treg) cells can further be blocked due to in situ oxygen production by hydrogen peroxide catalysis. Meanwhile, Dox and R848 released from the nanoplatform can induce immunogenic cell death and enhance the activation of dendritic cells (DCs), respectively. Thus, the improved microenvironment by A2AR blockade and the stimulation of the DCs to enhance the CD8+ T cells mediated immune response were achieved. Consequently, the expression of Treg cells decreased to 9.79% in tumor tissue and the inhibition rate of tumor growth reached 73.58%. Therefore, this nanoplatform provides a potential strategy for clinical application in cancer immunotherapy.


Assuntos
Nanopartículas , Neoplasias , Humanos , Linfócitos T CD8-Positivos , Receptor A2A de Adenosina/metabolismo , Dióxido de Silício/metabolismo , Imunoterapia , Neoplasias/tratamento farmacológico , Doxorrubicina/farmacologia , Oxigênio/metabolismo , Adenosina , Macrófagos/metabolismo , Microambiente Tumoral
15.
Nano Lett ; 22(15): 6418-6427, 2022 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-35856800

RESUMO

Aberrant energy metabolism not only endows tumor cells with unlimited proliferative capacity but also contributes to the establishment of the glucose-deficient/lactate-rich immunosuppressive tumor microenvironment (ITM) impairing antitumor immunity. Herein, a novel metabolic nanoregulator (D/B/CQ@ZIF-8@CS) was developed by enveloping 2-deoxy-d-glucose (2-DG), BAY-876, and chloroquine (CQ) into zeolitic imidazolate framework-8 (ZIF-8) to simultaneously deprive the energy/nutrition supply of tumor cells and relieve the ITM for synergetic tumor starvation-immunotherapy. Aerobic glycolysis, glucose uptake, and autophagy flux could be concurrently blocked by D/B/CQ@ZIF-8@CS, cutting off the nutrition/energy supply and the source of lactate. Furthermore, inhibition of glucose uptake and aerobic glycolysis could effectively reverse the glucose-deficient/lactate-rich ITM, thus functionally inactivating regulatory T cells and augmenting anti-CTLA-4 immunotherapy. Such a two-pronged strategy would provide new insights for the design of metabolic intervention-based synergistic cancer therapy.


Assuntos
Glicólise , Neoplasias , Linhagem Celular Tumoral , Metabolismo Energético , Glucose/metabolismo , Humanos , Terapia de Imunossupressão , Lactatos , Neoplasias/tratamento farmacológico , Microambiente Tumoral
16.
Acta Pharm Sin B ; 12(6): 2683-2694, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35755281

RESUMO

Remodeling the tumor microenvironment through reprogramming tumor-associated macrophages (TAMs) and increasing the immunogenicity of tumors via immunogenic cell death (ICD) have been emerging as promising anticancer immunotherapy strategies. However, the heterogeneous distribution of TAMs in tumor tissues and the heterogeneity of the tumor cells make the immune activation challenging. To overcome these dilemmas, a hybrid bacterium with tumor targeting and penetration, TAM polarization, and photothermal conversion capabilities is developed for improving antitumor immunotherapy in vivo. The hybrid bacteria (B.b@QDs) are prepared by loading Ag2S quantum dots (QDs) on the Bifidobacterium bifidum (B.b) through electrostatic interactions. The hybrid bacteria with hypoxia targeting ability can effectively accumulate and penetrate the tumor tissues, enabling the B.b to fully contact with the TAMs and mediate their polarization toward M1 phenotype to reverse the immunosuppressive tumor microenvironment. It also enables to overcome the intratumoral heterogeneity and obtain abundant tumor-associated antigens by coupling tumor penetration of the B.b with photothermal effect of the QDs, resulting in an enhanced immune effect. This strategy that combines B.b-triggered TAM polarization and QD-induced ICD achieved a remarkable inhibition of tumor growth in orthotopic breast cancer.

17.
ACS Nano ; 16(4): 5778-5794, 2022 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-35324153

RESUMO

How to precisely reprogram tumor-associated macrophages (TAMs) and combine them with immunogenic cell death (ICD) is still a great challenge in enhancing the antitumor immunotherapeutic effect. Here, we developed a localized drug delivery system with a step-by-step cell internalization ability based on a hierarchical-structured fiber device. The chemotherapeutic agent-loaded nanomicelles are encapsulated in the internal chambers of the fiber, which could first be internalized by actively targeting tumor cells to induce ICD. Next, the rod-like microparticles can be gradually formed from long to short shape through hydrolysis of the fiber matrix in the tumor microenvironment and selectively phagocytosed by TAMs but not to tumor cells when the length becomes less than 3 µm. The toll-like receptors 7 (TLR7) agonist imiquimod could be released from these microparticles in the cytoplasm to reprogram M2-like TAMs. The in vivo results exhibit that this localized system can synergistically induce an antitumor immune response and achieve an excellent antitumor efficiency. Therefore, this system will provide a promising treatment platform for cancer immunotherapy.


Assuntos
Macrófagos , Neoplasias , Humanos , Macrófagos/metabolismo , Imunoterapia/métodos , Microambiente Tumoral , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Linhagem Celular Tumoral
18.
Chem Commun (Camb) ; 58(14): 2299-2302, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35075473

RESUMO

Effective delivery of therapeutics to tumors is generally hampered by the limited penetration of biological barriers imposed by the tumor microenvironment. Despite the broad applications of cell-penetrating peptides (CPPs) for intracellular delivery of therapeutics across membrane bilayers, the discovery of novel CPPs with enhanced tumor tissue permeability remains largely unexplored. Herein, we identified two short stapled CPPs with aromatic cross-links that confer superior dual-penetration in tumor cells and tissues over their linear counterparts. This work may benefit the future applications of constrained CPPs as powerful molecular transporters to access deeper tumor tissues.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Neoplasias/metabolismo , Animais , Transporte Biológico , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/química , Camundongos , Estrutura Molecular , Neoplasias/diagnóstico por imagem , Imagem Óptica
20.
Adv Mater ; 34(7): e2106307, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34859919

RESUMO

The checkpoint inhibitor therapy that blocks programmed death-1 (PD-1) and its major ligand PD-L1 has achieved encouraging clinical efficacy in certain cancers. However, the binding of checkpoint inhibitors with other immune cells that express PD-L1 often results in a low response rate to the blockade and severe adverse effects. Herein, an LyP1 polypeptide-modified outer-membrane vesicle (LOMV) loaded with a PD-1 plasmid is developed to achieve self-blockade of PD-L1 in tumor cells. The nanocarriers accumulate in the tumor tissue through OMV-targeting ability and are internalized into the tumor cells via the LyP1-mediated target, subsequently delivering PD-1 plasmid into the nucleus, leading to the expression of PD-1 by the tumor cells. In addition, a magnetic particle chemiluminescence kit is developed to quantitatively detect the binding rate of PD-1/PD-L1. The self-expressed PD-1 bonded with the PD-L1 is expressed by both autologous and neighboring tumor cells, achieving self-blockade. Simultaneously, the outer-membrane protein of LOMV recruits cytotoxic lymphocyte cells and natural killer cells to tumor tissues and stimulates them to secrete IFN-γ  , improving the antitumor activity of the PD-1/PD-L1 self-blocking therapy.


Assuntos
Antígeno B7-H1 , Neoplasias , Antígeno B7-H1/metabolismo , Bactérias/metabolismo , Humanos , Imunoterapia/métodos , Células Matadoras Naturais/metabolismo , Neoplasias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA