Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Small ; : e2400542, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38593309

RESUMO

Osteoarthritis (OA) management remains challenging because of its intricate pathogenesis. Intra-articular injections of drugs, such as glucocorticoids and hyaluronic acid (HA), have certain limitations, including the risk of joint infection, pain, and swelling. Hydrogel-based therapeutic strategies have attracted considerable attention because of their enormous therapeutic potential. Herein, a supramolecular nanofiber hydrogel is developed using dexamethasone sodium phosphate (DexP) as a vector to deliver lentivirus-encoding hyaluronan synthase 2 (HAS2) (HAS2@DexP-Gel). During hydrogel degradation, HAS2 lentivirus and DexP molecules are slowly released. Intra-articular injection of HAS2@DexP-Gel promotes endogenous HA production and suppresses synovial inflammation. Additionally, HAS2@DexP-Gel reduces subchondral bone resorption in the anterior cruciate ligament transection-induced OA mice, attenuates cartilage degeneration, and delays OA progression. HAS2@DexP-Gel exhibited good biocompatibility both in vitro and in vivo. The therapeutic mechanisms of the HAS2@DexP-Gel are investigated using single-cell RNA sequencing. HAS2@DexP-Gel optimizes the microenvironment of the synovial tissue by modulating the proportion of synovial cell subpopulations and regulating the interactions between synovial fibroblasts and macrophages. The innovative nanofiber hydrogel, HAS2@DexP-Gel, effectively enhances endogenous HA production while reducing synovial inflammation. This comprehensive approach holds promise for improving joint function, alleviating pain, and slowing OA progression, thereby providing significant benefits to patients.

2.
Nat Commun ; 15(1): 820, 2024 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-38280876

RESUMO

Transdermal drug delivery has been regarded as an alternative to oral delivery and subcutaneous injection. However, needleless transdermal delivery of biomacromolecules remains a challenge. Herein, a transdermal delivery platform based on biocompatible fluorocarbon modified chitosan (FCS) is developed to achieve highly efficient non-invasive delivery of biomacromolecules including antibodies and antigens. The formed nanocomplexes exhibits effective transdermal penetration ability via both intercellular and transappendageal routes. Non-invasive transdermal delivery of immune checkpoint blockade antibodies induces stronger immune responses for melanoma in female mice and reduces systemic toxicity compared to intravenous injection. Moreover, transdermal delivery of a SARS-CoV-2 vaccine in female mice results in comparable humoral immunity as well as improved cellular immunity and immune memory compared to that achieved with subcutaneous vaccine injection. Additionally, FCS-based protein delivery systems demonstrate transdermal ability for rabbit and porcine skins. Thus, FCS-based transdermal delivery systems may provide a compelling opportunity to overcome the skin barrier for efficient transdermal delivery of bio-therapeutics.


Assuntos
Quitosana , Melanoma , Vacinas Virais , Suínos , Feminino , Humanos , Animais , Camundongos , Coelhos , Melanoma/tratamento farmacológico , Vacinas contra COVID-19 , Imunoterapia , Sistemas de Liberação de Medicamentos
3.
Sci Adv ; 9(44): eadh1582, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37910617

RESUMO

Choroidal melanoma, a common intraocular malignant tumor, relies on local radiotherapy and enucleation for treatment. However, cancer recurrence and visual impairment remain important challenges. Here, a therapeutic artificial vitreous body (AVB) hydrogel based on tetra-armed poly(ethylene glycol) was developed to control the recurrence of choroidal melanoma and preserve vision after vitrectomy. AVB loaded with melphalan (Mel) and anti-programmed cell death ligand-1 (αPDL1), was injected after surgical resection in the choroidal melanoma mouse model. Afterwards, the sequentially released Mel and αPDL1 from AVB could achieve a synergistic antitumor effect to inhibit tumor recurrence. AVB with similar physical properties to native vitreous body could maintain the normal structure and visual function of eye after vitrectomy, which has been evidenced by standard examinations of ophthalmology in the mouse model. Thus, the immunotherapeutic AVB may be a promising candidate as an infill biomaterial to assist surgical treatment of intraocular malignant tumors.


Assuntos
Neoplasias da Coroide , Melanoma , Animais , Camundongos , Corpo Vítreo , Vitrectomia , Hidrogéis , Recidiva Local de Neoplasia/patologia , Melanoma/patologia , Neoplasias da Coroide/cirurgia , Neoplasias da Coroide/patologia , Melfalan , Imunoterapia
4.
Adv Mater ; 35(29): e2302220, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37178454

RESUMO

Radiotherapy (RT) is an extensively used strategy for cancer treatment, but its therapeutic effect is usually limited by the abnormal tumor microenvironment (TME) and it lacks the ability to control tumor metastases. In this work, a nanoscale coordination polymer, Hf-nIm@PEG (HNP), is prepared by the coordination of hafnium ions (Hf4+ ) with 2-nitroimidazole (2-nIm), and then modified with lipid bilayers containing poly(ethylene glycol) (PEG). Under low-dose X-ray irradiation, on the one hand, Hf4+ with high computed tomography signal enhancement ability can deposit radiation energy to induce DNA damage, and on the other hand, NO can be persistently released from 2-nIm, which can not only directly react with the radical DNA to prevent the repair of damaged DNA but also relieves the hypoxic immunosuppressive TME to sensitize radiotherapy. Additionally, NO can also react with superoxide ions to generate reactive nitrogen species (RNS) to induce cell apoptosis. More interestingly, it is discovered that Hf4+ can effectively activate the cyclic-di-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway to promote the immune responses induced by radiotherapy. Thus, this work presents a simple but multifunctional nanoscale coordination polymer to deposit radiation energy, trigger the release of NO, modulate the TME, activate the cGAS-STING pathway, and finally realize synergistic radio-immunotherapy.


Assuntos
Neoplasias , Óxido Nítrico , Humanos , Raios X , Háfnio , Nucleotidiltransferases , Imunoterapia , Microambiente Tumoral , Neoplasias/radioterapia
5.
Sensors (Basel) ; 23(7)2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-37050426

RESUMO

The quantitative defect detection of wire rope is crucial to guarantee safety in various application scenes, and sophisticated inspection conditions usually lead to the accurate testing of difficulties and challenges. Thus, a magnetic flux leakage (MFL) signal analysis and convolutional neural networks (CNNs)-based wire rope defect recognition method was proposed to solve this challenge. Typical wire rope defect inspection data obtained from one-dimensional (1D) MFL testing were first analyzed both in time and frequency domains. After the signal denoising through a new combination of Haar wavelet transform and differentiated operation and signal preprocessing by normalization, ten main features were used in the datasets, and then the principles of the proposed MFL and 1D-CNNs-based wire rope defect classifications were presented. Finally, the performance of the novel method was evaluated and compared with six machine learning methods and related algorithms, which demonstrated that the proposed method featured the highest testing accuracy (>98%) and was valid and feasible for the quantitative and accurate detection of broken wire defects. Additionally, the considerable application potential as well as the limitations of the proposed methods, and future work, were discussed.

6.
Biomaterials ; 295: 122052, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36827893

RESUMO

Physiological barriers and immunosuppressive microenvironments of solid tumors present considerable hurdles to Chimeric antigen receptor T (CAR-T) cell therapy. Herein, we discovered that metformin, a prescribed drug for type 2 diabetes, could up-regulate the oxidative phosphorylation of CAR-T cells, increase their energy metabolism, and further promote their proliferation. Inspired by this finding, we designed a hydrogel scaffold to co-deliver metformin and CAR-T cells by adding CAR-T cells into a lyophilized alginate hydrogel containing metformin. The obtained hydrogel scaffold after being implanted into the tumor resection cavity could act as a cell reservoir to sustainably release both CAR-T cells and metformin. While the released metformin could suppress oxidative and glycolytic metabolism of cancer cells and lead to decreased tumor hypoxia, CAR-T cells would respond to metformin by markedly up-regulating oxidative metabolism and adopting a long-lived, highly activated phenotype, contributing to elevated antitumor responses. As demonstrated in several post-surgical tumor models, the proliferation and tumor-infiltration of CAR-T cells were significantly enhanced and the treatment efficacy of CAR-T cells was augmented, against both local tumors and distant abscopal tumors, while showing reduced systemic immune-related adverse effects. Our work presents a new strategy to achieve effective yet safe CAR-T therapy against solid tumors using a cell-delivery scaffold based on clinically validated drugs and biomaterials.


Assuntos
Diabetes Mellitus Tipo 2 , Metformina , Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Metformina/farmacologia , Metformina/uso terapêutico , Metformina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hidrogéis/metabolismo , Linfócitos T , Neoplasias/metabolismo , Imunoterapia Adotiva , Microambiente Tumoral
7.
ACS Nano ; 17(5): 4373-4386, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36802527

RESUMO

Therapeutic proteins are playing increasingly important roles in treating numerous types of diseases. However, oral administration of proteins, especially large ones (e.g., antibodies), remains a great challenge due to their difficulties in penetrating intestinal barriers. Herein, fluorocarbon-modified chitosan (FCS) is developed for efficient oral delivery of different therapeutic proteins, in particular large ones such as immune checkpoint blockade antibodies. In our design, therapeutic proteins are mixed with FCS to form nanoparticles, lyophilized with appropriate excipients, and then filled into enteric capsules for oral administration. It has been found that FCS could promote transmucosal delivery of its cargo protein via inducing transitory rearrangement of tight junction associated proteins between intestinal epithelial cells and subsequently release free proteins into blood circulation. It is shown that at a 5-fold dose oral delivery of anti-programmed cell death protein-1 (αPD1) or its combination with anti-cytotoxic T-lymphocyte antigen 4 (αCTLA4) using this method could achieve comparable antitumor therapeutic responses to that achieved by intravenous injection of corresponding free antibodies in various types of tumor models and, more excitingly, result in significantly reduced immune-related adverse events. Our work successfully demonstrates the enhanced oral delivery of antibody drugs to achieve systemic therapeutic responses and may revolutionize the future clinical usage of protein therapeutics.


Assuntos
Excipientes , Nanopartículas , Anticorpos , Polímeros , Imunoterapia
8.
Sci Adv ; 9(4): eabq3104, 2023 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-36706184

RESUMO

Therapeutic antibodies are extensively used to treat fundus diseases by intravitreal injection, as eyedrop formulation has been rather challenging due to the presence of ocular barriers. Here, an innovative penetrating carrier was developed for antibody delivery in eyedrop formulations. We found that fluorocarbon-modified chitosan (FCS) would self-assemble with proteins to form nanocomplexes, which could effectively pass across the complicated ocular structure to reach the posterior eye segments in both mice and rabbits. In a choroidal melanoma-bearing mouse model, eyedrops containing FCS/anti-PDL1 could induce stronger antitumor immune responses than those triggered by intravenous injection of anti-PDL1. Moreover, in choroidal neovascularization-bearing mouse and rabbit models, FCS/anti-VEGFA eyedrops effectively inhibited vascular proliferation, achieving comparable therapeutic responses to those observed with intravitreal injection of anti-VEGFA. Our work presents an effective delivery carrier to treat fundus diseases using eyedrop of therapeutic proteins, which may enable at-home treatment of many eye diseases with great patient compliance.


Assuntos
Neovascularização de Coroide , Coelhos , Animais , Camundongos , Soluções Oftálmicas , Fundo de Olho , Modelos Animais de Doenças , Neovascularização de Coroide/tratamento farmacológico
9.
Adv Mater ; 35(2): e2204994, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36349821

RESUMO

Age-related macular degeneration (AMD) is a major cause of visual impairment and severe vision loss worldwide, while the currently available treatments are often unsatisfactory. Previous studies have demonstrated both inflammation and oxidative-stress-induced damage to the retinal pigment epithelium are involved in the pathogenesis of aberrant development of blood vessels in wet AMD (wet-AMD). Although antivascular endothelial growth factor (VEGF) therapy (e.g., Ranibizumab) can impair the growth of new blood vessels, side effects are still found with repeated monthly intravitreal injections. Here, an injectable antibody-loaded supramolecular nanofiber hydrogel is fabricated by simply mixing betamethasone phosphate (BetP), a clinic anti-inflammatory drug, anti-VEGF, the gold-standard anti-VEGF drug for AMD treatment, with CaCl2 . Upon intravitreal injection, such BetP-based hydrogel (BetP-Gel), while enabling long-term sustained release of anti-VEGF to inhibit vascular proliferation in the retina and attenuate choroidal neovascularization, can also scavenge reactive oxygen species to reduce local inflammation. Remarkably, such BetP-Gel can dramatically prolong the effective treatment time of conventional anti-VEGF therapy. Notably, anti-VEGF-loaded supramolecular hydrogel based on all clinically approved agents may be readily translated into clinical use for AMD treatment, with the potential to replace the current anti-VEGF therapy.


Assuntos
Nanofibras , Degeneração Macular Exsudativa , Humanos , Inibidores da Angiogênese/uso terapêutico , Fator A de Crescimento do Endotélio Vascular , Hidrogéis/uso terapêutico , Degeneração Macular Exsudativa/tratamento farmacológico , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Inflamação/tratamento farmacológico
10.
Small ; 17(52): e2104773, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34729889

RESUMO

Tumor radiofrequency ablation (RFA) is a local and minimally invasive application using high temperature to induce coagulative necrosis of tumor, which has been commonly used in clinic. Although the tumor fragments generated by RFA can activate the host's immune system, it may be insufficient to inhibit cancer recurrence due to many factors such as the inefficient antigen presentation by dendritic cells (DCs). In this research, a convenient local administration strategy by blocking rho-associated kinases (ROCK) is applied to amplify the immune responses triggered by RFA via promoting the phagocytosis capacity of DCs. Briefly, ROCK inhibitor, Y27632, is successfully dispersed in the amphiphilic copolymer poly(D,L-lactide-co-glycolide)-b-poly(ethyleneglycol)-b-poly(D,L-lactideco-glycolide) (PLGA-PEG-PLGA) solution, which is sol at room temperature and forms hydrogel quickly at body temperature, obviously prolonging the retention of Y27632 after injection. Interestingly, in the melanoma tumor model, the generated tumor fragments after RFA treatment are swallowed by DCs and undergo reinforced antigen presentation process with the help of gradual released Y27632, further effectively activating T cell mediated anti-tumor immune responses and significantly improving the therapeutic efficiency of RFA. Overall, such strategy remarkably prolongs the survival of mice after RFA treatment, showing great potential for clinical translation as an improvement strategy for RFA.


Assuntos
Neoplasias , Ablação por Radiofrequência , Animais , Hidrogéis , Imunidade , Imunoterapia , Camundongos
11.
Adv Mater ; 33(9): e2007888, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33491820

RESUMO

Radiodynamic therapy (RDT), an emerging therapeutic approach for cancer treatment by employing ionizing irradiation to induce localized photodynamic therapy (PDT) can overcome the drawbacks of the limited penetration depth for traditional PDT and the unconcentrated energy in the tumor for traditional radiotherapy (RT). Taking advantage of aggregation-induced emission (AIE) photosensitizers with bright fluorescence and efficient singlet oxygen production in the aggregate state, Hf-AIE coordination polymer nanoparticles (CPNs), which show both strong RT and RDT effect under X-ray irradiation, are developed. Furthermore, to enhance the tumor accumulation and prolong the tumor retention of the CPNs, bioorthogonal click chemistry is applied in the system through coupling between dibenzocyclooctyne (DBCO)-modified CPNs (Hf-AIE-PEG-DBCO) (PEG: poly(ethylene glycol)) and azide groups on the cell membrane formed by metabolic glycoengineering. Thanks to the high penetration of X-ray irradiation, the bioorthogonal-assisted RT and RDT combination therapy realizes significant killing of cancer cells without showing noticeable biotoxicity after intravenous administration of CPNs.


Assuntos
Antineoplásicos/química , Háfnio/química , Nanopartículas/química , Neoplasias/radioterapia , Fármacos Fotossensibilizantes/química , Animais , Antineoplásicos/uso terapêutico , Transporte Biológico , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Proliferação de Células , Ciclo-Octanos/química , Humanos , Camundongos , Neoplasias Experimentais , Ácidos Fosfatídicos/química , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Polietilenoglicóis/química , Oxigênio Singlete/química
12.
ACS Appl Mater Interfaces ; 12(45): 50248-50259, 2020 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-33135879

RESUMO

Chemotherapeutic agents have been widely used for cancer treatment in clinics. Aside from their direct cytotoxicity to cancer cells, some of them could activate the immune system of the host, contributing to the enhanced antitumor activity. Here, the reactive oxygen species (ROS)-responsive hydrogel, covalently cross-linked by phenylboronic acid-modified 7-ethyl-10-hydroxycamptothecin (SN38-SA-BA) with poly(vinyl alcohol) (PVA), is fabricated for topical delivery of anti-programmed cell death protein ligand 1 antibodies (aPDL1). In the presence of endogenous ROS, SN38-SA-BA will be oxidized and hydrolyzed, leading to the degradation of hydrogel and the release of initial free SN38 and encapsulated aPDL1. It is demonstrated that SN38 could elicit specific immune responses by triggering immunogenic cell death (ICD) of cancer cells, a distinct cell death pathway featured with the release of immunostimulatory damage-associated molecular patterns (DAMPs). Meanwhile, the released aPDL1 could bind to programmed cell death protein ligand 1 (PDL1) expressed on cancer cells to augment antitumor T cell responses. Thus, the ROS-responsive prodrug hydrogel loaded with aPDL1 could induce effective innate and adaptive antitumor immune responses after local injection, significantly inhibiting or even eliminating those tumors.


Assuntos
Antineoplásicos/farmacologia , Imunoterapia , Irinotecano/farmacologia , Pró-Fármacos/farmacologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Ácidos Borônicos/química , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Feminino , Irinotecano/química , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Tamanho da Partícula , Álcool de Polivinil/química , Pró-Fármacos/síntese química , Pró-Fármacos/química , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Espécies Reativas de Oxigênio/imunologia , Propriedades de Superfície , Linfócitos T/imunologia
13.
Biomater Sci ; 8(21): 5846-5858, 2020 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-33016276

RESUMO

With the development of phototherapy, which is a type of light-induced cancer treatment, various biomaterials have been well designed as photoabsorbing/sensitizing agents or effective carriers to enhance the therapeutic efficacy and evade the side effects of phototherapy. In recent years, the immunological responses induced by phototherapy have been widely explored, which are mainly triggered by the tumor associated antigens (TAAs) released from the dying cancer cells after phototherapy, together with the secretion of damage associated molecular patterns (DAMPs) and various pro-inflammatory cytokines/factors. To amplify these immunological responses induced by phototherapy, various adjuvant nano/micromaterials are introduced to boost the immune system to recognize and kill cancer cells. Moreover, such immune responses are further demonstrated to work in synergy with other immunotherapies such as immune checkpoint blockade (ICB), chimeric antigen receptor (CAR)-T cell and cytokine therapy, achieving significantly increased immune response rates and successful therapeutic outcomes. Here, this minireview will focus on the recent progress in engineering biomaterials for enhanced photoimmunotherapy and discuss the challenges, opportunities and future prospects in this field.


Assuntos
Materiais Biocompatíveis , Neoplasias , Antígenos de Neoplasias , Humanos , Imunoterapia , Neoplasias/terapia , Fototerapia
14.
Nano Lett ; 20(9): 6763-6773, 2020 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-32787149

RESUMO

Despite the great promise achieved by immune checkpoint blockade (ICB) therapy in harnessing the immune system to combat different tumors, limitations such as low objective response rates and adverse effects remain to be resolved. Here, an anti-inflammatory nanofiber hydrogel self-assembled by steroid drugs is developed for local delivery of antiprogrammed cell death protein ligand 1 (αPDL1). Interestingly, on the one hand this carrier-free system based on steroid drugs can reprogram the pro-tumoral immunosuppressive tumor microenvironment (TME) to antitumoral TME; on the other hand, it would serve as a reservoir for sustained release of αPDL1 so as to synergistically boost the immune system. By local injection of such αPDL1-loaded hydrogel, effective therapeutic effects were observed in inhibiting both local tumors and abscopal tumors without any treatment. This work presents a unique hydrogel-based delivery system using clinically approved drugs, showing promise in improving the objective response rate of ICB therapy and minimizing its systemic toxicity.


Assuntos
Hidrogéis , Nanofibras , Anti-Inflamatórios , Hidrogéis/farmacologia , Imunoterapia , Microambiente Tumoral
15.
Nat Commun ; 11(1): 3712, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32709842

RESUMO

Metal hydrides have been rarely used in biomedicine. Herein, we fabricate titanium hydride (TiH1.924) nanodots from its powder form via the liquid-phase exfoliation, and apply these metal hydride nanodots for effective cancer treatment. The liquid-phase exfoliation is an effective method to synthesize these metal hydride nanomaterials, and its efficiency is determined by the matching of surface energy between the solvent and the metal hydrides. The obtained TiH1.924 nanodots can produce reactive oxygen species (ROS) under ultrasound, presenting a highly efficient sono-sensitizing effect. Meanwhile, TiH1.924 nanodots with strong near-infrared (NIR) absorbance can serve as a robust photothermal agent. By using the mild photothermal effect to enhance intra-tumoral blood flow and improve tumor oxygenation, a remarkable synergistic therapeutic effect is achieved in the combined photothermal-sonodynamic therapy. Importantly, most of these TiH1.924 nanodots can be cleared out from the body. This work presents the promises of functional metal hydride nanomaterials for biomedical applications.


Assuntos
Antineoplásicos/química , Nanopartículas/uso terapêutico , Nanoestruturas/química , Titânio/química , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Nanotecnologia , Espécies Reativas de Oxigênio , Difração de Raios X
16.
Nanomicro Lett ; 12(1): 100, 2020 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-34138094

RESUMO

Radiotherapy (RT) is a widely used way for cancer treatment. However, the efficiency of RT may come with various challenges such as low specificity, limitation by resistance, high dose and so on. Nitric oxide (NO) is known a very effective radiosensitizer of hypoxic tumor. However, NO cannot circulate in body with high concentration. Herein, an NIR light-responsive NO delivery system is developed for controlled and precisely release of NO to hypoxic tumors during radiotherapy. Tert-Butyl nitrite, which is an efficient NO source, is coupled to Ag2S quantum dots (QDs). NO could be generated and released from the Ag2S QDs effectively under the NIR irradiation due to the thermal effect. In addition, Ag is also a type of heavy metal that can benefit the RT therapy. We demonstrate that Ag2S NO delivery platforms remarkably maximize radiotherapy effects to inhibit tumor growth in CT26 tumor model. Furthermore, immunosuppressive tumor microenvironment is improved by our NO delivery system, significantly enhancing the anti-PD-L1 immune checkpoint blockade therapy. 100% survival rate is achieved by the radio-immune combined therapy strategy based on the Ag2S NO delivery platforms. Our results suggest the promise of Ag2S NO delivery platforms for multifunctional cancer radioimmunotherapy.

17.
Biomaterials ; 228: 119568, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31677393

RESUMO

A general therapeutic strategy to treat breast cancer is attractive as different subtypes of breast cancers often exhibit distinct response to existing cancer therapeutics. To this end, we prepare a catalyst couple of glucose oxidase (GOx) and gallic acid-ferrous (GA-Fe) nanocomplexes, a type of near-infrared (NIR) absorbing Fenton catalyst, to enable NIR-trigger in-situ gelation and enhanced chemodynamic/starvation therapy that appears to be effective for different types of breast cancer cells. In this system, GOx is mixed with GA-Fe in a solution of N,N-dimethylacrylamide (DMAA) and poly (ethylene glycol) double acrylate (PEGDA). Upon intratumoral injection and NIR laser exposure, such GA-Fe show rapid temperature increase, which would simultaneously increase the catalytic efficiencies of GA-Fe and GOx. The cascade production of hydroxyl radicals (•OH) from glucose is then initiated to enable polymerization of DMAA and PEGDA to form a hydrogel at the injection site within the tumor. The continuous production of cytotoxic •OH together with glucose depletion by the intratumorally fixed catalyst couple would further confer effective destruction of breast cancer tumors by such chemodynamic/starvation therapy. Our work presents a hydrogel-based therapeutic strategy for local treatment of solid tumors with high tumor destruction efficacy and low systemic toxicity.


Assuntos
Neoplasias da Mama , Hipertermia Induzida , Neoplasias da Mama/tratamento farmacológico , Glucose , Glucose Oxidase , Humanos , Radical Hidroxila
18.
Chem Soc Rev ; 48(22): 5506-5526, 2019 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-31589233

RESUMO

Cancer immunotherapy by educating or stimulating patients' own immune systems to attack cancer cells has demonstrated promising therapeutic responses in the clinic. However, although the number of approved immunotherapeutics is rapidly increasing, key challenges such as limited clinical response rate and significant autoimmunity-related adverse effects remain to be resolved. Recently, it has been discovered that a diverse range of biomaterials-assisted local treatment methods including localized radiotherapy, chemotherapy or phototherapy are able to stimulate the immune systems, often by inducing immunogenic cell death (ICD). The triggered tumor-specific immunological responses after such local treatments, especially in combination with immune checkpoint blockade (ICB) therapy, can achieve a significant abscopal effect to attack whole-body spreading metastatic cancer cells, and later on result in immune memory to inhibit tumor recurrence. Moreover, local delivery of immunomodulatory therapeutics with biomaterials has also been demonstrated to be an alternative strategy to improve the therapeutic responses and reduce side effects of cancer immunotherapy. In this review, we would like to summarize the latest advances, challenges and opportunities in utilizing biomaterials-assisted local treatment strategies for enhancing anticancer immunity, and discuss further prospects in this field together with how this strategy may possibly be translated into clinical use.


Assuntos
Antineoplásicos/uso terapêutico , Materiais Biocompatíveis/química , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Humanos , Neoplasias/patologia
19.
Nano Lett ; 19(2): 805-815, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30592897

RESUMO

Amplification of intracellular oxidative stress has been found to be an effective strategy to induce cancer cell death. To this end, we prepare a unique type of ultrasmall gallic acid-ferrous (GA-Fe(II)) nanocomplexes as the catalyst of Fenton reaction to enable persistent conversion of H2O2 to highly cytotoxic hydroxyl radicals (•OH). Then, both GA-Fe(II) and l-buthionine sulfoximine (BSO), an inhibitor of glutathione (GSH) synthesis, are coencapsulated within a stealth liposomal nanocarrier. Interestingly, the obtained BSO/GA-Fe(II)@liposome is able to efficiently amplify intracellular oxidative stress via increasing •OH generation and reducing GSH biosynthesis. After chelating with 99mTc4+ radioisotope, such BSO/GA-Fe(II)@liposome could be tracked under in vivo single-photon-emission-computed-tomography (SPECT) imaging, which illustrates the time-dependent tumor homing of such liposomal nanoparticles after intravenous injection. With GA-Fe(II)-mediated •OH production and BSO-mediated GSH depletion, treatment with such BSO/GA-Fe(II)@liposome would lead to dramatically enhanced intratumoral oxidative stresses, which then result in remarkably improved therapeutic efficacies of concurrently applied chemotherapy or radiotherapy. This work thus presents the concise fabrication of biocompatible BSO/GA-Fe(II)@liposome as an effective adjuvant nanomedicine to promote clinically used conventional cancer chemotherapy and radiotherapy, by greatly amplifying the intratumoral oxidative stress.


Assuntos
Butionina Sulfoximina/uso terapêutico , Compostos Ferrosos/uso terapêutico , Ácido Gálico/uso terapêutico , Glutationa/antagonistas & inibidores , Neoplasias Mamárias Animais/terapia , Estresse Oxidativo/efeitos dos fármacos , Animais , Butionina Sulfoximina/administração & dosagem , Linhagem Celular Tumoral , Feminino , Compostos Ferrosos/administração & dosagem , Ácido Gálico/administração & dosagem , Glutationa/metabolismo , Radical Hidroxila/metabolismo , Lipossomos/química , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/radioterapia , Camundongos , Camundongos Endogâmicos BALB C , Tomografia Computadorizada de Emissão de Fóton Único
20.
Biomaterials ; 181: 81-91, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30077139

RESUMO

Owing to the existence of severe tumor hypoxia and limited X-ray absorption of solid tumors, the therapeutic efficacy of radiotherapy is far from satisfactory. Herein, ultrasmall iridium nanocrystals (IrNCs) with homogeneous size distribution are successfully synthesized. The obtained IrNCs show catalase-like catalytic activity towards hydrogen peroxide (H2O2) with great temperatures/pH stability. As free IrNCs are prone to be toxified by thiol-containing biomolecules, we encapsulate as-prepared IrNCs within stealth liposomal carriers, obtaining Ir@liposome with well-protected catalytic activity in physiological conditions. By utilizing its efficient photothermal conversion ability, such Ir@liposome shows effective near-infrared-(NIR)-responsive catalytic activity towards H2O2 decomposition. As revealed by in vivo photoacoustic imaging, our Ir@liposome exhibits efficient passive tumor accumulation upon intravenous injection, and could efficiently decompose the tumor endogenous H2O2 into O2, particularly upon exposure to the NIR laser. As the results of relieved tumor hypoxia after such treatment and the radiosensitization capability of Ir as a high-Z element, greatly enhanced radio-therapeutic efficacy with Ir@liposome is then achieved. This work thus presents a unique type of NIR light controllable theranostic nanozyme based on noble metal nanocrystals as a nanoscale radiosensitizer with great performance in enhancing cancer radiotherapy.


Assuntos
Raios Infravermelhos , Irídio/química , Lipossomos/química , Nanopartículas/química , Neoplasias/radioterapia , Linhagem Celular Tumoral , Humanos , Peróxido de Hidrogênio/química , Concentração de Íons de Hidrogênio , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...