Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Small ; : e2310865, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38678537

RESUMO

Photopharmacology, incorporating photoswitches such as azobenezes into drugs, is an emerging therapeutic method to realize spatiotemporal control of pharmacological activity by light. However, most photoswitchable molecules are triggered by UV light with limited tissue penetration, which greatly restricts the in vivo application. Here, this study proves that 131I can trigger the trans-cis photoisomerization of a reported azobenezen incorporating PROTACs (azoPROTAC). With the presence of 50 µCi mL-1 131I, the azoPROTAC can effectively down-regulate BRD4 and c-Myc levels in 4T1 cells at a similar level as it does under light irradiation (405 nm, 60 mW cm-2). What's more, the degradation of BRD4 can further benefit the 131I-based radiotherapy. The in vivo experiment proves that intratumoral co-adminstration of 131I (300 µCi) and azoPROTC (25 mg kg-1) via hydrogel not only successfully induce protein degradation in 4T1 tumor bearing-mice but also efficiently inhibit tumor growth with enhanced radiotherapeutic effect and anti-tumor immunological effect. This is the first time that a radioisotope is successfully used as a trigger in photopharmacology in a mouse model. It believes that this study will benefit photopharmacology in deep tissue.

2.
J Mater Chem B ; 12(13): 3240-3248, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38437473

RESUMO

Proteolysis targeting chimeras (PROTACs) is a promising strategy for cancer therapy. However, the always-on bioactivity of PROTACs may lead to non-target toxicity, which restricts their antitumor performance. Here, we developed an X-ray radiation responsive PROTAC nanomicelle (RCNprotac) by covalently conjugating a reported small molecule PROTAC (MZ1) to hydrophilic PEG via a diselenide bond-containing carbon chain, which then self-assembled into a 141.80 ± 5.66 nm nanomicelle. The RCNprotac displayed no bioactivity during circulation due to the occupation of the hydroxyl group on the E3 ubiquitin ligand component and could effectively accumulate at the tumor site owing to the enhanced permeability and retention effect. Upon exposure to X-ray radiation, the radiation-sensitive diselenide bonds were broken to specifically release MZ1 for tumor BRD4 protein degradation. Furthermore, the reduction in the BRD4 protein level could increase the tumor's sensitivity to radiation. RCNprotac showed a synergistic enhancement of antitumor effects both in vitro and in vivo. We believe that this X-ray-responsive PROTAC nanomicelle could provide a new strategy for the X-ray-activated spatiotemporally controlled protein degradation and for the BRD4 proteolysis enhanced tumor radiosensitivity.


Assuntos
Nanopartículas , Neoplasias , Humanos , Proteólise , Proteínas Nucleares/metabolismo , Fatores de Transcrição , Neoplasias/patologia , Proteínas que Contêm Bromodomínio , Proteínas de Ciclo Celular/metabolismo
3.
Acta Pharm Sin B ; 14(2): 795-807, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38322334

RESUMO

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

4.
J Control Release ; 364: 206-215, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37884209

RESUMO

Cerenkov radiation induced photodynamic therapy (CR-PDT) can tackle the tissue penetration limitation of traditional PDT. However, co-delivery of radionuclides and photosensitizer may cause continuous phototoxicity in normal tissues during the circulation. 5-aminolevulinic acid (ALA) which can intracellularly transform into photosensitive protoporphyrin IX (PpIX) is a cancer-selective photosensitizer with negligible side effect. However, the hydrophilic nature of ALA and the further conversion of PpIX to photoinactive Heme severely hinder the therapeutic benefits of ALA-based PDT. Herein, we developed an 89Zr-labeled, pH responsive ALA and artemisinin (ART) co-loaded liposome (89Zr-ALA-Liposome-ART) for highly selective cancer therapy. 89Zr can serve as the internal excitation source to self-activate PpIX for CR-PDT, and the photoinactive Heme can activate the chemotherapeutic effect of ART. The 89Zr-ALA-Liposome-ART exhibited excellent tumor inhibition capability in subcutaneous 4T1-tumor-bearing Balb/c mice via CR-PDT and chemotherapy. Combined with anti-PD-L1, the 89Zr-ALA-Liposome-ART elicited strong antitumor immunity to against tumor recurrence.


Assuntos
Neoplasias , Fotoquimioterapia , Pró-Fármacos , Camundongos , Animais , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/farmacologia , Pró-Fármacos/uso terapêutico , Lipossomos , Ácido Aminolevulínico/uso terapêutico , Protoporfirinas , Neoplasias/tratamento farmacológico , Heme , Linhagem Celular Tumoral
5.
Bioeng Transl Med ; 8(4): e10427, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37476071

RESUMO

The enriched collagens in the extracellular matrix (ECM) of breast cancer substantially impede drug delivery. Halofuginone (HF), a potent antifibrotic agent, was effective to deplete the collagens and remodel the ECM by inhibiting the TGFß pathway. However, the application of HF was hindered by its strong liver toxicity. Herein, mesoporous platinum (mPt) nanoparticles were constructed to load HF as theranostic nanoplatforms. mPt had a uniform spherical structure with a diameter of 79.83 ± 6.97 nm and an average pore diameter of 20 nm and exhibited good photothermal conversion efficiency of 62.4%. The obtained HF-loaded nanoplatform (PEG@mPt-HF) showed enhanced cytotoxicity through the combination of photothermal therapy and the anti-TGFß effect induced by HF. The animal imaging and histochemical assays confirmed the PEG@mPt-HF could efficiently deliver HF to tumors (monitored by CT) and remodel the ECM by TGFß pathway inhibition, which resulted in increased anti-cancer efficacy. Importantly, the liver toxicity observed in HF-treated mice was negligible in those treated by PEG@mPt-HF. Overall, this study designed a theranostic nanoplatform to remodel the ECM with remarkably reduced systematic toxicity and enhance the therapeutic efficacy through combination treatment.

6.
Nanoscale ; 15(26): 11280-11289, 2023 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-37350173

RESUMO

Protein therapy, an innovative therapeutic strategy, has been extensively used in the treatment of cancer in recent years. However, the sequential delivery of multiple proteins acting separately intracellular and extracellular to their sites of action remains a challenge. Here, we construct a nanosystem (PEI-PEG-TRAIL@IONP-GOx) to sequentially release tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) extracellularly and glucose oxidase (GOx) intracellularly for synergistic cancer treatment. The nanosystem is built as a core-shell structure. The core is a pH responsive nanoassembly of boronic acid modified iron oxide nanoparticles (FPBA-IONPs) and polyphenol decorated GOx. The shell is a PEGylated polyethyleneimine (PEI-PEG) polymer on which TRAIL was coupled by a matrix metalloproteinase-2 (MMP-2) responsive peptide. Once the nanosystems were magnetically guided to the tumor site, TRAIL was quickly released by the extracellular MMP-2 to induce tumor apoptosis and enhanced the cellular uptake of the cores. After cytosolic delivery, FPBA-IONPs and GOx were disassembled intracellularly to trigger a cascade reaction to generate free radicals for tumor inhibition. Both in vitro and in vivo experiments proved the separate delivery of TRAIL and GOx and their remarkable synergistic anti-cancer effect. We believe that this nanosystem can offer a new approach for the multistage delivery of proteins and accomplish the objective of protein cooperation for cancer treatment.


Assuntos
Nanopartículas , Neoplasias , Humanos , Metaloproteinase 2 da Matriz , Neoplasias/tratamento farmacológico , Polímeros , Peptídeos , Fenômenos Magnéticos , Linhagem Celular Tumoral , Nanopartículas/química
7.
J Control Release ; 357: 40-51, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36948418

RESUMO

Cancer immunotherapy is an emerging cancer therapeutic method by activating the patient's immune system but suffers from low immunogenicity at tumor sites. Fever-like heat is known to modulate an immune-friendly tumor microenvironment. Here, temperature-responsive iron oxide nanoassemblies (IONAs) are developed by crosslinking iron oxide nanoparticles (IONPs) and loaded with JQ1 (JQ1/IONAs), an immuno-modulating agent known to down-regulate PD-L1. In the presence of an alternating magnetic field (AMF), the IONAs demonstrate a much more effective magnetic thermal effect than IONPs and are responsively disassembled to prevent overheating. Compared with IONPs + AMF (∼ 41 °C) and unresponsive nanoassemblies (uIONAs) + AMF (∼ 50 °C), the IONAs + AMF with a temperature heated around 45 °C show a much better immune response and anti-tumor effect. Further combining the mild thermal therapy with controlled release of JQ1, the JQ1/IONAs + AMF completely eradicate the primary tumors and trigger a strong immune effect to inhibit the distant tumor growth as well as prevent tumor recurrence and metastasis. Our JQ1/IONAs not only provide a magnetic thermal agent with effective heating and temperature self-regulation ability but also serve as a heat-triggered JQ1 carrier to spontaneously combine mild magnetic thermal therapy with immune checkpoint blockade therapy.


Assuntos
Hipertermia Induzida , Neoplasias , Humanos , Hipertermia Induzida/métodos , Temperatura Alta , Campos Magnéticos , Neoplasias/terapia , Linhagem Celular Tumoral , Microambiente Tumoral
8.
Biomater Sci ; 11(6): 2167-2176, 2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36734805

RESUMO

Frequency upconversion luminescence (FUCL) based on hot-band absorption has attracted considerable attention in bioimaging and phototherapy fields for deep-seated cancer treatment. Photoimmunotherapy, a promising therapeutic approach induced by photodynamic therapy (PDT), can selectively kill cancer cells, reverse the immunosuppressive system, boost host immune response, and elicit durable antitumor immunity. To date, few near-infrared organic photosensitizers for photodynamic immunotherapy have been reported based on hot-band absorption. Herein, we report an upconversion luminescent phthalocyanine photosensitizer PdPc(OBu)8 with anti-Stokes emission at 748 nm and highly efficient singlet oxygen generation with hot-band absorption at 808 nm. Taking advantage of nanoliposomes, FUCL phthalocyanine nano-photosensitizers (PdPc NPs) were obtained to reduce the aggregation-caused quenching and improve water solubility and biocompatibility. PdPc NPs could be effectively accumulated in tumor tissues through intravenous administration, causing FUCL-induced PDT under 808 nm irradiation. Considering its finite immune responses and tumor ablation after PDT, a combination of PdPc NP-based PDT with checkpoint inhibitors (anti-PD-L1) for near-infrared photoimmunotherapy has been used to potentiate the antitumor efficacy that could simultaneously ablate primary tumors and inhibit the progression of distant tumors. This study can promote the development of upconversion-based PDT combined with immunotherapy for tumor precision therapy.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Fotoquimioterapia/métodos , Luminescência , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Imunoterapia , Linhagem Celular Tumoral
9.
Nanoscale ; 15(7): 3188-3198, 2023 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-36723141

RESUMO

Multifunctional nanoplatforms with the synergistic effects of multiple therapeutic modalities have become a research focus due to their superior anti-tumor properties over single therapeutic modalities. Herein, we developed around 14 nm porous hollow copper iron oxide nanoparticles (PHCuFeNPs) with pore sizes of around 2-3 nm as a cisplatin carrier and photothermal therapeutic agent. The PHCuFeNPs were synthesized via a galvanic reaction between Cu2S nanoparticles and iron pentacarbonyl (Fe(CO)5) followed by etching in the organic phase to make the pores. They were stable under normal physiological conditions, but the pores were etched in a weak acidic tumor microenvironment, resulting in the controlled release of Cu and Fe ions for enhanced chemodynamic therapy and accelerated cisplatin release for chemotherapy. Under 980 nm laser irradiation, the PHCuFeNPs could effectively heat up to further promote the release process for synergistic therapy. Besides, they were proved to mediate immunogenic cell death to activate the immune system for potential immunotherapy. Together with their ability to degrade into fragments for fast renal metabolism, we believe that these PHCuFeNPs could provide a biocompatible and efficient multi-antitumor therapeutic approach.


Assuntos
Cisplatino , Nanopartículas , Cisplatino/farmacologia , Cobre/farmacologia , Porosidade , Fototerapia/métodos , Nanopartículas/uso terapêutico , Nanopartículas Magnéticas de Óxido de Ferro , Linhagem Celular Tumoral
10.
Biomaterials ; 294: 122017, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36680943

RESUMO

The cancer photodynamic therapy (PDT) is limited by a congenital defect, namely the tumor hypoxia. Cancer cells are characterized by the vigorous oxygen-consuming glycolysis, which is well-known as the "Warburg effect" and one of the primary causes for the hypoxia. Herein, we employed the glucose metabolism as the cancer-specific target to enhance the performance of PDT. The Salvianolic acid B as the inhibitor of glucose uptake and aerobic glycolysis was concomitantly delivered with the photosensitizer chlorin e6 by a redox-responsive organosilica cross-linked micelle. The results demonstrated that the Salvianolic acid B suppressed the glucose metabolism, retarded the oxygen consumption to retain adequate oxygen as the ammo for PDT, which remarkably improve the efficacy of PDT both in vitro and in vivo. Our study not only provides an alternative strategy to address the hypoxia problem for PDT, but also enhances the selectivity of the treatment by targeting the cancer-specific Warburg effect.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Porfirinas , Humanos , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/uso terapêutico , Fármacos Fotossensibilizantes/farmacologia , Fotoquimioterapia/métodos , Porfirinas/uso terapêutico , Porfirinas/farmacologia , Glucose , Oxigênio , Hipóxia/tratamento farmacológico , Neoplasias/tratamento farmacológico
11.
Sci Total Environ ; 855: 158715, 2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36113792

RESUMO

Many nanomaterials containing different valences of iron have been designed for applications in biomedicine, energy, catalyzers, nanoenzymes, and so on. However, the toxic effects of the valence state of iron in iron-based nanomaterials are still unclear. Here, three different-valence iron-based nanomaterials (nFe@Fe3O4, nFe3O4 and nFe2O3) were synthesized and exposed to zebrafish embryos and mammalian cardiomyocytes. All of them induced ferroptosis along with an increase in valence through iron overload and the Fenton reaction. Specifically, we exposed Tg (cmlc2:EGFP) zebrafish to the three iron-based nanomaterials and found that nFe@Fe3O4 treatments led to enlarged ventricles, while nFe3O4 and nFe2O3 increased atrial size, which was consistent with the results from hematoxylin-eosin staining and in situ hybridization. Moreover, we used ferroptosis inhibitors (ferrostatin-1 or deferoxamine) to treat zebrafish along with nanoparticles exposure and found that the cardiac developmental defects caused by nFe3O4 and nFe2O3, but not nFe@Fe3O4, could be completely rescued by ferroptosis inhibitors. We further found that nFe@Fe3O4, rather than nFe3O4 and nFe2O3, reduced the dissolved oxygen in the medium, which resulted in hypoxia and acceleration of heart tube formation and ventricular enlargement, and both were fully rescued by oxygen donors combined with ferroptosis inhibitors. Consistently, these findings were also observed in mammalian cardiomyocytes. In summary, our study demonstrates that the valence state of iron-based nanomaterials determines the ferroptosis potential. Our study also clarifies that high-valence iron-based nanomaterials induce an enlarged atrium via ferroptosis, while low-valence ones increase the ventricular size through both hypoxia and ferroptosis, which is helpful to understand the potential adverse effects of different valences of iron-based nanomaterials on environmental health and assure the responsible and sustainable development of nanotechnology.


Assuntos
Ferroptose , Nanoestruturas , Animais , Ferro/toxicidade , Peixe-Zebra , Nanoestruturas/toxicidade , Hipóxia , Oxigênio , Mamíferos
12.
Theranostics ; 12(17): 7404-7419, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36438500

RESUMO

Cerenkov radiation (CR) from radionuclides and megavoltage X-ray radiation can act as an in situ light source for deep cancer theranostics, overcoming the limitations of external light sources. Despite the blue-weighted emission and low quantum yield of CR, activatable probes-mediated CR can enhance the in-vivo diagnostic signals by Cerenkov resonance energy transfer and also can produce therapeutic effects by reactive species generation/drug release, greatly promoting the biomedical applications of CR. In this review, we describe the principles and sources of CR, construction of CR-activated probes and their application to tumor optical imaging and therapy. Finally, future prospects for the design and biomedical application of CR-activated probes are discussed.


Assuntos
Neoplasias , Medicina de Precisão , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/radioterapia , Imagem Óptica , Liberação Controlada de Fármacos , Vibração
13.
J Med Chem ; 64(21): 15702-15715, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34670362

RESUMO

Dual inhibition of tubulin and poly(ADP-ribose) polymerase-1 (PARP-1) may become an attractive approach for cancer therapy. Here, we discover a dual tubulin/PARP-1 inhibitor (termed as TP-3) using structure-based virtual screening. TP-3 shows strong dual inhibitory effects on both tubulin and PARP-1. Cellular assays reveal that TP-3 shows superior antiproliferative activities against human cancer cells, including breast, liver, ovarian, and cervical cancers. Further studies indicate that TP-3 plays an antitumor role through multiple mechanisms, including the disturbance of the microtubule network and the PARP-1 DNA repairing function, accumulation of DNA double-strand breaks, inhibition of the tube formation, and induction of G2/M cell cycle arrest and apoptosis. In vivo assessment indicates that TP-3 inhibits the growth of MDA-MB-231 xenograft tumors in nude mouse with no notable side effects. These data demonstrate that TP-3 is a dual-targeting, high-efficacy, and low-toxic antitumor agent.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Descoberta de Drogas , Inibidores de Poli(ADP-Ribose) Polimerases/química , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Animais , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Ensaios de Seleção de Medicamentos Antitumorais , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Modelos Moleculares , Simulação de Acoplamento Molecular , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Nanobiotechnology ; 19(1): 350, 2021 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-34717646

RESUMO

BACKGROUND: Nanoscale drug delivery systems have emerged as broadly applicable approach for chemo-photothermal therapy. However, these nanoscale drug delivery systems suffer from carrier-induced toxicity, uncontrolled drug release and low drug carrying capacity issues. Thus, to develop carrier-free nanoparticles self-assembled from amphiphilic drug molecules, containing photothermal agent and anticancer drug, are very attractive. RESULTS: In this study, we conjugated camptothecin (CPT) with a photothermal agent new indocyanine green (IR820) via a redox-responsive disulfide linker. The resulting amphiphilic drug-drug conjugate (IR820-SS-CPT) can self-assemble into nanoparticles (IR820-SS-CPT NPs) in aqueous solution, thus remarkably improving the membrane permeability of IR820 and the aqueous solubility of CPT. The disulfide bond in the IR820-SS-CPT NPs could be cleaved in GSH rich tumor microenvironment, leading to the on demand release of the conjugated drug. Importantly, the IR820-SS-CPT NPs displayed an extremely high therapeutic agent loading efficiency (approaching 100%). Besides, in vitro experimental results indicated that IR820-SS-CPT NPs displayed remarkable tumor cell killing efficiency. Especially, the IR820-SS-CPT NPs exhibited excellent anti-tumor effects in vivo. Both in vitro and in vivo experiments were conducted, which have indicated that the design of IR820-SS-CPT NPs can provide an efficient nanotherapeutics for chemo-photothermal therapy. CONCLUSION: A novel activatable amphiphilic small molecular prodrug IR820-SS-CPT has been developed in this study, which integrated multiple advantages of GSH-triggered drug release, high therapeutic agent content, and combined chemo-photothermal therapy into one drug delivery system.


Assuntos
Camptotecina/administração & dosagem , Camptotecina/química , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Terapia Fototérmica/métodos , Pró-Fármacos/química , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Humanos , Verde de Indocianina , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fototerapia , Solubilidade
15.
J Am Chem Soc ; 143(38): 15812-15823, 2021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34473493

RESUMO

Iron-based nanoparticles have attracted much attention because of their ability to induce ferroptosis via a catalyzing Fenton reaction and to further potentiate immunotherapy. However, current iron-based nanoparticles need to be used in cooperation with other treatments or be applied in a high dose for effective therapy because of their low reactive oxygen species production efficacy. Here, we synthesized ultrasmall single-crystal Fe nanoparticles (bcc-USINPs) that stayed stable in a normal physiological environment but were highly active in a tumor microenvironment because of the selective acidic etching of an Fe3O4 shell and the exposure of the Fe(0) core. The bcc-USINPs could efficiently induce tumor cell ferroptosis and immunogenetic cell death at a very low concentration. Intravenous injection of iRGD-bcc-USINPs at three doses of 1 mg/kg could effectively suppress the tumor growth, promote the maturation of dendritic cells, and trigger the adaptive T cell response. Combined with programmed death-ligand 1 (PD-L1) immune checkpoint blockade immunotherapy, the iRGD-bcc-USINP-mediated ferroptosis therapy greatly potentiated the immune response and developed strong immune memory. In addition, these USINPs were quickly renal excreted with no side effects in normal tissues. These iRGD-bcc-USINPs provide a simple, safe, effective, and selectively tumor-responsive Fe(0) delivery system for ferroptosis-based immunotherapy.


Assuntos
Antineoplásicos/química , Ferroptose/efeitos dos fármacos , Ferro/química , Nanopartículas Metálicas/química , Animais , Antineoplásicos/farmacocinética , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Imunoterapia , Ferro/farmacocinética , Rim , Camundongos , Terapia de Alvo Molecular , Espécies Reativas de Oxigênio/metabolismo , Especificidade por Substrato , Microambiente Tumoral
16.
Nanoscale ; 13(31): 13231-13240, 2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34477731

RESUMO

Although artemisinin (ART) has shown initial promise in cancer therapy, its therapeutic efficacy is limited by its low tumor inhibitory efficacy and unfavorable distribution. Considering the important role of heme in the specific parasite-killing effect of ART, we designed a liposomal nanostructure self-assembled from hemin-lipid (Hemesome) to co-deliver ART and hemin for cancer therapy. The synergistic chemotherapeutic and immunotherapeutic effects of hemin and ART were demonstrated both in vitro and in vivo. The liposome-like structure was relatively stable in the blood circulation and gastrointestinal tract environment, but dissociated in the tumor cell environment. The folic acid (FA) modification not only increased their efficiency for transport across the epithelium, but also increased their tumor accumulation. In mouse models, following oral administration of FA-Hemesome-ART nanoparticles (5 mg kg-1 ART in total) every other day and intraperitoneal injection with a programmed death-ligand 1 antibody (aPD-L1, 70 µg per mouse in total), MC38 tumors were completely inhibited within 30 days. The cured mice remained tumor-free 30 days after rechallenging them with another inoculation of MC38 cells due to the strong immune memory effect.


Assuntos
Artemisininas , Nanopartículas , Neoplasias , Animais , Linhagem Celular Tumoral , Hemina , Imunoterapia , Lipídeos , Camundongos , Neoplasias/tratamento farmacológico
18.
Angew Chem Int Ed Engl ; 60(40): 21884-21889, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34374188

RESUMO

Stimulating photosensitizers (PS) by Cerenkov radiation (CR) can overcome the light penetration limitation in traditional photodynamic therapy. However, separate injection of radiopharmaceuticals and PS cannot guarantee their efficient interaction in tumor areas, while co-delivery of radionuclides and PS face the problem of nonnegligible phototoxicity in normal tissues. Here, we describe a 131 I-labeled smart photosensitizer, composed of pyropheophorbide-a (photosensitizer), a diisopropylamino group (pH-sensitive group), an 131 I-labeled tyrosine group (CR donor), and polyethylene glycol, which can self-assemble into nanoparticles (131 I-sPS NPs). The 131 I-sPS NPs showed low phototoxicity in normal tissues due to aggregation-caused quenching effect, but could self-produce reactive oxygen species in tumor sites upon disassembly. Upon intravenous injection, 131 I-sPS NPs showed great tumor inhibition capability in subcutaneous 4T1-tumor-bearing Balb/c mice and orthotopic VX2 liver tumor bearing rabbits. We believed 131 I-sPS NPs could expand the application of CR and provide an effective strategy for deep tumor theranostics.


Assuntos
Antineoplásicos/farmacologia , Clorofila/análogos & derivados , Neoplasias Hepáticas/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Clorofila/química , Clorofila/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Radioisótopos do Iodo , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/patologia , Camundongos , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único
19.
Theranostics ; 11(14): 7045-7056, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093870

RESUMO

Rationale: Precise treatment of tumors is attracting increasing attention. Molecular probes simultaneously demonstrating the diagnostic signal and pharmacological effect in response to tumor microenvironment are highly desired. γ-glutamyl transpeptidase (GGT) is a biomarker with significantly up-regulated expression in the tumor area. We developed a GGT responsive near-infrared (NIR) nanoassembly for tumor-specific fluorescence imaging-guided photothermal therapy. Methods: The GGT responsive NIR probe was constructed by conjugating GGT-specific substrate γ-glutamic acid (γ-Glu) with cyanine fluorophore (NRh-NH2) via amide reaction. The resulting NRh-G spontaneously assembled into nanoparticles (NRh-G-NPs) around 50 nm. The NPs were characterized and the properties evaluated in the presence or absence of GGT. Subsequently, we studied fluorescence imaging and photothermal therapy of NRh-G-NPs in vitro and in vivo. Results: NRh-G-NPs, upon specific reaction with GGT, turned into NRh-NH2-NPs, showing a ~180-fold fluorescence enhancement and excellent photothermal effect recovery. NRh-G-NPs could selectively light up U87MG tumor cells while their fluorescence was weak in L02 human normal liver cells. The NPs also showed excellent tumor cell ablation upon laser irradiation. After intravenous injection into tumor-bearing mice, NRh-G-NPs could arrive in the tumor area and specifically light up the tumor. Following laser irradiation, the tumor could be completely erased with no tumor reoccurrence for up to 40 days. Conclusions: NRh-G-NPs were specifically responsive to GGT overexpressed in U87MG tumor cells and selectively lit up the tumor for imaging-guided therapy. Besides, the recovery of photothermal property in the tumor area could improve cancer therapy precision and decreased side effects in normal tissues.


Assuntos
Glioma/tratamento farmacológico , Glioma/radioterapia , Hipertermia Induzida/métodos , Nanopartículas/química , Terapia Fototérmica/métodos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos da radiação , gama-Glutamiltransferase/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Feminino , Fluorescência , Corantes Fluorescentes/química , Ácido Glutâmico/química , Humanos , Lasers , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Espectroscopia de Luz Próxima ao Infravermelho , gama-Glutamiltransferase/genética
20.
Acta Biomater ; 129: 84-95, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34010690

RESUMO

Hydrogels have drawn considerable attention in the field of drug delivery, yet their poor mechanical strength and uncontrollable drug release behavior have hindered further applications in clinical practice. Taking utility of metal-ligand coordination for structurally reinforcing the hydrogel network, we report design and synthesis of magnetic nanocomposite hydrogels (HA-DOPA·MNPs) that are crosslinked by DOPA-Fe(III) coordination existing between dopamine-conjugated hyaluronan (HA-DOPA) and iron oxide magnetic nanoparticles (MNPs). The MNPs in the nanocomposite hydrogel not only serve as structural crosslinkers, but also facilitate magnetic hyperthermia and on-demand release of doxorubicin (DOX) in HA-DOPA·MNPs/DOX hydrogels, for release rate of DOX accelerates when external alternating magnetic field (AMF) is ON, and it restores to a slow pace when AMF is OFF. Importantly, HA-DOPA·MNPs/DOX hydrogel shows a longer retention time than HA-DOPA/DOX gel or DOX solution in vivo. Further experiments confirm the efficacious anticancer potency of HA-DOPA·MNPs/DOX in vitro and in vivo, that is mediated by a combination therapy consisting of chemotherapy (DOX) and hyperthermia (MNPs). In contrast, single-modality treatment (DOX or hyperthermia only) fails to show an equivalent efficacy at the same dose. STATEMENT OF SIGNIFICANCE: This study reports the design of a class of magnetic nanocomposite hydrogel (HA-DOPA·MNPs) that was structurally reinforced by DOPA-Fe (III) coordination between HA-DOPA and iron oxide MNPs. On one hand, MNPs served as crosslinking centers for structurally reinforcing the nanocomposite hydrogel; on the other hand, MNPs facilitated temperature rise under an external MNPs, which prompted on-demand drug release as well as a combination therapy. Comparing to single modality treatment (chemotherapy or hyperthermia alone), the HA-DOPA·MNPs/DOX formulation with AMF demonstrated better efficacy against proliferation of tumor cells (A375) both in vitro and in vivo. We believe that design of HA-DOPA·MNPs/DOX hydrogel in this report provides a general approach to fabricate structurally-reinforced nanocomposite hydrogels for on-demand drug delivery and efficacious combination therapy.


Assuntos
Compostos Férricos , Hipertermia Induzida , Catecóis , Doxorrubicina/farmacologia , Hidrogéis , Nanogéis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA