Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Pharm ; 21(3): 1537-1547, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38356224

RESUMO

Mitochondria-targeting photothermal therapy could significantly enhance the tumor cell killing effect. However, since therapeutic reagents need to overcome a series of physiological obstacles to arrive at mitochondria accurately, precise mitochondria-targeting photothermal therapy still faces great challenges. In this study, we developed a self-delivery nanoplatform that specifically targeted the mitochondria of tumor cells for precise photothermal therapy. Photothermal agent IR780 was encapsulated by amphiphilic apoptotic peptide KLA with mitochondria-targeting ability to form nanomicelle KI by self-assembly through hydrophilic and hydrophobic interactions. Subsequently, negatively charged tumor-targeting polymer HA was coated on the surface of KI through electrostatic interactions, to obtain tumor mitochondria-targeting self-delivery nanoplatform HKI. Through CD44 receptor-mediated recognition, HKI was internalizated by tumor cells and then disassembled in an acidic environment with hyaluronidase in endosomes, resulting in the release of apoptotic peptide KLA and photothermal agent IR780 with mitochondria anchoring capacity, which achieved precise mitochondria guidance and destruction. This tumor mitochondria-targeting self-delivery nanoplatform was able to effectively deliver photothermal agents and apoptotic peptides to tumor cell mitochondria, resulting in precise destruction to mitochondria and enhancing tumor cell inhibition at the subcellular organelle level.


Assuntos
Nanopartículas , Neoplasias , Humanos , Terapia Fototérmica , Peptídeos , Mitocôndrias , Apoptose , Nanopartículas/química , Linhagem Celular Tumoral , Fototerapia
2.
Molecules ; 29(16)2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39202916

RESUMO

The diselenide bond has attracted intense interest for drug delivery systems (DDSs) for tumor chemotherapy, owing to it possessing higher redox sensitivity than the disulfide one. Various redox-responsive diselenide-containing carriers have been developed for chemotherapeutics delivery. However, the premature drug leakage from these DDSs was significant enough to cause toxic side effects on normal cells. Here, a pH/redox co-triggered degradable polyprodrug was designed as a drug self-delivery system (DSDS) by incorporating drug molecules as structural units in the polymer main chains, using a facile one-pot two-step approach. The proposed PDOX could only degrade and release drugs by breaking both the neighboring acid-labile acylhydrazone and the redox-cleavable diselenide conjugations in the drug's structural units, triggered by the higher acidity and glutathione (GSH) or reactive oxygen species (ROS) levels in the tumor cells. Therefore, a slow solubility-controlled drug release was achieved for tumor-specific chemotherapy, indicating promising potential as a safe and efficient long-acting DSDS for future tumor treatment.


Assuntos
Antineoplásicos , Oxirredução , Pró-Fármacos , Concentração de Íons de Hidrogênio , Humanos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Liberação Controlada de Fármacos , Espécies Reativas de Oxigênio/metabolismo , Sistemas de Liberação de Medicamentos , Portadores de Fármacos/química , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Polímeros/química , Glutationa/química , Glutationa/metabolismo , Doxorrubicina/química , Doxorrubicina/farmacologia , Doxorrubicina/administração & dosagem
3.
Small ; : e2309994, 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38095445

RESUMO

A systemic treatment strategy is urgently demanded to suppress the rapid growth and easy metastasis characteristics of breast cancer. In this work, a chimeric peptide-engineered self-delivery nanomedicine (designated as ChiP-CeR) for photodynamic-triggered breast cancer immunotherapy by macrophage polarization. Among these, ChiP-CeR is composed of the photosensitizer of chlorine e6 (Ce6) and the TLR7/8 agonist of lmiquimod (R837), which is further modified with tumor matrix targeting peptide (Fmoc-K(Fmoc)-PEG8 -CREKA. ChiP-CeR is preferred to actively accumulate at the tumor site via specific recognition of fibronectin, which can eradicate primary tumor growth through photodynamic therapy (PDT). Meanwhile, the destruction of primary tumors would trigger immunogenic cell death (ICD) effects to release high-mobility group box-1(HMGB1) and expose calreticulin (CRT). Moreover, ChiP-CeR can also polarize M2-type tumor-associated macrophages (TAMs) into M1-type TAMs, which can activate T cell antitumor immunity in combination with ICD. Overall, ChiP-CeR possesses superior antitumor effects against primary and lung metastatic tumors, which provide an applicable nanomedicine and a feasible strategy for the systemic management of metastatic breast cancer.

4.
Small ; 18(4): e2104521, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34821029

RESUMO

High-performance photothermal theranostics is urgently desired for cancer therapy because of their good controllability and noninvasive features. The relatively low photothermal conversion efficiency is still at the drawbacks because of the absence of efficient extraneous carriers. Herein, a carrier-free nanomedicine is developed to in vivo self-deliver organic photothermal agents for efficient cancer phototheranostics. By a facile self-assembly strategy, the near-infrared (NIR)-absorbing conjugated oligomer IDIC-4F is fabricated into a carrier-free nanoparticle (DCF-P), showing ultrasmall size of nearly 4.0 nm with a nearly 100% of drug loading capacity. Notably, DCF-P achieves a superhigh photothermal conversion efficiency of 80.5% that is far greater than that of IDIC-4F-loaded nanomicelle DCF-M (57.3%). With the guidance of NIR fluorescence and photoacoustic dual-imaging, it is verified that DCF-P could well achieve tumor-preferential accumulation and retention at 4 h postinjection, and meanwhile shows highly efficient in vivo tumor elimination with good biosafety. This study thus contributes a novel concept for designing ultrasmall nanoparticle characteristics of preferential accumulation in tumors, and also provides a strategy for creating high-performance carrier-free nanomedicine via highly ordered molecular stacking.


Assuntos
Neoplasias , Técnicas Fotoacústicas , Nanomedicina Teranóstica , Portadores de Fármacos/química , Humanos , Nanopartículas , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Técnicas Fotoacústicas/métodos , Fototerapia/métodos , Medicina de Precisão , Nanomedicina Teranóstica/métodos
5.
Small ; 17(40): e2102470, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34480417

RESUMO

Tumor cells adapt to excessive oxidative stress by actuating reactive oxygen species (ROS)-defensing system, leading to a resistance to oxidation therapy. In this work, self-delivery photodynamic synergists (designated as PhotoSyn) are developed for oxidative damage amplified tumor therapy. Specifically, PhotoSyn are fabricated by the self-assembly of chlorine e6 (Ce6) and TH588 through π-π stacking and hydrophobic interactions. Without additional carriers, nanoscale PhotoSyn possess an extremely high drug loading rate (up to 100%) and they are found to be fairly stable in aqueous phase with a uniform size distribution. Intravenously injected PhotoSyn prefer to accumulate at tumor sites for effective cellular uptake. More importantly, TH588-mediated MTH1 inhibition could destroy the ROS-defensing system of tumor cells by preventing the elimination of 8-oxo-2'-deoxyguanosine triphosphate (8-oxo-dG), thereby exacerbating the oxidative DNA damage induced by the photodynamic therapy (PDT) of Ce6 under light irradiation. As a consequence, PhotoSyn exhibit enhanced photo toxicity and a significant antitumor effect. This amplified oxidative damage strategy improves the PDT efficiency with a reduced side effect by increasing the lethality of ROS without generating superabundant ROS, which would provide a new insight for developing self-delivery nanoplatforms in photodynamic tumor therapy in clinic.


Assuntos
Nanopartículas , Fotoquimioterapia , Porfirinas , Linhagem Celular Tumoral , Estresse Oxidativo , Fármacos Fotossensibilizantes/uso terapêutico , Espécies Reativas de Oxigênio
6.
Nano Lett ; 20(3): 2062-2071, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32096643

RESUMO

Tumor hypoxia is the Achilles heel of oxygen-dependent photodynamic therapy (PDT), and tremendous challenges are confronted to reverse the tumor hypoxia. In this work, an oxidative phosphorylation inhibitor of atovaquone (ATO) and a photosensitizer of chlorine e6 (Ce6)-based self-delivery nanomedicine (designated as ACSN) were prepared via π-π stacking and hydrophobic interaction for O2-economized PDT against hypoxic tumors. Specifically, carrier-free ACSN exhibited an extremely high drug loading rate and avoided the excipient-induced systemic toxicity. Moreover, ACSN not only dramatically improved the solubility and stability of ATO and Ce6 but also enhanced the cellular internalization and intratumoral permeability. Abundant investigations confirmed that ACSN effectively suppressed the oxygen consumption to reverse the tumor hypoxia by inhibiting mitochondrial respiration. Benefiting from the synergistic mechanism, an enhanced PDT effect of ACSN was observed on the inhibition of tumor growth. This self-delivery system for oxygen-economized PDT might be a potential appealing clinical strategy for tumor eradication.


Assuntos
Neoplasias Mamárias Experimentais , Nanomedicina , Nanopartículas , Fotoquimioterapia , Porfirinas , Animais , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Clorofilídeos , Feminino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Nanopartículas/química , Nanopartículas/uso terapêutico , Porfirinas/química , Porfirinas/farmacocinética , Porfirinas/farmacologia
7.
Nano Lett ; 20(4): 2219-2229, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-31823615

RESUMO

Distal metastases of tumors result from the interaction between "seeds" (circulating tumor cells, CTCs) and "soil" (premetastatic niche, PMN). Various strategies focus on CTC inhibition, but only a few strategies inhibit PMN formation. The main predisposition of PMN formation in melanoma lies in the pulmonary recruitment of granulocytic myeloid-derived suppressor cells (G-MDSCs, CD11b+Ly6G+ cells) induced by tumors, which increase vascular permeability by secreting matrix metalloproteinase-9 (MMP-9) and result in immunosuppression by secreting interleukin-10 (IL-10) in premetastatic lungs. Here, a micellar hypotoxic low molecular weight heparin-tocopherol succinate nanoparticle (LMWH-TOS nanoparticle, LT NP) was established and investigated for its influence on PMN formation in this study. We first demonstrated that the hydrophilic segment LMWH in LT NPs can inhibit early pulmonary recruitment of G-MDSCs through interrupting their extravasation by inhibiting P-selectin/PSGL-1-mediated adhesion between vascular endothelial cells and G-MDSCs. In addition, the hydrophobic segment (TOS) in LT NPs significantly inhibited the expression of MMP-9 in G-MDSCs. As a result, the drug-free nanoparticles could maintain the normal microenvironment of lungs, thus effectively inhibiting implantation and colonization of CTCs. Further, phenylboronic acid (PBA)-modified and doxorubicin/immunopotentiator α-galactosylceramide (αGC)-coloaded nanoparticles (PLT/DOX/αGC NPs) were exploited. PBA modification achieved targeted chemotherapy by binding to overexpressed sialic acid residues on the tumor cell surface. This nanosystem effectively inhibited the postoperative metastasis and tumor recurrence simultaneously. Our work provides a proof of concept that the prevention of PMN formation through interfering G-MDSCs with self-delivery nanosystems is a safe and effective antimetastasis strategy.


Assuntos
Células Endoteliais/efeitos dos fármacos , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/tratamento farmacológico , Células Supressoras Mieloides/efeitos dos fármacos , Nanopartículas/uso terapêutico , Células Neoplásicas Circulantes/efeitos dos fármacos , Animais , Células Endoteliais/patologia , Heparina de Baixo Peso Molecular/uso terapêutico , Neoplasias Pulmonares/secundário , Melanoma Experimental/patologia , Camundongos , Micelas , Células Supressoras Mieloides/patologia , Nanomedicina , Células Neoplásicas Circulantes/patologia , Microambiente Tumoral/efeitos dos fármacos , alfa-Tocoferol/análogos & derivados , alfa-Tocoferol/uso terapêutico
8.
Mol Pharm ; 17(2): 710-716, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31910025

RESUMO

Drug self-delivery systems (DSDSs) have attracted intense attention due to their high drug content. However, their practical application still suffers from their premature drug leakage, slow drug release, and/or low antitumor efficacy of the released small molecular drugs. Here, acid-labile poly(Doxazolidine) (P(Doxaz)) is designed as a polyprodrug for the self-delivery of high antitumor chemotherapeutics (Doxazolidine (Doxaz)), with an ultrahigh Doxaz content of 92.45%. The P(Doxaz) nanoparticles could completely degrade into Doxaz within 10 h in the simulated tumor intracellular microenvironment, with a low drug leakage of 12.9% over 12 h in the normal physiological media. Owing to the ultrahigh drug content, fast acid-triggered degradation and drug release, and high antitumor efficacy of Doxaz, the proposed DSDS possesses an enhanced antiproliferation efficacy compared to the free DOX, demonstrating its potential in future tumor treatments.


Assuntos
Acetatos/química , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Doxorrubicina/análogos & derivados , Sistemas de Liberação de Medicamentos/métodos , Oxazóis/síntese química , Oxazóis/farmacologia , Polímeros/síntese química , Pró-Fármacos/síntese química , Pró-Fármacos/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/síntese química , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Nanopartículas/química
9.
Mol Pharm ; 16(9): 3770-3779, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31348660

RESUMO

Drug self-delivery systems consisting of small-molecule active drugs with nanoscale features for intracellular delivery without the need for additional polymeric carriers have drawn much attention recently. In this work, we proposed a highly efficient strategy to fabricate protonized and reduction-responsive self-assembled drug nanoparticles from an amphiphilic small-molecule camptothecin-ss-1,2,3-triazole-gemcitabine conjugate (abbreviated as CPT-ss-triazole-GEM) for combination chemotherapy, which was prepared via a Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) "click" reaction. To obtain this drug-triazole-drug conjugate, we first prepared a CPT derivate containing a propargyl group linked with a disulfide group and a GEM derivate attached to an azide group. Subsequently, the two kinds of modified drugs were connected together through a CuAAC reaction between the alkynyl and azide groups to yield the CPT-ss-triazole-GEM prodrug. The characterizations of chemical structures of these intermediates and the final product were performed by 1H NMR, Fourier transform infrared, and liquid chromatography/mass spectrometry measurements. This amphiphilic small-molecule drug-triazole-drug conjugate displayed a high drug loading content, that is, 36.0% of CPT and 27.2% of GEM. This kind of amphiphilic small-molecule prodrugs could form spherical nanoparticles in an aqueous solution in the absence of any other polymeric carriers, in which the hydrophobic CPT formed the core of the nanoparticles, whereas the hydrophilic GEM and protonated 1,2,3-triazole group yielded the shell. In the tumor microenvironment, the prodrug nanoparticles could release both pristine drugs simultaneously. Under the conditions of pH 7.4, and pH 7.4 and 2 µM glutathione (GSH), the prodrug nanoparticles could maintain stability and only 7% of CPT was leaked. However, in a high-GSH environment (pH 7.4 and 10 mM GSH) with the same incubation time, the disulfide linkage would be dissociated and lead to about 34% of CPT release. The results of the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test demonstrated that these prodrug nanoparticles showed a higher cytotoxicity toward HepG2 cells than free CPT and free GEM on both 48 and 72 h of incubation. Both in vitro cellular uptake and flow cytometry results implied that these prodrug nanoparticles could be internalized by HepG2 cells with efficient drug release inside cells. The pharmacokinetics and tissue distribution of the prodrug showed a moderate half-life in vivo, and the prodrug peak concentration in most of the collected tissues appeared at 0.25 h after administration. In addition, the CPT-ss-triazole-GEM prodrug could not cross the blood-brain barrier. Even more important is the fact that there is no accumulation in tissues and a rapid elimination of this small-molecule prodrug could be achieved. In brief, this protonized and reduction-sensitive prodrug simultaneously binds both antitumor drugs and has good self-delivery behavior through the donor-acceptor interaction of the H-bonding ligand, that is, the 1,2,3-triazole group. It provides a new method for combined drug therapy.


Assuntos
Camptotecina/química , Química Click/métodos , Desoxicitidina/análogos & derivados , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Pró-Fármacos/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/química , Desoxicitidina/farmacocinética , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Meia-Vida , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Masculino , Pró-Fármacos/farmacocinética , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual , Triazóis/química , Triazóis/farmacocinética , Gencitabina
10.
Adv Sci (Weinh) ; 11(15): e2309204, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38239040

RESUMO

The chemo-regulation abilities of chemotherapeutic medications are appealing to address the low immunogenicity, immunosuppressive lactate microenvironment, and adaptive immune resistance of colorectal cancer. In this work, the proteolysis targeting chimera (PROTAC) of BRD4 (dBET57) is found to downregulate colorectal cancer glycolysis through the transcription inhibition of c-Myc, which also inhibits the expression of programmed death ligand 1 (PD-L1) to reverse immune evasion and avoid adaptive immune resistance. Based on this, self-delivery nano-PROTACs (designated as DdLD NPs) are further fabricated by the self-assembly of doxorubicin (DOX) and dBET57 with the assistance of DSPE-PEG2000. DdLD NPs can improve the stability, intracellular delivery, and tumor targeting accumulation of DOX and dBET57. Meanwhile, the chemotherapeutic effect of DdLD NPs can efficiently destroy colorectal cancer cells to trigger a robust immunogenic cell death (ICD). More importantly, the chemo-regulation effects of DdLD NPs can inhibit colorectal cancer glycolysis to reduce the lactate production, and downregulate the PD-L1 expression through BRD4 degradation. Taking advantages of the chemotherapy and chemo-regulation ability, DdLD NPs systemically activated the antitumor immunity to suppress the primary and metastatic colorectal cancer progression without inducing any systemic side effects. Such self-delivery nano-PROTACs may provide a new insight for chemotherapy-enabled tumor immunotherapy.


Assuntos
Antígeno B7-H1 , Neoplasias Colorretais , Humanos , Quimera de Direcionamento de Proteólise , Proteínas Nucleares , Linhagem Celular Tumoral , Fatores de Transcrição , Doxorrubicina/uso terapêutico , Doxorrubicina/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Imunoterapia , Lactatos/farmacologia , Microambiente Tumoral , Proteínas que Contêm Bromodomínio , Proteínas de Ciclo Celular
11.
ACS Appl Mater Interfaces ; 16(26): 33169-33181, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38915234

RESUMO

Inducing immunogenic cell death (ICD) is a promising strategy for cancer immunotherapy. Shikonin (SHK), a naphthoquinone compound from Lithospermum erythrorhizon, can stimulate antitumor immunity by inducing ICD. Nevertheless, the immunogenicity of tumor cells killed by SHK is weak. Endoplasmic reticulum (ER) stress is an important intracellular pathway of the ICD effect. Curcumin (CUR) can directly induce ER stress by disrupting Ca2+ homeostasis, which might enhance SHK-induced ICD effect. A self-delivery ICD effect nanobooster (CS-PEG NPs) was developed by the self-assembly of SHK (ICD inducer) and CUR (ICD enhancer) with the assistance of DSPE-PEG2K for cancer chemoimmunotherapy. CS-PEG NPs possessed effective CT26 tumor cell cellular uptake and tumor accumulation ability. Moreover, enhanced cytotoxicity against tumor cells and apoptosis promotion were achieved due to the synergistic effect of CUR and SHK. Notably, CS-PEG NPs induced obvious Ca2+ homeostasis disruption, ER stress, and ICD effect. Subsequently, the neoantigens produced by the robust ICD effect in vivo promoted dendritic cell maturation, which further recruited and activated cytotoxic T lymphocytes. Superior antitumor efficacy and systemic antitumor immunity were observed in the CT26-bearing BALB/c mouse model without side effects in major organs. This study offers a promising self-delivery nanobooster to induce strong ICD effect and antitumor immunity for cancer chemoimmunotherapy.


Assuntos
Curcumina , Estresse do Retículo Endoplasmático , Morte Celular Imunogênica , Imunoterapia , Camundongos Endogâmicos BALB C , Naftoquinonas , Animais , Naftoquinonas/química , Naftoquinonas/farmacologia , Morte Celular Imunogênica/efeitos dos fármacos , Camundongos , Curcumina/química , Curcumina/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Linhagem Celular Tumoral , Nanopartículas/química , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/patologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Feminino
12.
J Colloid Interface Sci ; 678(Pt A): 494-502, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39214001

RESUMO

Corrosion of steel in the marine environment greatly reduces their service life. Polymeric coatings are the most popular anticorrosion technology, but seawater penetration cannot be prohibited because of the distinct stacking structure of the macromolecular chains. In this context, a novel anticorrosive hyperbranched polyurethane-based coating with dopamine (DOPA) at the terminals is prepared herein. The built-in DOPA is able to capture the iron ions released from the corroded substrate and form DOPA-Fe3+ complexation, which further cooperates with the surrounding seawater and imparts self-passivation, self-delivery and self-healing capabilities to the coating. Under the joint action of these measures, the corrosion of tinplate (serving as the steel model) is reduced to a record-low level (corrosion current = 1 × 10-9 A cm-2, corrosion rate = 1 × 10-5 mm year-1). Conceptually, the present dynamic active anticorrosion strategy greatly outperforms the traditional static passive approach, and turns the unfavorable but unavoidable seawater into a favorable factor, which paves the way for the development of long-lasting marine coatings.

13.
Adv Healthc Mater ; 13(17): e2303896, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38551494

RESUMO

Glutathione (GSH) depletion-induced ferroptosis has emerged as a promising treatment for malignant cancer. It works by inactivating glutathione peroxidase 4 (GPX4) and facilitating lipid peroxidation. However, effectively delivering inducers and depleting intracellular GSH remains challenging due to the short half-lives and high hydrophobicity of small-molecule ferroptosis inducers. These inducers often require additional carriers. Herein, diselenide-containing polymers can consume GSH to induce ferroptosis for pancreatic cancer therapy. The diselenide bonds are controllably built into the backbone of the polycarbonate with a targeting peptide CRGD (Cys-Arg-Gly-Asp), which allows for self-assembly into stable nanoparticles (denoted CRNSe) for self-delivery. Significantly, at a concentration of 12 µg mL-1, CRNSe binds to the active site cysteine of GSH resulting in a thorough depletion of GSH. In contrast, the disulfide-containing analog only causes a slight decrease in GSH level. Moreover, the depletion of GSH inactivates GPX4, ultimately inducing ferroptosis due to the accumulation of lipid peroxide in BxPC-3 cells. Both in vitro and in vivo studies have demonstrated that CRNSe exhibits potent tumor suppressive ability with few side effects on normal tissue. This study validates the anti-tumor mechanism of diselenide-containing polymers in addition to apoptosis and also provides a new strategy for inherently inducing ferroptosis in cancer therapy.


Assuntos
Ferroptose , Glutationa , Ferroptose/efeitos dos fármacos , Humanos , Glutationa/metabolismo , Animais , Linhagem Celular Tumoral , Camundongos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Nanopartículas/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Polímeros/química , Polímeros/farmacologia , Camundongos Nus , Cimento de Policarboxilato/química , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Camundongos Endogâmicos BALB C
14.
Beilstein J Org Chem ; 9: 908-17, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23766806

RESUMO

Here we report supramolecular hydrogelators made of nonsteroidal anti-inflammatory drugs (NSAID) and small peptides. The covalent linkage of Phe-Phe and NSAIDs results in conjugates that self-assemble in water to form molecular nanofibers as the matrices of hydrogels. When the NSAID is naproxen (1), the resultant hydrogelator 1a forms a hydrogel at a critical concentration (cgc) of 0.2 wt % at pH 7.0. Hydrogelator 1a, also acting as a general motif, enables enzymatic hydrogelation in which the precursor turns into a hydrogelator upon hydrolysis catalyzed by a phosphatase at physiological conditions. The conjugates of Phe-Phe with other NSAIDs, such as (R)-flurbiprofen (2), racemic flurbiprofen (3), and racemic ibuprofen (4), are able to form molecular hydrogels, except in the case of aspirin (5). After the conjugation with the small peptides, NSAIDs exhibit improved selectivity to their targets. In addition, the peptides made of D-amino acids help preserve the activities of NSAIDs. Besides demonstrating that common NSAIDs are excellent candidates to promote aromatic-aromatic interaction in water to form hydrogels, this work contributes to the development of functional molecules that have dual or multiple roles and ultimately may lead to new molecular hydrogels of therapeutic agents for topical use.

15.
ACS Nano ; 17(18): 18227-18239, 2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37668306

RESUMO

Nanoprecipitation, which is achieved through the diffusion and precipitation of drug molecules in blended solvent and antisolvent phases, is a classic route for constructing nanodrugs (NDs) and previously directed by diffusion-controlled theory. However, the diffusion-controlled mechanism is out of date in the recent preparation of self-delivery supramolecular NDs (SDSNDs), characterized by the construction of drug nanoparticles through supramolecular interactions in the absence of carriers and surfactants. Herein, a "reaction"-like complement, contributed from supramolecular interactions, is proposed for the preparation of naphthoquinone SDSNDs. Different from the diffusion-controlled process, the formation rate of SDSNDs via the "reaction"-like process is almost constant and highly dependent on the supramolecular interaction-determined Gibbs free energy of molecular binding. Thus, the formation rate and drug availability of SDSNDs are greatly improved by engineering the supramolecular interactions, which facilitates the preparation of SDSNDs with expected sizes, components, and therapeutic functions. As a deep understanding of supramolecular-interaction-involved nanoprecipitation, the current "reaction"-like protocol not only provides a theoretical supplement for classic nanoprecipitation but also highlights the potential of nanoprecipitation in shaping self-assembled, coassembled, and metal-ion-associated SDSNDs.

16.
Adv Healthc Mater ; 12(10): e2202769, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36538727

RESUMO

Drug self-delivery systems (DSDSs) offer new ways to create novel drug delivery systems (DDSs). In typical DSDSs, therapeutic reagents are not considered passive cargos but active delivery agents of actionable targets. As an advanced drug delivery strategy, DSDSs with positive cooperativity of both free drugs and nanocarriers exhibit the clear merits of unprecedented drug-loading capacity, minimized systemic toxicity, and flexible preparation of nanoscale deliverables for passive targeted therapy. This review highlights the recent advances and future trends in DSDSs on the basis of two differently constructed structures: covalent and noncovalent bond-based DSDSs. Specifically, various chemical and architectural designs, fabrication strategies, and responsive and functional features are comprehensively discussed for these two types of DSDSs. In addition, additional comments on the current development status of DSDSs and the potential applications of their molecular designs are presented in the corresponding discussion. Finally, the promising potential of DSDSs in biological applications is revealed and the relationship between preliminary molecular design of DSDSs and therapeutic effects of subsequent DSDSs biological applications is clarified.


Assuntos
Sistemas de Liberação de Medicamentos
17.
Adv Mater ; 35(30): e2301409, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37084041

RESUMO

Small interfering RNA (siRNA) holds immense promise for suppressing gene expression and treating various life-threatening diseases, including cancer. However, efficient delivery and lysosomal escape remain critical challenges that hinder the therapeutic effectiveness of siRNA. Herein, cationic photosensitizer (NB-Br) is grafted onto polo-like kinase 1 (PLK1) siRNA to form an amphiphilic siRNA-photosensitizer conjugate (siPLK1-NB), which can self-assemble into nanoparticles (siPLK1-NB NPs) via electrostatic attraction. Notably, siPLK1-NB NPs exhibit rapid and efficient cell endocytosis, as well as outstanding tumor-targeting property in multiple tumor-bearing mice models. When siPLK1-NB NPs are located inside tumor cell lysosomes, the generated reactive oxygen species (ROS) after photoactivation can disrupt the lysosome membrane structure and facilitate siRNA escape from lysosomes. Under light irradiation, siPLK1-NB NPs can downregulate PLK1 expression and induce photodynamic killing, effectively inhibiting tumor cell growth both in vitro and in vivo. Consequently, this study provides a novel design strategy for carrier-free siRNA delivery systems. As far as it is known, this is the first report of a carrier-free siRNA delivery system based on electrostatic attraction.


Assuntos
Nanopartículas , Fármacos Fotossensibilizantes , Animais , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Eletricidade Estática , Linhagem Celular Tumoral , Terapia Genética , Nanopartículas/química
18.
J Control Release ; 357: 460-471, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37068523

RESUMO

Immune checkpoint blockade (ICB) has shown significant clinical success, yet its responses can vary due to immunosuppressive tumor microenvironments. To enhance antitumor immunity, combining ICB therapy with tumor metabolism reprogramming may be a promising strategy. In this study, we developed a photodynamic immunostimulant called BVC aiming to boost immune recognition and prevent immune escape for metastatic tumor eradication by reprogramming glutamine metabolism. BVC, a carrier free self-assembled nanoparticle, comprises a photosensitizer (chlorin e6), an ASCT2 inhibitor (V9302) and a PD1/PDL1 blocker (BMS-1), offering favorable stability and enhanced drug delivery efficiency. The potent photodynamic therapy (PDT) capability of BVC is attributed to its regulation of glutamine metabolism, which influences the redox microenvironment within tumor tissues. By targeting ASCT2-mediated glutamine metabolism, BVC inhibits glutamine transport and GSH synthesis, leading to the upregulation of Fas and PDL1. Additionally, BVC-mediated PDT induces immunogenic cell death, triggering a cascade of immune responses. Consequently, BVC not only enhances immune recognition between CD8+ T cells and Fas-overexpressing tumor cells but also reduces tumor cell immune escape through PD1/PDL1 blockade, significantly benefiting metastatic tumor eradication. This study paves a novel approach for multi-synergistic tumor treatment.


Assuntos
Glutamina , Fotoquimioterapia , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/uso terapêutico , Imunoterapia , Microambiente Tumoral
19.
ACS Appl Bio Mater ; 6(7): 2816-2825, 2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37326439

RESUMO

Inflammation activation is accompanied by tumor growth, migration, and differentiation. Photodynamic therapy (PDT) can trigger an inflammatory response to cause negative feedback of tumor inhibition. In this paper, a feedback-elevated antitumor amplifier is developed by constructing self-delivery nanomedicine for PDT and cascade anti-inflammation therapy. Based on the photosensitizer chlorin e6 (Ce6) and COX-2 inhibitor indomethacin (Indo), the nanomedicine is prepared via molecular self-assembly technology without additional drug carriers. It is exciting that the optimized nanomedicine (designated as CeIndo) possesses favorable stability and dispersibility in the aqueous phase. Moreover, the drug delivery efficiency of CeIndo is significantly improved, which could be effectively accumulated at the tumor site and internalized by tumor cells. Importantly, CeIndo not only exhibits a robust PDT efficacy on tumor cells but also drastically decreases the PDT-induced inflammatory response in vivo, resulting in feedback-elevated tumor inhibition. By virtue of the synergistic effect of PDT and cascade inflammation suppression, CeIndo tremendously reduces tumor growth and leads to a low side effect. This study presents a paradigm for the development of codelivery nanomedicine for enhanced tumor therapy through inflammation suppression.


Assuntos
Fotoquimioterapia , Humanos , Fotoquimioterapia/efeitos adversos , Fotoquimioterapia/métodos , Nanomedicina , Retroalimentação , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/farmacologia , Inflamação/tratamento farmacológico
20.
Acta Biomater ; 158: 599-610, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36603734

RESUMO

Reactive oxygen species (ROS) generated during photodynamic therapy (PDT) can induce autophagy to protect tumor cell from PDT-induced apoptosis. In this work, a self-delivery autophagy regulator (designated as CeCe) is developed for autophagy promotion sensitized PDT against tumor. Briefly, CeCe is prepared by the assembly of a photosensitizer of chlorin e6 (Ce6) and autophagy promoter of celastrol. By virtue of intermolecular interactions, Ce6 and celastrol are able to self-assemble into nanomedicine with great photodynamic performance and autophagy regulation capacity. Under light irradiation, CeCe would produce ROS in tumor cells to amplify the oxidative stress and promote cell autophagy. As a result, CeCe exhibits an enhanced photo toxicity by inducing autophagic cell death. In vivo experiments indicate that CeCe can predominantly accumulate in tumor tissue for a robust PDT. Moreover, CeCe has a superior therapeutic efficiency compared to monotherapy and combined treatment of Ce6 and celastrol, suggesting a synergistic antitumor effect of PDT and autophagy promotion. This self-delivery nanomedicine may advance the development of the co-delivery nanoplatform to improve the antitumor efficacy of PDT by promoting autophagy. STATEMENT OF SIGNIFICANCE: Autophagy is a "double-edged sword" in cellular homeostasis and metabolism, which can promote tumor progression but also induce an unknown impact on tumor inhibition. In this work, a self-delivery autophagy regulator (designated as CeCe) was developed for autophagy promotion sensitized photodynamic therapy (PDT). By virtue of intermolecular interactions, Ce6 and celastrol were found to self-assemble into stable CeCe without drug excipients, which exhibited great photodynamic performance and autophagy regulation capacity. In vitro and in vivo findings demonstrated a superior tumor suppression ability of CeCe over the monotherapy as well as the combined treatment of Ce6 and celastrol, suggesting a synergistic antitumor efficacy by PDT and autophagy promotion.


Assuntos
Nanopartículas , Fotoquimioterapia , Porfirinas , Espécies Reativas de Oxigênio/metabolismo , Retroalimentação , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Autofagia , Porfirinas/farmacologia , Nanopartículas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA