Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Acta Biomater ; 150: 353-366, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35843594

RESUMO

Immunosuppressive tumor microenvironment (ITM), poor immunogenicity, and low tumor penetration markedly reduce the capability of tumor immunotherapy. To address these challenges, we successfully engineered acidity-triggered nanoparticles (NPs) with size reduction and charge switchable features to boost tumor immunotherapy based on indoleamine 2,3-dioxygenase 1 siRNA (IDO1 siRNA) and immunogenic cell death (ICD). The NPs significantly augmented tumor penetrating ability and improved cellular uptake via the detachment of 2,3-dimethylmaleic anhydride-grafted poly(ethylene glycol)-poly(L-lysine) copolymer (mPEG-PLL-DMA, PLM) from large-sized NPs with a negative charge. Subsequently, the NPs with a positive charge and small size rapidly escaped from the lysosomes and released mitoxantrone (MIT) and IDO1 siRNA. The antitumor immune response of IDO1 siRNA and MIT provided good antitumor capability by enhancing DC maturation, improving the number of CTLs, and downregulating the level of Tregs in tumor tissues. In summary, the results demonstrated that charge-switchable NPs based on the blockage of the IDO1 pathway and ICD activation induce an efficient antitumor immune response, thus showing high potential for treating primary/distant tumors and reducing metastasis. STATEMENT OF SIGNIFICANCE: Acidity-triggered nanoparticles (NPs) with size reduction and charge reversal to boost tumor immunotherapy based on indoleamine 2,3-dioxygenase 1 siRNA (IDO1 siRNA) and immunogenic cell death (ICD) were engineered. NPs augmented tumor penetrating ability and improved cellular uptake through the detachment of mPEG-PLL-DMA (PLM) from the large-sized MIT/siR-PLM/PPA NPs with negative charge to expose miniature and positively charged MIT/siR-PPA NPs. The NPs rapidly escaped from the lysosome and sequentially released mitoxantrone (MIT) and IDO1 siRNA. The antitumor synergistic effect of inhibiting the IDO1 pathway by IDO1 siRNA and inducing ICD by MIT provided good antitumor capability by enhancing DC maturation, improving the number of CTLs, and downregulating the level of Tregs in tumor tissues. Thus, the NPs showed a promising pathway against aggressive and difficult-to-treat cancers.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Morte Celular Imunogênica , Imunoterapia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Mitoxantrona , Neoplasias/terapia , RNA Interferente Pequeno/genética , Microambiente Tumoral
2.
Biomaterials ; 283: 121448, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35245730

RESUMO

Multiple biological barriers and tumor metastasis severely impede the tumor therapy. To address these adversities, an acid-activated poly (ethylene glycol)-poly-l-lysine-2,3-dimethylmaleic anhydride/poly (ε-caprolactone)-poly(l-arginine)/ß-lapachone nanoparticles (mPEG-PLL-DMA/PCL-P(L-arg)/ß-Lap, PLM/PPA/ß-Lap NPs) were constructed with charge-reversal and size-reduction for ß-Lap delivery with a cascade reaction of reactive oxygen species (ROS) and nitric oxide (NO) production. The nanosystem exhibited highly penetrable, superior cellular uptake and desirable endo-lysosomal escape thanks to size-reduction, charge-reversal and proton sponge, respectively. The vast bulk of ROS, which rapidly generated from ß-Lap under high concentration quinone oxidoreductase 1 (NQO1), catalyzed guanidine groups to produce NO and generated highly toxic peroxynitrite (ONOO-). ONOO- would activate pro-matrix metalloproteinases (pro-MMPs) to generate MMPs, degrade the dense extracellular matrix (ECM) to augment the penetration capability, and aggravate DNA damage. NO and ONOO- influenced mitochondrial function by decreasing mitochondrial membrane potential and prevented the production of adenosine triphosphate (ATP), which inhibited the ATP-dependent tumor-derived microvesicles (TMVs) and restrained tumor metastasis. NO was defined as an epithelial mesenchymal transition (EMT) inhibitor to restrain tumor metastasis. All consequences demonstrated that PLM/PPA/ß-lap NPs exhibited efficient penetration capability, outstanding anti-metastasis activity and favorable antitumor efficacy. Those novel acid-activated NPs are intended to provide further inspiration for multifunctional NO gas therapy.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Óxido Nítrico/metabolismo , Ácido Peroxinitroso , Espécies Reativas de Oxigênio
3.
Acta Pharm Sin B ; 12(9): 3694-3709, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36176905

RESUMO

Amplifying "eat me signal" during tumor immunogenic cell death (ICD) cascade is crucial for tumor immunotherapy. Inspired by the indispensable role of adenosine triphosphate (ATP, a necessary "eat me signal" for ICD), a versatile ICD amplifier was developed for chemotherapy-sensitized immunotherapy. Doxorubicin (DOX), ATP and ferrous ions (Fe2+) were co-assembled into nanosized amplifier (ADO-Fe) through π‒π stacking and coordination effect. Meanwhile, phenylboric acid-polyethylene glycol-phenylboric acid (PBA-PEG-PBA) was modified on the surface of ADO-Fe (denoted as PADO-Fe) by the virtue of d-ribose unit of ATP. PADO-Fe could display active targetability against tumor cells via sialic acid/PBA interaction. In acidic microenvironment, PBA-PEG-PBA would dissociate from amplifier. Moreover, high H2O2 concentration would induce hydroxyl radical (·OH) and oxygen (O2) generation through Fenton reaction by Fe2+. DOX and ATP would be released from the amplifier, which could induce ICD effect and "ICD adjuvant" to amplify this process. Together with programmed death ligands 1 (PD-L1) checkpoint blockade immunotherapy, PADO-Fe could not only activate immune response against primary tumor, but also strong abscopal effect against distant tumor. Our simple and multifunctional ICD amplifier opens a new window for enhancing ICD effect and immune checkpoint blockade therapy.

4.
Acta Biomater ; 138: 505-517, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34798317

RESUMO

The limited infiltration of specific T cells in an immunosuppressive microenvironment is a major challenge for cancer immunotherapy. Reversing tumor microenvironment and inducing an antitumor immune response are crucial for cancer therapy. Here, phenylboronic acid (PBA) derivative-stabilized ultrasmall platinum nanoparticles (PBA-Pt) and dextran-coated BLZ-945 nanoparticles (DNPs) were co-assembled through a pH-responsive borate ester bond to construct a versatile reversible shielding multifunctional nanoplatform (Pt@DNPs) for the first time. Pt@DNPs dissociated into two individual components, namely PBA-Pt and DNPs, in the tumor acid microenvironment. Both in vitro and in vivo studies revealed that Pt@DNPs induced immunogenic cell death (ICD) (through multimechanisms involving PtⅡ release and a multienzyme catalytic process by PBA-Pt) and relieved immunosuppressive microenvironment (depletion of tumor-associated macrophages by BLZ-945), which led to tumor-associated antigen release, maturation of dendritic cells, and infiltration of cytotoxic T cells for efficient antitumor immune response against both primary tumor and pulmonary metastatic tumor nodules. Therefore, Pt@DNPs is a promising option for cancer chemo-immunotherapy. STATEMENT OF SIGNIFICANCE: A versatile reversible shielding multifunctional nanoplatform (Pt@DNPs) was engineered for the first time for combinational cancer chemo-immunotherapy. Multimechanisms involving induction of immunogenic cell death by PBA-Pt and sufficient TAM depletion by DNPs could efficiently relieve tumor immunosuppressive microenvironment and activate the antitumor immune response. The synergistic effect not only increased the infiltration of specific T cells in primary tumor, but it also induced a strong immune response against pulmonary metastatic nodules. Collectively, this nanoplatform may represent a promising strategy for combinational chemo-immunotherapy for cancers.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Linhagem Celular Tumoral , Concentração de Íons de Hidrogênio , Imunoterapia , Platina , Microambiente Tumoral
5.
ACS Appl Mater Interfaces ; 13(33): 39934-39948, 2021 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-34396771

RESUMO

There are two severe obstacles in cancer immunotherapy. The first is that the low response rate challenges the immune response owing to the immunosuppressive tumor microenvironment (ITM) and poor immunogenicity of the tumor. The second obstacle is that the dense and intricate pathophysiology barrier seriously restricts deep drug delivery in solid tumors. A laser/glutathione (GSH)-activatable nanosystem with tumor penetration for achieving highly efficient immunotherapy is reported. The core of the nanosystem was synthesized by coordinating zinc ions with GSH-activatable oxaliplatin (OXA) prodrugs and carboxylated phthalocyanine. Such an OXA/phthalocyanine-based coordination polymer nanoparticle (OPCPN) was wrapped by a phospholipid bilayer and NTKPEG. NTKPEG is a PEGylated indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor prodrug containing a thioketal (TK) linker, which was modified on the OPCPN (OPCPN@NTKPEG). Upon the laser irradiation tumor site, ROS production of the OPCPN@NTKPEG triggers cleavage of NTKPEG by degradation of TK for promoted tumor penetration and uptake. OXA, phthalocyanine, and IDO1 inhibitor were released by the intracellular high-level GSH. OXA inhibits cell growth and is combined with photodynamic therapy (PDT) to induce immunogenic cell death (ICD). The IDO1 inhibitor reversed the ITM by suppressing IDO1-mediated Trp degradation and exhaustion of cytotoxic T cells. Laser/GSH-activatable drug delivery was more conducive to enhancing ICD and reversing ITM in deep tumors. Chemo-PDT with OPCPN@NTKPEG significantly regressed tumor growth and reduced metastasis by improved cancer immunotherapy.


Assuntos
Antineoplásicos/química , Portadores de Fármacos/química , Glutationa/química , Indóis/química , Nanopartículas/química , Oxaliplatina/química , Fármacos Fotossensibilizantes/química , Animais , Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Liberação Controlada de Fármacos , Glutationa/metabolismo , Humanos , Morte Celular Imunogênica/efeitos dos fármacos , Morte Celular Imunogênica/efeitos da radiação , Imunoterapia , Indóis/farmacocinética , Isoindóis , Lasers , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Oxaliplatina/farmacocinética , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacocinética , Polietilenoglicóis/química , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Distribuição Tecidual , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos da radiação
6.
ACS Biomater Sci Eng ; 7(7): 3141-3155, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34137580

RESUMO

In cancer therapy, it is acknowledged that large-size nanoparticles stay in the circulation system for a long time, but their permeability to tumor tissues is poor. To address the conflicting need for prolonging circulation time and favorable tumor tissue penetration ability, a charge conversional multifunctional nanoplatform was strategically designed to improve the efficacy of small interfering RNA (siRNA) therapy against nonsmall cell lung cancer (NSCLC). The development of nanodrug delivery systems (NDDSs) was constructed by loading siRNA on polyamidoamine (PAMAM) dendrimers to build small-sized PAM/siRNA via electrostatic interaction and then capped with a pH-triggered copolymer poly(ethylene glycol) methyl ether (mPEG)-poly-l-lysine (PLL)-2,3-dimethylmaleic anhydride (DMA) (shorted as PLM) under physiological conditions. While in the tumor microenvironment, the acidic reaction of the PLM copolymer changes from negative charge to positive charge due to the cleavable amide bond between mPEG-PLL and DMA, leading to large-size nanoparticles (NPs) with a negative charge that turns into a positive charge and small NPs with a high tumor-penetrating ability. All of the in vitro and in vivo studies validated that PLM/PAM/siRNA NPs possess desirable features including excellent biocompatibility, a prolonged circulation time, significant pH sensitivity, high tumor tissue penetration ability, and sufficient endo-/lysosomal escape. Taken together, all results suggest tremendous potential of the gene therapy based on the stimuli-sensitive PLM/PAM/siRNA NPs, providing a profound application prospective treatment strategy in cancer gene therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Nanopartículas , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Estudos Prospectivos , RNA Interferente Pequeno/genética , Microambiente Tumoral
7.
J Mater Chem B ; 8(8): 1616-1628, 2020 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-32010914

RESUMO

Small interfering RNA (siRNA) has extensive potential for the treatment of non-small cell lung cancer (NSCLC). While both cationic lipids and polymers have demonstrated promise to facilitate siRNA encapsulation, they can also hamper cytosolic siRNA release and induce severe cytotoxicity. To address these issues, a unique polymer hybrid nanoparticle (NP) nanoplatform was developed for multistage siRNA delivery based on both pH-responsive and endo/lysosomal escape characteristics, which was formed via a combination of an electrostatic interactions between the copolymer methoxy poly(ethylene glycol)-poly(l-histidine)-poly(sulfadimethoxine) (mPEG-PHis-PSD, shortened to PHD), dendritic poly-l-lysine (PLL) and PLK1 siRNA (shortened to siPLK1). The biological composition of the proton sponge effect polymer of the PHis chain, which was in position to make efficient endo/lysosomal escape, and the pH-responsive polymer of the PSD fragment, which could accelerate the release of siPLK1. In the present study, the NP illustrated excellent physiochemical properties and rapid endo/lysosomal escape in vitro. Besides this, compared with the PD/PLL/siRNA formulation, the PHD/PLL/siRNA NP indicated higher cellular uptake, and higher cell cytotoxicity in vitro. The in vivo results demonstrated that the PHD/PLL/siRNA NP exhibited the strongest tumor growth inhibition rate and ideal safety compared with the control and other siPLK1-treated formulations, which can be mainly attributed to pH-induced instantaneous dissociation and efficient endo/lysosomal escape arising from the PHD copolymer. Consequently, the above evidence indicates that the PHD/PLL/siRNA NP is a favorable gene delivery system and provides a potential strategy for siRNA delivery.


Assuntos
Antineoplásicos/química , Histidina/química , Nanopartículas/química , RNA Interferente Pequeno/metabolismo , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Endossomos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Tamanho da Partícula , Polietilenoglicóis/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/química , Distribuição Tecidual , Quinase 1 Polo-Like
8.
Biomater Sci ; 7(9): 3751-3763, 2019 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-31355389

RESUMO

Poor prognosis in lung cancer has been proved to be associated with the presence of lung cancer stem cells (LCSCs). Similar to bulk cancer cells and always existing in the interior of a solid tumor, there are also insurmountable barriers for the elimination of CSCs. To overcome these drawbacks, a versatile polyion complex was rationally designed, which can respond to a tumor microenvironment and exhibits a size-variable property, which allows it to possess remarkable tumor penetration capability and to accumulate around LCSCs. Protein tyrosine kinase 7 (PTK7) antibody mediated active targetability can facilitate the cellular uptake of triphenylphosphine-docetaxel (TD) and microRNA-31(miR-31) and the breakage of the disulfide bond can also enhance intracellular drug release. TD possesses a favorable apoptosis-inducing effect via a mitochondria pathway, while miR-31 could significantly regulate the MET-PI3K-Akt signaling pathway to exert the capability to eliminate LCSCs. APSP/HA might support new insights into LCSC eradiation in cancer management.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Polímeros/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Células A549 , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Liberação Controlada de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Polímeros/química , Relação Estrutura-Atividade , Células Tumorais Cultivadas
9.
ACS Appl Mater Interfaces ; 11(12): 11865-11875, 2019 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-30830746

RESUMO

It is generally known that, for nanoparticles in cancer therapy, sufficient tumor penetration needs a minor particle size, while long in vivo circulation time needs a larger particle size. It is hard to balance them because they are standing on either side of a seesaw. To address these two different requirements, a dual-responsive size-shrinking nanocluster can self-adaptively respond to a complicated tumor microenvironment and transform its particulate property to overcome sequential in vivo barriers and reach a preferable antitumor activity. The nanocluster (RPSPT@SNCs) could preferentially accumulate into tumor tissue and dissociate under extracellular matrix metalloproteinase-2 (MMP-2) to release small-sized micelle formulations (RPSPTs). RPSPT possesses favorable tumor penetration and tumor targeting capability to deliver the antitumor agent paclitaxel (PTX) into deep regions of solid tumor. The intracellular redox microenvironment can also accelerate drug accumulation. The prepared RPSPT@SNCs possesses enhanced cell cytotoxicity and tumor penetration capability on MCF-7 cells and a favorable antitumor activity on the xenograft tumor mouse model.


Assuntos
Nanopartículas/química , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Micelas , Microscopia Confocal , Nanopartículas/uso terapêutico , Nanopartículas/toxicidade , Neoplasias/tratamento farmacológico , Neoplasias/mortalidade , Neoplasias/patologia , Paclitaxel/química , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Tamanho da Partícula , Polímeros/química , Taxa de Sobrevida , Distribuição Tecidual , Transplante Heterólogo
10.
Drug Deliv ; 25(1): 723-737, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29513049

RESUMO

Mitochondrial-targeting therapy was considered to be a promising approach for the efficient treatment of cancer while positive charge induced nonspecific cytotoxicity severely limits its application. To overcome this drawback, a novel mitochondria targeted conjugate triphenylphosphine-docetaxel (TD) has been synthesized successfully and incorporated it into liposomes (EPSLP/TD), which possessed excellent pH-sensitive characteristic, EphA 10 mediated active targetability as well as mitochondria-targeting capability. EPSLP/TD was characterized to have a small particle size, high-encapsulation efficiency and excellent pH-sensitive characteristic. Compared with DTX-loaded liposomes (EPSLP/DTX), EPSLP/TD possessed higher cytotoxicity against MCF-7 cell line. Mitochondrial-targeting assay demonstrated mitochondria-targeting moiety triphenylphosphine (TPP) could efficiently deliver DTX to mitochondria. Western immunoblotting assay indicated that EPSLP/TD could efficiently deliver antitumor drug to mitochondria and induce cell apoptosis via mitochondria-mediated apoptosis pathway. In vivo antitumor study demonstrated EPSLP/TD owed excellent in vivo antitumor activity. Histological assay demonstrated EPSLP/TD showed strongly apoptosis inducing effect, anti-proliferation effect and anti-angiogenesis effect. This work investigated the potential of hierarchical targeting pH-sensitive liposomes is a suitable carrier to activate mitochondria-mediated apoptosis pathway for cancer therapy.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Compostos Organofosforados/administração & dosagem , Receptores da Família Eph/administração & dosagem , Taxoides/administração & dosagem , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Docetaxel , Feminino , Humanos , Concentração de Íons de Hidrogênio , Lipossomos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Int J Nanomedicine ; 13: 4209-4226, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30140154

RESUMO

INTRODUCTION: Multidrug resistance (MDR) of breast cancer is the major challenge to successful chemotherapy while mitochondria-targeting therapy was a promising strategy to overcome MDR. MATERIALS AND METHODS: In this study, HER-2 peptide-PEG2000-Schiff base-cholesterol (HPSC) derivate was synthesized successfully and incorporated it on the surface of the doxorubicin (DOX)-loaded dequalinium (DQA) chloride vesicle (HPS-DQAsomes) to treat drug-resistant breast cancer. Evaluations were performed using human breast cancer cell and DOX-resistant breast cancer cell lines (MCF-7 and MCF-7/ADR). RESULTS: The particle size of HPS-DQAsomes was ~110 nm with spherical shape. In vitro cytotoxicity assay indicated that HPS-DQAsomes could increase the cytotoxicity against MCF-7/ADR cell line. Cellular uptake and mitochondria-targeting assay demonstrated that HPS-DQAsomes could target delivering therapeutical agent to mitochondria and inducing mitochondria-driven apoptosis process. In vivo antitumor assay suggested that HPS-DQAsomes could reach favorable antitumor activity due to both tumor targetability and sub-organelles' targetability. Histological assay also indicated that HPS-DQAsomes showed a strong apoptosis-inducing effect. No obvious systematic toxicity of HPS-DQAsomes could be observed. CONCLUSION: In summary, multifunctional HPS-DQAsomes provide a novel and versatile approach for overcoming MDR via mitochondrial pathway in cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Dequalínio/farmacologia , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Mitocôndrias/metabolismo , Peptídeos/farmacologia , Receptor ErbB-2/metabolismo , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Colesterol/química , Citocromos c/metabolismo , Dequalínio/uso terapêutico , Doxorrubicina/uso terapêutico , Liberação Controlada de Fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Lipossomos , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Tamanho da Partícula , Polietilenoglicóis/química , Bases de Schiff/química , Testes de Toxicidade
12.
ACS Appl Mater Interfaces ; 10(25): 21590-21600, 2018 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-29798663

RESUMO

The therapeutic efficacy of chemotherapy is dramatically hindered by multidrug resistance (MDR), which is induced by the overexpression of P-glycoprotein (P-gp). The codelivery of an antitumor drug and siRNA is an effective strategy recently applied in overcoming P-gp-related MDR. In this study, a multifunctional drug delivery system with both pH-sensitive feature and active targetability was designed, in which MDR1-siRNA and DOX were successfully loaded. The resulting carrier EphA10 antibody-conjugated pH-sensitive doxorubicin (DOX), MDR1-siRNA coloading lipoplexes (shortened as DOX + siRNA/ePL) with high serum stability had favorable physicochemical properties. DOX + siRNA/ePL exhibited an incremental cellular uptake, enhanced P-gp downregulation efficacy, as well as a better cell cytotoxicity in human breast cancer cell line/adriamycin drug-resistant (MCF-7/ADR) cells. The results of the intracellular colocalization study indicated that DOX + siRNA/ePL possessed the ability for pH-responsive rapid endosomal escape in a time-dependent characteristic. Meanwhile, the in vivo antitumor activities suggested that DOX + siRNA/ePL could prolong the circulation time as well as specifically accumulate in the tumor cells via receptor-mediated endocytosis after intravenous administration into the blood system. The histological study further demonstrated that DOX + siRNA/ePL could inhibit the proliferation, induce apoptosis effect, and downregulate the P-gp expression in vivo. Altogether, DOX + siRNA/ePL was expected to be a suitable codelivery system for overcoming the MDR effect.


Assuntos
Resistência a Múltiplos Medicamentos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Linhagem Celular Tumoral , Doxorrubicina , Resistencia a Medicamentos Antineoplásicos , Humanos , Concentração de Íons de Hidrogênio , Nanopartículas , RNA Interferente Pequeno , Receptores da Família Eph
13.
Int J Nanomedicine ; 13: 6885-6902, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30498349

RESUMO

INTRODUCTION: Specific polo-like kinase (PLK1) silencing with small interface RNA (siRNA) may be an effective approach for PLK1-overexpressed lung cancer. However, low siRNA concentration into cytoplasm of tumor tissue severely limits its application. MATERIALS AND METHODS: In this study, a novel triblock copolymer methoxy poly(ethylene glycol)-poly(histidine)-poly(sulfadimethoxine) (mPEG-PHis-PSD, shorten as PHD) was synthesized and used to construct novel nonviral gene vector with cationic liposomes. RESULTS: The resulting hybrid nanoparticles (PHD/LR) loaded with siPLK1 possessed excellent physiochemical properties. In vitro study indicated that PHD/LR could be efficiently internalized into human lung adenocarcinoma A549 cells and downregulated PLK1 protein expression to induce cell apoptosis, which was attributed to pH-induced instantaneous dissociation, efficient endo/lysosomal escape arose from PHD copolymer. Furthermore, in vivo antitumor activity demonstrated that PHD/LR could efficiently accumulated into tumor tissue and silenced PLK1 expression to possess antitumor activity. CONCLUSION: Taken all these together, PHD/LR was expected to be a suitable carrier for specific delivering siRNA for lung cancer therapy.


Assuntos
Adenocarcinoma/terapia , Proteínas de Ciclo Celular/antagonistas & inibidores , Terapia Genética , Neoplasias Pulmonares/terapia , Nanopartículas/administração & dosagem , Polímeros/administração & dosagem , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , RNA Interferente Pequeno/administração & dosagem , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Apoptose , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proliferação de Células , Humanos , Concentração de Íons de Hidrogênio , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , Nanopartículas/química , Polímeros/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Quinase 1 Polo-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA