Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Nano Lett ; 23(21): 9963-9971, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37729438

RESUMO

Given the key roles of cancer associated fibroblasts (CAFs) in shaping tumor stroma, this study shows a CAF-associated ITGB1-inactivating peptide-enriched membrane nanodelivery system (designated as PMNPs-D) to simultaneously target CAFs and tumor cells for boosted chemotherapy through promoted drug perfusion. In the structure of PMNPs-D, the PLGA-based inner core is loaded with the chemotherapeutic drug doxorubicin, and the outer surface is cloaked by hybrid biomembranes with the insertion of integrin ß1 (ITGB1) inhibiting peptide (i.e., FNIII14). After prolonged blood circulation and actively targeting in tumor sites, PMNPs-D can respond to CAF-overexpressed fibroblast activation protein-α (FAP-α) to trigger the release of FNIII14, which will bind to ITGB1 and inhibit CAFs' biological function in producing the stromal matrix, thereby loosening the condensed stromal structure and enhancing the permeability of nanotherapeutics in tumors. As a result, this tailor-designed nanosystem shows substantial tumor inhibition and metastasis retardation in aggressive adenoid cystic carcinoma (ACC) tumor-harboring mice.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias , Animais , Camundongos , Fibroblastos Associados a Câncer/patologia , Neoplasias/patologia , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Membranas , Peptídeos/metabolismo , Microambiente Tumoral , Linhagem Celular Tumoral , Fibroblastos/metabolismo
2.
Nano Lett ; 22(21): 8735-8743, 2022 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-36286590

RESUMO

The chemotherapy efficacy of nanodrugs is restricted by poor tumor targeting and uptake. Here, an engineered biohybrid living material (designated as EcN@HPB) is constructed by integrating paclitaxel and BAY-876 bound human serum albumin nanodrugs (HPB) with Escherichia coli Nissle 1917 (EcN). Due to the inherent tumor tropism of EcN, EcN@HPB could actively target the tumor site and competitively deprive glucose through bacterial respiration. Thus, albumin would be used as an alternative nutrient source for tumor metabolism, which significantly promotes the internalization of HPB by tumor cells. Subsequently, BAY-876 internalized along with HPB nanodrugs would further depress glucose uptake of tumor cells via inhibiting glucose transporter 1 (GLUT1). Together, the decline of glucose bioavailability of tumor cells would activate and promote the macropinocytosis in an AMP-activated protein kinase (AMPK)-dependent manner, resulting in more uptake of HPB by tumor cells and boosting the therapeutic outcome of paclitaxel.


Assuntos
Infecções por Escherichia coli , Nanopartículas , Neoplasias , Humanos , Disponibilidade Biológica , Escherichia coli/genética , Escherichia coli/metabolismo , Glucose/metabolismo , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico
3.
Chem Soc Rev ; 50(2): 945-985, 2021 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-33226037

RESUMO

Cell primitive-based functional materials that combine the advantages of natural substances and nanotechnology have emerged as attractive therapeutic agents for cancer therapy. Cell primitives are characterized by distinctive biological functions, such as long-term circulation, tumor specific targeting, immune modulation etc. Moreover, synthetic nanomaterials featuring unique physical/chemical properties have been widely used as effective drug delivery vehicles or anticancer agents to treat cancer. The combination of these two kinds of materials will catalyze the generation of innovative biomaterials with multiple functions, high biocompatibility and negligible immunogenicity for precise cancer therapy. In this review, we summarize the most recent advances in the development of cell primitive-based functional materials for cancer therapy. Different cell primitives, including bacteria, phages, cells, cell membranes, and other bioactive substances are introduced with their unique bioactive functions, and strategies in combining with synthetic materials, especially nanoparticulate systems, for the construction of function-enhanced biomaterials are also summarized. Furthermore, foreseeable challenges and future perspectives are also included for the future research direction in this field.


Assuntos
Antineoplásicos/farmacologia , Materiais Biomiméticos/farmacologia , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Materiais Biomiméticos/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Neoplasias/patologia
4.
J Am Chem Soc ; 142(51): 21460-21468, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33290051

RESUMO

Inspired by nature, where dynamic networks control the levels of gene expression and the activities of transcribed/translated proteins, we introduce nucleic acid-based constitutional dynamic networks (CDNs) as functional modules mimicking native circuits by demonstrating CDNs-guided programmed synthesis of genes, controlled transcription of RNAs, and dictated transcription/translation synthesis of proteins. An auxiliary CDN consisting of four dynamically equilibrated constituents AA', AB', BA', and BB' is orthogonally triggered by two different inputs yielding two different compositionally reconfigured CDNs. Subjecting the parent auxiliary CDN to two hairpins, HA and HB, and two templates TA and TB and a nicking/replication machinery leads to the cleavage of the hairpins and to the activation of the nicking/replication machineries that synthesize two "genes", e.g., the histidine-dependent DNAzyme g1 and the Zn2+-ion-dependent DNAzyme g2. The triggered orthogonal reconfiguration of the parent CDN to the respective CDNs leads to the programmed preferred CDN-guided synthesis of g1 or g2. Similarly, the triggered reconfigured CDNs are subjected to two hairpins HC and HD, the templates I'/I and J'/J, and the RNA polymerase (RNAp)/NTPs machinery. While the cleavage of the hairpins by the constituents associated with the parent CDN leads to the transcription of the broccoli aptamer recognizing the DFHBI ligand and of the aptamer recognizing the malachite green (MG) ligand, the orthogonally triggered CDNs lead to the CDNs-guided enhanced transcription of either the DFHBI aptamer or the MG aptamer. In addition, subjecting the triggered reconfigured CDNs to predesigned hairpins HE and HF, the templates M'/M and N'/N, the RNAp/NTPs machinery, and the cell-free ribosome t-RNA machinery leads to the CDNs-guided transcription/translation of the green fluorescence protein (GFP) or red fluorescence protein (RFP).


Assuntos
Redes Reguladoras de Genes , Biossíntese de Proteínas/genética , Animais , Aptâmeros de Nucleotídeos/genética , Proteínas de Fluorescência Verde/genética , RNA Mensageiro/genética
5.
Nano Lett ; 19(12): 9121-9130, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31729224

RESUMO

The broadband C3N4 semiconductor absorbs in the UV region, λ = 330-380 nm, a feature limiting its application for light-to-energy conversion. The unique surface adsorption properties of C3N4 allow, however, the binding of a photosensitizer, operating in the visible-solar spectrum to the surface of C3N4. Coupling of the energy levels of the photosensitizer with the energy levels of C3N4 allows effective photoinduced electron-transfer quenching and subsequent charge separation in the hybrid structures. Two methods to adsorb a photosensitizer on the C3N4 nanoparticles are described. One is exemplified by the adsorption of Zn(II)-protoporphyrin IX on C3N4 using π-π interactions. The second method utilizes the specific binding interactions of single-stranded nucleic acids on C3N4 and involves the binding of a Ru(II)-tris-bipyridine-modified nucleic acid on the C3N4 nanoparticles. Effective electron-transfer quenching of the photoexcited photosensitizers by C3N4 proceeds in the two hybrid systems. The two hybrid photosystems induce the effective photosensitized reduction of N,N'-dimethyl-4,4'-bipyridinium, MV2+, to MV+•, in the presence of Na2EDTA as a sacrificial electron donor. The generation of MV+• is ca. 5-fold higher as compared to the formation of MV+• in the presence of the photosensitizer alone (in the absence of C3N4). The effective generation of MV+• in the photosystems is attributed to the efficient quenching of the photosensitizers, followed by effective charge separation of the electrons in the conduction band of C3N4 and the holes in the oxidized photosensitizer. The subsequent transfer of the conduction-band electrons to MV2+ and the oxidation of Na2EDTA by the oxidized photosensitizers lead to the effective formation of MV+•. The photogenerated MV+• by the two hybrid photosystems is used to catalyze H2 evolution in the presence of Pt nanoparticle catalysts and to mediate the reduction of NADP+ to NADPH, in the presence of ferredoxin-NADP+ reductase, FNR. The ability to couple the photogenerated NADPH to drive NADP+-dependent biocatalytic transformations is demonstrated.

6.
Nano Lett ; 19(9): 6621-6628, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31407917

RESUMO

Sequence-specific aptamers act as functional scaffolds for the assembly of photosynthetic model systems. The Ru(II)-tris-bipyridine photosensitizer is conjugated by different binding modes to the antityrosinamide aptamer to yield a set of photosensitizer-aptamer binding scaffolds. The N-methyl-N'-(3-aminopropane)-4,4'-bipyridinium electron acceptor, MV2+, is covalently linked to tyrosinamide, TA, to yield the conjugate TA-MV2+. The tyrosinamide unit in TA-MV2+ acts as a ligand for anchoring TA-MV2+ to the Ru(II)-tris-bipyridine-aptamer scaffold, generating the diversity of photosensitizer-aptamer/electron acceptor supramolecular conjugates. Effective electron transfer quenching in the photosynthetic model systems is demonstrated, and the quenching efficiencies are controlled by the structural features of the conjugates. The redox species generated by the photosensitizer-aptamer/electron acceptor supramolecular systems mediate the ferredoxin-NADP+ reductase, FNR, catalyzed synthesis of NADPH, and the Pt-nanoparticle-catalyzed evolution of hydrogen (H2). The novelty of the study rests on the unprecedented use of aptamer scaffolds as functional units for organizing photosynthetic model systems.


Assuntos
Aptâmeros de Nucleotídeos/química , Nanopartículas Metálicas/química , Modelos Químicos , Fármacos Fotossensibilizantes/química , Fotossíntese , Platina/química , Transporte de Elétrons , Ferredoxina-NADP Redutase/química , NADP/química
7.
Angew Chem Int Ed Engl ; 59(23): 9163-9170, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32125762

RESUMO

All-DNA scaffolds act as templates for the organization of photosystem I model systems. A series of DNA templates composed of ZnII -protoporphyrin IX (ZnII PPIX)-functionalized G-quadruplex conjugated to the 3'- or 5'-end of the tyrosinamide (TA) aptamer and ZnII PPIX/G-quadruplex linked to the 3'- and 5'-ends of the TA aptamer through a four-thymidine bridge. Effective photoinduced electron transfer (ET) from ZnII PPIX/G-quadruplex to bipyridinium-functionalized tyrosinamide, TA-MV2+ , bound to the TA aptamer units is demonstrated. The effectiveness of the primary ET quenching of ZnII PPIX/G-quadruplex by TA-MV2+ controls the efficiency of the generation of TA-MV+. . The photosystem-controlled formation of TA-MV+. by the different photosystems dictates the secondary activation of the ET cascade corresponding to the ferredoxin-NADP+ reductase (FNR)-catalysed reduction of NADP+ to NADPH by TA-MV+. , and the sequestered alcohol dehydrogenase catalysed reduction of acetophenone to 1-phenylethanol by NADPH.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , DNA/química , DNA/metabolismo , Quadruplex G , Modelos Biológicos , Fotossíntese , Protoporfirinas/metabolismo , Transporte de Elétrons
8.
J Am Chem Soc ; 141(39): 15567-15576, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31478647

RESUMO

In this study, we report on a redox-controllable and reversible complete "ON"/"OFF"-switchable aptamer binding to ATP. A series of methylene blue-modified ATP-aptamers was synthesized, revealing improved binding affinities toward ATP as compared to the nonmodified aptamer. These binding affinities were dependent on the conjugation site of the redox label on the aptamer scaffold. Importantly, we find that the oxidized methylene blue-modified aptamers bind to ATP with micromolar affinity, while the reduced form lacks binding affinity toward ATP, resulting in an unprecedented complete "ON"/"OFF" redox-controllable aptamer switch. We demonstrate the cyclic "ON"/"OFF" binding of ATP to the methylene blue-functionalized aptamer through cyclic oxidation and reduction of the redox label using both chemical and electrochemical means. Molecular dynamics and docking simulations were performed to account for the redox-switchable properties of the conjugated aptamers and to rationalize the enhanced binding affinities of the different aptamer designs.


Assuntos
Trifosfato de Adenosina/química , Aptâmeros de Nucleotídeos/química , Técnicas Biossensoriais/métodos , Fenômenos Químicos , Técnicas Eletroquímicas , Modelos Moleculares , Conformação de Ácido Nucleico , Oxirredução
9.
Small ; 15(17): e1900935, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30920730

RESUMO

UiO-68 metal-organic framework nanoparticles (NMOFs) are loaded with a doxorubicin drug (fluorescent dye analogs) and locked by means of structurally engineered duplex nucleic acid structures, where one strand is covalently linked to the NMOFs and the second strand is hybridized with the anchor strand. Besides the complementarity of the second strand to the anchor sequence, it includes the complementary sequence to the microRNAs (miRNA)-21 or miRNA-221 that is specific miRNA biomarker for MCF-7 breast cancer cells or OVCAR-3 ovarian cancer cells. In the presence of the respective miRNA biomarkers, the miRNA-induced displacement of the strand associated with the anchor strand proceeds, resulting in the release of DNA/miRNA duplexes. The released duplexes are, however, engineered to be digested in the presence of exonuclease III, Exo III, a process that recycles the miRNAs and provides the autonomous amplified unlocking of the NMOFs and the release of the doxorubicin load (or the fluorescent dye analogs) even at low concentrations of miRNA. Preliminary cell experiments reveal that the respective NMOFs are unlocked by the miRNA-21 or miRNA-221, resulting in selective cytotoxicity toward MCF-7 breast cancer cells or OVCAR-3 ovarian cancer cells.


Assuntos
Sistemas de Liberação de Medicamentos , Estruturas Metalorgânicas , MicroRNAs/química , Nanomedicina/métodos , Neoplasias/tratamento farmacológico , Apoptose , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular , Doxorrubicina/química , Feminino , Humanos , Ligantes , Células MCF-7 , MicroRNAs/metabolismo , Nanopartículas/química , Compostos Orgânicos , Neoplasias Ovarianas/tratamento farmacológico
10.
Small ; 13(18)2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28266809

RESUMO

The nanoplatform GNR-ACPP-PpIX (designated as GNR-ACPI) is designed for dual image guided combined activatable photodynamic therapy (PDT) and photothermal therapy (PTT). In GNR-ACPI, gold nanorods (GNRs) are modified with a protoporphyrin (PpIX, a PDT agent) conjugated activatable cell penetrating peptide (ACPP), which consists of the matrix metalloproteinases-2 (MMP-2) sensitive peptide sequence GPLGLAG. First, the photoactivity of PpIX is effectively quenched by GNRs due to the strong near infrared region light absorption of GNR and the special "U type" structure of ACPP induced close contact between PpIX and GNR. However, once arriving at the tumor site, the GPLGLAG sequence is hydrolyzed by the MMP-2 overexpressed by tumor cells, resulting in the release of the residual cell membrane penetrating peptide (CPP) attached PpIX (CPP-PpIX) with the recovery of photoactivity of PpIX. In addition, with the help of CPP, more efficient cellular uptake of PpIX by tumor cells can be achieved, which will greatly improve the PDT efficacy. Moreover, the GNR can also be utilized for photothermic imaging as well as PTT for tumors. It is found that the combination of PTT and PDT under the guidance of dual-mode imaging greatly enhances the antitumor effects, while possessing negligible systematic toxicity.


Assuntos
Ouro/química , Nanotubos/química , Fotoquimioterapia/métodos , Peptídeos Penetradores de Células/química
11.
Small ; 12(6): 733-44, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26708101

RESUMO

In this work, a ZnO based nanococktail with programmed functions is designed and synthesized for self-synergistic tumor targeting therapy. The nanococktail can actively target tumors via specific interaction of hyaluronic acid (HA) with CD44 receptors and respond to HAase-rich tumor microenvironment to induce intracellular cascade reaction for controlled therapy. The exposed cell-penetrating peptide (R8) potentiates the cellular uptake of therapeutic nanoparticles into targeted tumor cells. Then ZnO cocktail will readily degrade in acidic endo/lysosomes and induce the production of desired reactive oxygen species (ROS) in situ. The destructive ROS not only leads to serious cell damage but also triggers the on-demand drug release for precise chemotherapy, thus achieving enhanced antitumor efficiency synergistically. After tail vein injection of ZnO cocktail, a favorable tumor apoptosis rate (71.2 ± 8.2%) is detected, which is significantly superior to that of free drug, doxorubicin (12.9 ± 5.2%). Both in vitro and in vivo studies demonstrate that the tailor-made ZnO cocktail with favorable biocompatibility, promising tumor specificity, and self-synergistically therapeutic capacity opens new avenues for cancer therapy.


Assuntos
Espaço Intracelular/metabolismo , Nanopartículas/química , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Citometria de Fluxo , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Espécies Reativas de Oxigênio/metabolismo
12.
Small ; 11(39): 5230-42, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26285687

RESUMO

Nanotechnology-based drug delivery has a great potential to revolutionize cancer treatment by enhancing anticancer drug efficacy and reducing drug toxicity. Here, a bioinspired nano-prodrug (BiNp) assembled by an antineoplastic peptidic derivative (FA-KLA-Hy-DOX), a folate acid (FA)-incorporated proapoptotic peptide (KLAKLAK)(2) (KLA) to doxorubicin (DOX) via an acid-labile hydrozone bond (Hy) is constructed. The hydrophobic antineoplastic agent DOX is efficiently shielded in the core of nano-prodrug. With FA targeting moieties on the surface, the obtained BiNp shows significant tumor-targeting ability and enhances the specific uptake of cancer cells. Upon the trigger by the intracellular acidic microenvironment of endosomes, the antineoplastic agent DOX is released on-demand and promotes the apoptosis of cancer cells. Simultaneously, the liberated FA-KLA can induce the dysfunction of mitochondria and evoke mitochondria-dependent apoptosis. In vitro and in vivo results show that the nano-prodrug BiNp with integrated programmed functions exhibits remarkable inhibition of tumor and achieves a maximized therapeutic efficiency with a minimized side effect.


Assuntos
Doxorrubicina/administração & dosagem , Ácido Fólico/farmacocinética , Nanocápsulas/administração & dosagem , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Pró-Fármacos/administração & dosagem , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/química , Apoptose/efeitos dos fármacos , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/síntese química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Sinergismo Farmacológico , Feminino , Ácido Fólico/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanocápsulas/química , Nanocápsulas/ultraestrutura , Neoplasias Experimentais/química , Neoplasias Experimentais/patologia , Pró-Fármacos/síntese química
13.
Small ; 10(3): 599-608, 2014 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-24000121

RESUMO

Graphene oxide (GO)-based theranostic nanohybrid is designed for tumor induced imaging and potential combinational tumor therapy. The anti-tumor drug, Doxorubicin (DOX) is chemically conjugated to the poly(ethylenimine)-co-poly(ethylene glycol) (PEI-PEG) grafted GO via a MMP2-cleavable PLGLAG peptide linkage. The therapeutic efficacy of DOX is chemically locked and its intrinsic fluorescence is quenched by GO under normal physiological condition. Once stimulated by the MMP2 enzyme over-expressed in tumor tissues, the resulting peptide cleavage permits the unloading of DOX for tumor therapy and concurrent fluorescence recovery of DOX for in situ tumor cell imaging. Attractively, this PEI-bearing nanohybrid can mediate efficient DNA transfection and shows great potential for combinational drug/gene therapy. This tumor induced imaging and potential combinational therapy will open a window for tumor treatment by offering a unique theranostic approach through merging the diagnostic capability and pathology-responsive therapeutic function.


Assuntos
Diagnóstico por Imagem/métodos , Grafite , Nanopartículas , Neoplasias/diagnóstico , Neoplasias/terapia , Óxidos , Animais , Linhagem Celular , Sobrevivência Celular , Terapia Combinada , Humanos , Luciferases/metabolismo , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Polietilenoimina/análogos & derivados , Polietilenoimina/síntese química , Polietilenoimina/química , Espectrometria de Fluorescência , Espectroscopia de Infravermelho com Transformada de Fourier , Termogravimetria
14.
J Am Chem Soc ; 135(13): 5068-73, 2013 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-23464924

RESUMO

A novel type of cellular-uptake-shielding multifunctional envelope-type mesoporous silica nanoparticle (MEMSN) was designed for tumor-triggered targeting drug delivery to cancerous cells. ß-Cyclodextrin (ß-CD) was anchored on the surface of mesoporous silica nanoparticles via disulfide linking for glutathione-induced intracellular drug release. Then a peptide sequence containing Arg-Gly-Asp (RGD) motif and matrix metalloproteinase (MMP) substrate peptide Pro-Leu-Gly-Val-Arg (PLGVR) was introduced onto the surface of the nanoparticles via host-guest interaction. To protect the targeting ligand and prevent the nanoparticles from being uptaken by normal cells, the nanoparticles were further decorated with poly(aspartic acid) (PASP) to obtain MEMSN. In vitro study demonstrated that MEMSN was shielded against normal cells. After reaching the tumor cells, the targeting property could be switched on by removing the PASP protection layer via hydrolyzation of PLGVR at the MMP-rich tumor cells, which enabled the easy uptake of drug-loaded nanoparticles by tumor cells and subsequent glutathione-induced drug release intracellularly.


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Dióxido de Silício/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Portadores de Fármacos/síntese química , Humanos , Microscopia Eletrônica de Transmissão , Porosidade
15.
Langmuir ; 29(23): 6996-7004, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23663135

RESUMO

Controlling the formation of large and homogeneous arrays of bionanostructures through the self-assembly approach is still a great challenge. Here, we report the spontaneous formation of highly ordered arrays based on aligned peptide nanostructures in a solution as well as at an interface by self-assembly. By controlling the time and temperature of self-assembly in the solution, parallel fibrous alignments and more sophisticated two-dimensional "knitted" fibrous arrays could be formed from aligned rod-like fibers. During the formation of such arrays, the "disorder-to-order" transitions are controlled by the temperature-responsible motile short hydrophobic tails of the gemini-like amphiphilic peptides (GAPs) with asymmetric molecular conformation. In addition, the resulting long-range-ordered "knitted" fibrous arrays are able to direct mineralization of calcium phosphate to form organic-inorganic composite materials. In this study, the self-assembly behavior of these peptide building blocks at an interface was also studied. Highly ordered spatial arrays with vertically or horizontally aligned nanostructures such as nanofibers, microfibers, and microtubes could be formed through interfacial assembly. The regular structures and their alignments on the interface are controlled by the alkyl chain length of building blocks and the hydrophilicity/hydrophobicity property of the interface.


Assuntos
Nanoestruturas/química , Peptídeos/química , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformação Molecular , Tamanho da Partícula , Soluções , Propriedades de Superfície
16.
ACS Nano ; 17(17): 17217-17232, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37584451

RESUMO

Macrophage-mediated cellular phagocytosis (MMCP) plays a critical role in conducting antitumor immunotherapy but is usually impaired by the intrinsic phagocytosis evading ability of tumor cells and the immunosuppressive tumor microenvironment (TME). Herein, a MMCP-boosting hydrogel (TCCaGM) was elaborately engineered by encapsulating granulocyte-macrophage colony-stimulating factor (GM-CSF) and a therapeutic nanoplatform (TCCaN) that preloaded with the tunicamycin (Tuni) and catalase (CAT) with the assistance of CaCO3 nanoparticles (NPs). Strikingly, the hypoxic/acidic TME was efficiently alleviated by the engineered hydrogel, "eat me" signal calreticulin (CRT) was upregulated, while the "don't eat me" signal CD47 was downregulated on tumor cells, and the infiltrated DCs were recruited and activated, all of which contributed to boosting the macrophage-mediated phagocytosis and initiating tumor-specific CD8+ T cells responses. Meanwhile, the remodeled TME was beneficial to accelerate the polarization of tumor-associated macrophages (TAMs) to the antitumoral M1-like phenotype, further heightening tumoricidal immunity. With the combination of PD-1 antibody (αPD-1), the designed hydrogel significantly heightened systemic antitumor immune responses and long-term immunological effects to control the development of primary and distant tumors as well as suppress tumor metastasis and recurrence, which established an optimal strategy for high-performance antitumor immunotherapy.


Assuntos
Adjuvantes Imunológicos , Neoplasias , Humanos , Adjuvantes Imunológicos/farmacologia , Microambiente Tumoral , Linfócitos T CD8-Positivos , Hidrogéis/farmacologia , Macrófagos , Neoplasias/terapia , Neoplasias/patologia , Fagocitose , Antígeno CD47 , Imunoterapia
17.
Adv Mater ; 33(31): e2007630, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34050564

RESUMO

Immunotherapy that can activate immunity or enhance the immunogenicity of tumors has emerged as one of the most effective methods for cancer therapy. Nevertheless, single-mode immunotherapy is still confronted with several critical challenges, such as the low immune response, the low tumor infiltration, and the complex immunosuppression tumor microenvironment. Recently, the combination of immunotherapy with other therapeutic modalities has emerged as a powerful strategy to augment the therapeutic outcome in fighting against cancer. In this review, recent research advances of the combination of immunotherapy with chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, metabolic therapy, and microwave thermotherapy are summarized. Critical challenges and future research direction of immunotherapy-based cancer therapeutic strategy are also discussed.


Assuntos
Imunoterapia , Microambiente Tumoral , Terapia Combinada , Humanos , Terapia de Imunossupressão , Fototerapia
18.
Chem Commun (Camb) ; 57(74): 9398-9401, 2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34528964

RESUMO

A tumor cell membrane-camouflaged therapeutic system was fabricated to eliminate tumors by embedding apyrase and glucose oxidase (GOx) into zeolitic imidazolate framework-8 (ZIF-8) nanoparticles for tumor-targeted metabolic therapy. Experimental results demonstrated that these functional nanoparticles could disturb the energy supply of tumor cells by depleting ATP and glucose and efficiently induce tumor cell death.


Assuntos
Apirase/metabolismo , Materiais Biomiméticos/metabolismo , Glucose Oxidase/metabolismo , Estruturas Metalorgânicas/metabolismo , Nanopartículas/metabolismo , Neoplasias/metabolismo , Trifosfato de Adenosina/metabolismo , Materiais Biomiméticos/química , Morte Celular/efeitos dos fármacos , Glucose/metabolismo , Humanos , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Nanopartículas/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia
19.
ACS Nano ; 15(11): 17870-17884, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34747172

RESUMO

The excessive lactate in the tumor microenvironment always leads to poor therapeutic outcomes of chemotherapy. In this study, a self-driven bioreactor (defined as SO@MDH, where SO is Shewanella oneidensis MR-1 and MDH is MIL-101 metal-organic framework nanoparticles/doxorubicin/hyaluronic acid) is rationally constructed via the integration of doxorubicin (DOX)-loaded metal-organic framework (MOF) MIL-101 nanoparticles with SO to sensitize chemotherapy. Owing to the intrinsic tumor tropism and electron-driven respiration of SO, the biohybrid SO@MDH could actively target and colonize hypoxic and eutrophic tumor regions and anaerobically metabolize lactate accompanied by the transfer of electrons to Fe3+, which is the key component of the MIL-101 nanoparticles. As a result, the intratumoral lactate would undergo continuous catabolism coupled with the reduction of Fe3+ to Fe2+ and the subsequent degradation of MIL-101 frameworks, leading to an expeditious drug release for effective chemotherapy. Meanwhile, the generated Fe2+ will be promptly oxidized by the abundant hydrogen peroxide in the tumor microenvironment to reproduce Fe3+, which is, in turn, beneficial to circularly catabolize lactate and boost chemotherapy. More importantly, the consumption of intratumoral lactic acid could significantly inhibit the expression of multidrug resistance-related ABCB1 protein (also named P-glycoprotein (P-gp)) for conquering drug-resistant tumors. SO@MDH demonstrated here holds high tumor specificity and promising chemotherapeutic efficacy for suppressing tumor growth and overcoming multidrug resistance, confirming its potential prospects in cancer therapy.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Neoplasias , Humanos , Doxorrubicina/farmacologia , Neoplasias/terapia , Reatores Biológicos , Lactatos , Microambiente Tumoral
20.
ACS Macro Lett ; 9(6): 872-881, 2020 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35648534

RESUMO

Poly(N-isopropylacrylamide) (PNIPAAm)-based thermally responsive micelles are of great importance as smart materials for a number of applications such as drug delivery and biosensing, owing to their tunable lower critical solution temperature (LCST). Their design and synthesis in the nanoscale size range have been widely studied, and research interest in their structural and physic-chemical properties is continually growing. In this Viewpoint, representative research on the construction of PNIPAAm-based thermally responsive micelles as well as their applications are highlighted and discussed, which would serve as a good start for newcomers in this field and a positive guide for future research.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA