Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
J Control Release ; 373: 216-223, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39002797

RESUMO

Platinum(II)-based drugs (PtII), which hinder DNA replication, are the most widely used chemotherapeutics. However, current PtII drugs often miss their DNA targets, leading to severe side effects and drug resistance. To overcome this challenge, we developed a oxaliplatin-based platinum(IV) (PtIV) prodrug amphiphile (C16-OPtIV-R8K), integrating a long-chain hydrophobic lipid and a nucleus-targeting hydrophilic peptide (R8K). This design allows the prodrug to self-assemble into highly uniform lipid nanoparticles (NTPtIV) for enhanced targeting chemotherapy and immunotherapy. Subsequently, NTPtIV's bioactivity and effects were examined at diverse levels, encompassing cancer cells, 3D tumor spheres, and in vivo. Our in vitro studies show a 74% cancer cell nucleus localization of platinum drugs-3.6 times higher than that of oxaliplatin, achieving more than a ten-fold increase in eliminating drug-resistant cancer cells. In vivo, NTPtIV shows efficient tumor accumulation, leading to suppressed tumor growth of murine breast cancer. Moreover, NTPtIV recruited more CD4+ and CD8+ T cells and reduced CD4+ Foxp3+ Tregs to synergistically enhance targeted chemotherapy and immunotherapy. Overall, this strategy presents a promising advancement in nucleus-targeted cancer therapy, synergistically boosting the efficacy of chemotherapy and immunotherapy.

2.
Small ; : e2401397, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898735

RESUMO

Pyroptosis, an inflammatory cell death, plays a pivotal role in activating inflammatory response, reversing immunosuppression and enhancing anti-tumor immunity. However, challenges remain regarding how to induce pyroptosis efficiently and precisely in tumor cells to amplify anti-tumor immunotherapy. Herein, a pH-responsive polydopamine (PDA) nanocluster, perfluorocarbon (PFC)@octo-arginine (R8)-1-Hexadecylamine (He)-porphyrin (Por)@PDA-gambogic acid (GA)-cRGD (R-P@PDA-GC), is rationally design to augment phototherapy-induced pyroptosis and boost anti-tumor immunity through a two-input programmed cascade therapy. Briefly, oxygen doner PFC is encapsulated within R8 linked photosensitizer Por and He micelles as the core, followed by incorporation of GA and cRGD peptides modified PDA shell, yielding the ultimate R-P@PDA-GC nanoplatforms (NPs). The pH-responsive NPs effectively alleviate hypoxia by delivering oxygen via PFC and mitigate heat resistance in tumor cells through GA. Upon two-input programmed irradiation, R-P@PDA-GC NPs significantly enhance reactive oxygen species production within tumor cells, triggering pyroptosis via the Caspase-1/GSDMD pathway and releasing numerous inflammatory factors into the TME. This leads to the maturation of dendritic cells, robust infiltration of cytotoxic CD8+ T and NK cells, and diminution of immune suppressor Treg cells, thereby amplifying anti-tumor immunity.

3.
Int J Biol Macromol ; 272(Pt 2): 132919, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38843673

RESUMO

Poly (vinyl alcohol) (PVA) hydrogel showed potential applications in bioengineering and wearable sensors fields. It is still a huge challenge to prepare highly adhesive yet strong poly (vinyl alcohol) hydrogel with good biocompatibility. Herein, we prepared a highly self-adhesive and strong poly (vinyl alcohol)/tannic acid@cellulose nanocrystals (PVA/TA@CNCs) composite hydrogel using TA@CNCs as functional nanofiller via facile freezing-thawing method. Multiple networks consisting of hydrogen bonding and coordination interactions endowed the hydrogel with high mechanical strength, excellent flexibility and fracture toughness with adequate energy dissipation mechanism and relatively dense network structure. The tensile strength of PVA/TA@CNCs hydrogel reached the maximum of 463 kPa, increasing by 367 % in comparison with pure PVA hydrogel (99 kPa), demonstrating the synergistic reinforcing and toughening effect of TA@CNCs. The hydrogel exhibited extremely high adhesion not only for various dry and wet substrates such as plastic, metal, Teflon, rubber, glass, leaf, but also sweaty human skin, showing good adhesion durability. The highest adhesion strength to silicone rubber, steel plate and pigskin could reach 197 kPa, 100 kPa and 46.9 kPa, respectively. Meanwhile the hydrogel had negligible cytotoxicity to cells and showed good biocompatibility.


Assuntos
Celulose , Hidrogéis , Nanopartículas , Álcool de Polivinil , Álcool de Polivinil/química , Celulose/química , Nanopartículas/química , Humanos , Hidrogéis/química , Resistência à Tração , Materiais Biocompatíveis/química , Adesivos/química , Animais
4.
ACS Nano ; 18(20): 12870-12884, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38727063

RESUMO

Epirubicin (EPI) alone can trigger mildly protective autophagy in residual tumor cells, resulting in an immunosuppressive microenvironment. This accelerates the recurrence of residual tumors and leads to antiprogrammed death ligand 1 (anti-PD-1)/PD-L1 therapy resistance, posing a significant clinical challenge in tumor immunotherapy. The combination of checkpoint inhibitors targeting the PD-1/PD-L1 pathway and amplifying autophagy presents an innovative approach to tumor treatment, which can prevent tumor immune escape and enhance therapeutic recognition. Herein, we aimed to synthesize a redox-triggered autophagy-induced nanoplatform with SA&EA-induced PD-L1 inhibition. The hyaluronic acid (HA) skeleton and arginine segment promoted active nanoplatform targeting, cell uptake, and penetration. The PLGLAG peptide was cleaved by overexpressing matrix metalloproteinase-2 (MMP-2) in the tumor microenvironment, and the PD-L1 inhibitor D-PPA was released to inhibit tumor immune escape. The intense autophagy inducers, STF-62247 and EPI, were released owing to the cleavage of disulfide bonds influenced by the high glutathione (GSH) concentration in tumor cells. The combination of EPI and STF induced apoptosis and autophagic cell death, effectively eliminating a majority of tumor cells. This indicated that the SA&EA nanoplatform has better therapeutic efficacy than the single STF@AHMPP and EPI@AHMPTP groups. This research provided a way to set up a redox-triggered autophagy-induced nanoplatform with PD-L1 inhibition to enhance chemo-immunotherapy.


Assuntos
Autofagia , Antígeno B7-H1 , Imunoterapia , Nanopartículas , Animais , Humanos , Camundongos , Autofagia/efeitos dos fármacos , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/química , Nanopartículas/química , Oxirredução , Microambiente Tumoral/efeitos dos fármacos
5.
Adv Sci (Weinh) ; 11(25): e2401214, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38647420

RESUMO

Deep penetration and downregulation of heat shock protein (HSP) expression in multimodal synergistic therapy are promising approaches for curing cancer in clinical trials. However, free small-molecule drugs and most drug vehicles have a low delivery efficiency deep into the tumor owing to poor drug penetration and hypoxic conditions at the tumor site. In this study, the objective is to use reactive oxygen species (ROS)-responsive supramolecular gels co-loaded with the photosensitizer Zn(II) phthalocyanine tetrasulfonic acid (ZnPCS4) and functionalized tetrahedral DNA (TGSAs) (G@P/TGSAs) to enhance deep tissue and cell penetration and block the HSP90 pathway for chemo- photodynamic therapy (PDT) - photothermal therapy (PTT) trimodal synergistic therapy. The (G@P/TGSAs) are injected in situ into the tumor to release ZnPCS4 and TGSAs under high ROS concentrations originating from both the tumor and PDT. TGSAs penetrate deeply into tumor tissues and augment photothermal therapy by inhibiting the HSP90 pathway. Proteomics show that HSP-related proteins and molecular chaperones are inhibited/activated, inhibiting the HSP90 pathway. Simultaneously, the TGSA-regulated apoptotic pathway is activated. In vivo study demonstrates efficient tumor penetration and excellent trimodal synergistic therapy (45% tumor growth inhibition).


Assuntos
Proteínas de Choque Térmico HSP90 , Fotoquimioterapia , Espécies Reativas de Oxigênio , Espécies Reativas de Oxigênio/metabolismo , Animais , Camundongos , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Fotoquimioterapia/métodos , Géis , Fármacos Fotossensibilizantes/farmacologia , Terapia Fototérmica/métodos , Modelos Animais de Doenças , Indóis/farmacologia , Humanos , Terapia Combinada/métodos , Linhagem Celular Tumoral , Isoindóis
6.
Colloids Surf B Biointerfaces ; 237: 113871, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38547796

RESUMO

Highly toxic reactive oxygen species (ROS), crucial in inducing apoptosis and ferroptosis, are pivotal for cell death pathways in cancer therapy. However, the effectiveness of ROS-related tumor therapy is impeded by the limited intracellular ROS and substrates, coupled with the presence of abundant ROS scavengers like glutathione (GSH). In this research, we developed acid-responsive, iron-coordinated polymer nanoparticles (PPA/TF) encapsulating a mitochondrial-targeting drug alpha-tocopheryl succinate (α-TOS) for enhanced synergistic tumor treatment. The imidazole grafted micelles exhibit prolonged blood circulation and improve the delivery efficiency of the hydrophobic drug α-TOS. Additionally, PPA's design aids in delivering Fe3+, supplying ample iron ions for chemodynamic therapy (CDT) and ferroptosis through the attachment of imidazole groups to Fe3+. In the tumor's weakly acidic intracellular environment, PPA/TF facilitates pH-responsive drug release. α-TOS specifically targets mitochondria, generating ROS and replenishing those depleted by the Fenton reaction. Moreover, the presence of Fe3+ in PPA/TF amplifies ROS upregulation, promotes GSH depletion, and induces oxidative damage and ferroptosis, effectively inhibiting tumor growth. This research presents an innovative ROS-triggered amplification platform that optimizes CDT and ferroptosis for effective cancer treatment.


Assuntos
Ferroptose , Neoplasias , Humanos , Espécies Reativas de Oxigênio/metabolismo , Micelas , Linhagem Celular Tumoral , Polímeros/química , Neoplasias/patologia , Ferro/química , Concentração de Íons de Hidrogênio , Imidazóis/farmacologia , Peróxido de Hidrogênio/química , Glutationa/metabolismo , Microambiente Tumoral
7.
J Am Chem Soc ; 146(1): 1185-1195, 2024 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-38148611

RESUMO

Patients treated with Pt-based anticancer drugs (PtII) often experience severe side effects and are susceptible to cancer recurrence due to the limited bioavailability of PtII and tumor-induced immunosuppression. The exposure of phosphatidylserine on the cell's outer surface induced by PtII results in profound immunosuppression through the binding of phosphatidylserine to its receptors on immune cells. Here, we report a novel approach for enhanced cancer chemoimmunotherapy, where a novel nuclear-targeting lipid PtIV prodrug amphiphile was used to deliver a small interfering RNA (siXkr8) to simultaneously amplify Pt-DNA adducts and reduce the level of exposure of phosphatidylserine. This drug delivery vehicle is engineered by integrating the PtIV prodrug with self-assembly performance and siXkr8 into a lipid nanoparticle, which shows tumor accumulation, cancer cell nucleus targeting, and activatable in a reduced microenvironment. It is demonstrated that nuclear-targeting lipid PtIV prodrug increases the DNA cross-linking, resulting in increased Pt-DNA adduct formation. The synergistic effects of the PtIV prodrug and siXkr8 contribute to the improvement of the tumor immune microenvironment. Consequently, the increased Pt-DNA adducts and immunogenicity effectively inhibit primary tumor growth and prevent tumor recurrence. These results underscore the potential of utilizing the nuclear-targeting lipid PtIV prodrug amphiphile to enhance Pt-DNA adduct formation and employing siXkr8 to alleviate immunosuppression during chemotherapy.


Assuntos
Antineoplásicos , Neoplasias , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Adutos de DNA , Fosfatidilserinas , RNA Interferente Pequeno , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , RNA de Cadeia Dupla , Linhagem Celular Tumoral , Cisplatino , Microambiente Tumoral
8.
J Mater Chem B ; 12(2): 413-435, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38112639

RESUMO

Ferroptosis is a new form of non-apoptotic programmed cell death. Due to its effectiveness in cancer treatment, there are increasing studies on the application of nanoparticles based on ferroptosis in cancer therapy. In this paper, we present a summary of the latest progress in nanoparticles based on ferroptosis for effective tumor therapy. We also describe the combined treatment of ferroptosis with other therapies, including chemotherapy, radiotherapy, phototherapy, immunotherapy, and gene therapy. This summary of drug delivery systems based on ferroptosis aims to provide a basis and inspire opinions for researchers concentrating on exploring this field. Finally, we present some prospects and challenges for the application of nanotherapies to clinical treatment by promoting ferroptosis in cancer cells.


Assuntos
Ferroptose , Nanopartículas , Neoplasias , Terapia Combinada , Imunoterapia , Fototerapia , Neoplasias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...