Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; : e2400425, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38574376

RESUMO

Active transcytosis-mediated nanomedicine transport presents considerable potential in overcoming diverse delivery barriers, thereby facilitating tumor accumulation and penetration. Nevertheless, the persistent challenge lies in achieving a nuanced equilibrium between intracellular interception for drug release and transcytosis for tumor penetration. In this study, a comprehensive exploration is conducted involving a series of polyglutamine-paclitaxel conjugates featuring distinct hydrophilic/hydrophobic ratios (HHR) and tertiary amine-oxide proportions (TP) (OPGA-PTX). The screening process, meticulously focused on delineating their subcellular distribution, transcytosis capability, and tumor penetration, unveils a particularly promising candidate denoted as OPPX, characterized by an HHR of 10:1 and a TP of 100%. OPPX, distinguished by its rapid cellular internalization through multiple endocytic pathways, selectively engages in trafficking to the Golgi apparatus for transcytosis to facilitate accumulation within and penetration throughout tumor tissues and simultaneously sorted to lysosomes for cathepsin B-activated drug release. This study not only identifies OPPX as an exemplary nanomedicine but also underscores the feasibility of modulating subcellular distribution to optimize the active transport capabilities and intracellular release mechanisms of nanomedicines, providing an alternative approach to designing efficient anticancer nanomedicines.

2.
Oncogene ; 43(16): 1163-1177, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38472320

RESUMO

Neutrophils, the most abundant immune cells in human blood, play crucial and diverse roles in tumor development. In the tumor microenvironment (TME), cancer cells regulate the recruitment and behaviors of neutrophils, transforming some of them into a pro-tumor phenotype. Pro-tumor neutrophils interact with cancer cells in various ways to promote cancer initiation, growth, and metastasis, while anti-tumor neutrophils interact with cancer cells to induce senescence and death. Neutrophils can also interact with other cells in TME, including T cells, macrophages, stromal cells, etc. to exert anti- or pro-tumor functions. In this review, we will analyze the anti- and pro-tumor intercellular interactions mediated by neutrophils, with a focus on generalizing the mechanisms underlying the interaction of neutrophils with tumor cells and T cells. Furthermore, we will provide an overview of cancer treatment strategies targeting neutrophil-mediated cellular interactions.


Assuntos
Neoplasias , Neutrófilos , Humanos , Neoplasias/patologia , Linfócitos T , Fenótipo , Microambiente Tumoral
3.
Acta Biomater ; 175: 240-249, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38103850

RESUMO

DNA hydrogels with unique sequence programmability on nucleic acid framework manifest remarkable attributes, such as high payload capacities, biocompatibility and biosafety. The availability of DNA nanogels with multimodal functionalities remains limited due to the absence of facile gelation methods applicable at the nanometer scale. Here, we developed a one-step assembly of DNA dendrimers into nanogels (DNG) with couple hundred nanometers size. DNG showed robust stability against physical forces and biological degradation for easy purification and sustainable drug release. Long-term stability either in powder or aqueous solution endows DNG easy for shipping, handling and storage. By encoding dual functionalities into separate branches on DNA dendrimers, DNG can accommodate chemodrugs and aptamers with distinctive loading moduli. DNG significantly enhanced the drug efficacy against cancerous cells while minimizing cytotoxicity towards somatic cells, as demonstrated in vitro and in xenografted mice models of breast cancer. Thus, due to their facile assembly and storage, bi-entity encoding, and inherent biocompatibility, DNG exhibits immense prospects as nanoscale vesicles for the synergistic delivery of multimodal theranostics in anticancer treatments. STATEMENT OF SIGNIFICANCE: DNA nanogels were self-assembled via a facile protocol utilizing a DNA dendrimer structure. These nanogels displayed robust stability against physical forces, permitting long term storage in concentrated solutions or as a powder. Furthermore, they exhibited resilience to biological degradation, facilitating sustained drug release. The bi-entity encoded dendritic branches conferred dual functionalities, enabling both chemodrug encapsulation and the presentation of aptamers as targeting motifs. In vivo investigations confirmed the nanogels provide high efficacy in tumor targeting and chemotherapy with enhanced drug efficacy and reduced side effects.


Assuntos
Antineoplásicos , Dendrímeros , Animais , Camundongos , Nanogéis , Doxorrubicina/química , Dendrímeros/química , Pós , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Antineoplásicos/química , DNA , Portadores de Fármacos/química , Liberação Controlada de Fármacos
4.
Sci Bull (Beijing) ; 68(22): 2779-2792, 2023 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-37863773

RESUMO

Immunotherapy has revolutionized cancer therapy, using chemical or biological agents to reinvigorate the immune system. However, most of these agents have poor tumor penetration and inevitable side effects that complicate therapeutic outcomes. Electrical stimulation (ES) is a promising alternative therapy against cancers that does not involve chemical or biological agents but is limited in the fabrication and operation of complex micrometer-scale ES devices. Here, we present an optically microprinted flexible interdigital electrode with a gold-plated polymer microneedle array to generate alternating electric fields for cancer treatment. A flexible microneedle-array-integrated interdigital electrode (FMIE) was fabricated by combining optical 3D microprinting and electroless plating processes. FMIE-mediated ES of cancer cells induced necrotic cell death through mitochondrial Ca2+ overload and increased intracellular reactive oxygen species (ROS) production. This led to the release of damage-associated molecular patterns that activated the immune response and potentiated immunogenic cell death (ICD). FMIE-based ES has an excellent safety profile and systemic anti-tumor effects, inhibiting the growth of primary and distant tumors as well as melanoma lung metastasis. FMIE-based ES-driven cancer immunomodulation provides a new pathway for drug-free cancer therapy.


Assuntos
Imunoterapia , Neoplasias Pulmonares , Humanos , Eletrodos , Estimulação Elétrica , Fatores Biológicos
5.
Chem Rev ; 123(18): 10920-10989, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37713432

RESUMO

Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Nanomedicina , Neoplasias/terapia , Sistemas de Liberação de Medicamentos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Preparações Farmacêuticas , Microambiente Tumoral
6.
Med Rev (2021) ; 3(2): 184-187, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37724083

RESUMO

Cancer nanomedicines require different, even opposite, properties to voyage the cascade drug delivery process involving a series of biological barriers. Currently-approved nanomedicines can only alleviate adverse effects but cannot improve patient survival because they fail to meet all the requirements. Therefore, nanocarriers with synchronized functions are highly requisite to capacitate efficient drug delivery and enhanced therapeutic efficacies. This perspective article summarizes recent advances in the two main strategies for nanomedicine design, the All-in-One approach (integration of all the functions in one system) and the One-for-All approach (one functional group with proper affinity enables all the functions), and presents our views on future nanomedicine development.

7.
J Control Release ; 361: 792-802, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37595665

RESUMO

Drug self-delivery systems (DSDSs) have been extensively exploited to enhance drug loading capacity and avoid excipient-related toxicity issues. However, deficient tumor targeting, inferior tumor permeability, prominent burst release, and nonspecific subcellular distribution remain major obstacles. Herein, we reported a ROS-responsive amphiphilic prodrug (CPT-S-NO) synthesized by the conjugation of zwitterionic tertiary amine-oxide (TAO) moiety and hydrophobic camptothecin (CPT) through a thioether linkage, which formed a nanoparticulate DSDS in an aqueous solution. CPT-S-NO, compared with CPT-11 and the water-soluble TAO-modified CPT prodrug (CPT-NO), exhibited prolonged blood circulation, enhanced tumor accumulation, deep tumor penetration, efficient mitochondrial targeting, and ROS-activated drug release to induce mitochondrial dysfunction, corporately conducing to the superior antitumor efficacy in vivo. This TAO decoration strategy promises potential applications in designing multipotent DSDSs for various drugs.


Assuntos
Nanopartículas , Neoplasias , Pró-Fármacos , Humanos , Espécies Reativas de Oxigênio , Neoplasias/tratamento farmacológico , Mitocôndrias , Óxidos , Água , Nanopartículas/uso terapêutico
8.
Adv Sci (Weinh) ; 10(29): e2301216, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37551065

RESUMO

Chemo-immunotherapy has made significant progress in cancer treatment. However, the cancer cell self-defense mechanisms, including cell cycle checkpoint and programmed cell death-ligand 1 (PD-L1) upregulation, have greatly hindered the therapeutic efficacy. Herein, norcantharidin (NCTD)-platinum (Pt) codelivery nanoparticles (NC-NP) with tumor-sensitive release profiles are designed to overcome the self-defense mechanisms via synergistic chemo-immunotherapy. NC-NP remains stable under normal physiological conditions but quickly releases 1,2-diaminocyclohexane-platinum(II) (DACHPt, a parent drug of oxaliplatin) and NCTD in response to the tumor acidity. NCTD inhibits protein phosphatase 2A (PP2A) activity to relieve cell cycle arrest and downregulates the tumor PD-L1 expression to disrupt the programmed cell death-1 (PD-1)/PD-L1 interaction, synergistically enhancing Pt-based chemotherapy and immunogenic cell death-induced immunotherapy. As a result, NC-NP exhibits potent synergistic cytotoxicity and promotes T cell recruitment to generate robust antitumor immune responses. The dual synergism exhibits potent antitumor activity against orthotopic 4T1 tumors, providing a promising chemo-immunotherapy paradigm for cancer treatment.


Assuntos
Imunoterapia , Nanopartículas , Neoplasias , Humanos , Antígeno B7-H1 , Platina , Polímeros
9.
Adv Mater ; 35(46): e2303614, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37490011

RESUMO

Ionizable cationic lipids are recognized as an essential component of lipid nanoparticles (LNPs) for messenger RNA (mRNA) delivery but can be confounded by low lipoplex stability with mRNA during storage and in vivo delivery. Herein, the rational design and combinatorial synthesis of esterase-triggered decationizable quaternium lipid-like molecules (lipidoids) are reported to develop new LNPs with high delivery efficiency and improved storage stability. This top lipidoid carries positive charges at the physiological condition but promptly acquires negative charges in the presence of esterase, thus permitting stable mRNA encapsulation during storage and in vivo delivery while balancing efficient mRNA release in the cytosol. An optimal LNP formulation is then identified through orthogonal optimization, which enables efficacious mRNA transfection selectively in the spleen following intravenous administration. LNP-mediated delivery of ovalbumin (OVA)-encoding mRNA induces efficient antigen expression in antigen-presenting cells and elicits robust antigen-specific immune responses against OVA-transduced tumors. The work demonstrates the potential of decationizable quaternium lipidoids for spleen-selective RNA transfection and cancer immunotherapy.


Assuntos
Esterases , Nanopartículas , Baço/metabolismo , RNA Mensageiro/metabolismo , Transfecção , RNA Interferente Pequeno/genética
10.
J Control Release ; 357: 310-318, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37019286

RESUMO

Dendritic cells (DCs), the primary antigen-presenting cells in the immune system, play a critical role in regulating tumor immune responses. However, the tumor immunosuppressive microenvironment severely impedes the process of antigen-presenting and DC maturation, thereby limiting the efficacy of cancer immunotherapy. In this work, a pH-responsive polymer nanocarrier (PAG) modified with aminoguanidine (AG) was constructed for the efficient delivery of bortezomib (BTZ) through bidentate hydrogen bonds and electrostatic adsorption formed between guanidine groups of PAG and boronic acid groups of BTZ. The obtained PAG/BTZ nanoparticles exhibited pH-responsive release of BTZ and AG in the acidic tumor microenvironment. On the one hand, BTZ induced potent immune activation by eliciting immunogenic cell death (ICD) and releasing damage-associated molecular patterns. On the other hand, the cationic AG significantly promoted antigen uptake by DCs and activated DC maturation. As a result, PAG/BTZ significantly stimulated tumoral infiltration of cytotoxic T lymphocytes (CTLs) and triggered robust antitumor immune responses. Thus, it showed potent antitumor efficacy when synergizing with an immune checkpoint-blocking antibody.


Assuntos
Nanopartículas , Neoplasias , Humanos , Bortezomib/farmacologia , Portadores de Fármacos/química , Guanidina , Neoplasias/tratamento farmacológico , Antígenos , Imunidade , Nanopartículas/química , Imunoterapia , Linhagem Celular Tumoral , Microambiente Tumoral
11.
Nano Lett ; 23(9): 3904-3912, 2023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-37043295

RESUMO

Transcytosis-based active transport of cancer nanomedicine has shown great promise for enhancing its tumor extravasation and infiltration and antitumor activity, but how the key nanoproperties of nanomedicine, particularly particle size, influence the transcytosis remains unknown. Herein, we used a transcytosis-inducing polymer, poly[2-(N-oxide-N,N-diethylamino)ethyl methacrylate] (OPDEA), and fabricated stable OPDEA-based micelles with different sizes (30, 70, and 140 nm in diameter) from its amphiphilic block copolymer, OPDEA-block-polystyrene (OPDEA-PS). The study of the micelle size effects on cell transcytosis, tumor extravasation, and infiltration showed that the smallest micelles (30 nm) had the fastest transcytosis and, thus, the most efficient tumor extravasation and infiltration. So, the 7-ethyl-10-hydroxyl camptothecin (SN38)-conjugated OPDEA micelles of 30 nm had much enhanced antitumor activity compared with the 140 nm micelles. These results are instructive for the design of active cancer nanomedicine.


Assuntos
Camptotecina , Micelas , Linhagem Celular Tumoral , Camptotecina/farmacologia , Polímeros , Transcitose , Resultado do Tratamento , Tamanho da Partícula
12.
J Am Chem Soc ; 145(14): 7941-7951, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-36987634

RESUMO

Tumor-associated macrophages, especially M2-like macrophages, are extensively involved in tumor growth and metastasis, suppressing the innate immunity to help tumor cells escape and reshaping the microenvironment to help metastatic cells grow. However, in vivo, real-time visualized migration of M2-like macrophages has never been explored to monitor the tumor metastasis process. Herein, we prepared an M2-like macrophage-targeting nitric oxide (NO)-responsive nanoprobe (NRP@M-PHCQ) consisting of an amphiphilic block copolymer with mannose and hydroxychloroquine (HCQ) moieties (denoted as M-PHCQ) and a NO-responsive NIR-II probe (denoted as NRP). The mannose moieties provided M2-like macrophage-targeting capacity, and the HCQ moieties polarized M2-like macrophages to M1-like ones with enhanced NO secretion. Consequently, NRP@M-PHCQ was lit up by the secreted NO to visualize the migration and polarization of M2-like macrophages in real time. In vivo metastasis imaging with NRP@M-PHCQ successfully tracked early tumor metastasis in the lymph nodes and the lungs with high sensitivity, even superior to Luci-labeled bioluminescence imaging, suggesting the extensive distribution and critical role of M2-like macrophages in tumor metastasis. In general, this work provided a new strategy to sensitively image metastatic tumors by tracking the polarization of M2-like macrophages and visually disclosed the critical role of M2-like macrophages in early tumor metastasis.


Assuntos
Macrófagos , Manose , Linhagem Celular Tumoral
13.
Angew Chem Int Ed Engl ; 62(9): e202217408, 2023 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-36594796

RESUMO

Tumor enzyme-responsive charge-reversal carriers can induce efficient transcytosis and lead to efficient tumor infiltration and potent anticancer efficacy. However, the correlations of molecular structure with charge-reversal property, tumor penetration, and drug delivery efficiency are unknown. Herein, aminopeptidase N (APN)-responsive conjugates were synthesized to investigate these correlations. We found that the monomeric unit structure and the polymer chain structure determined the enzymatic hydrolysis and charge-reversal rates, and accordingly, the transcytosis and tumor accumulation and penetration of the APN-responsive conjugates. The conjugate with moderate APN responsiveness balanced the in vitro transcytosis and in vivo overall drug delivery process and achieved the best tumor delivery efficiency, giving potent antitumor efficacy. This work provides new insight into the design of tumor enzyme-responsive charge-reversal nanomedicines for efficient cancer drug delivery.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Antígenos CD13/uso terapêutico , Antineoplásicos/química , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Polímeros/química , Nanopartículas/química , Linhagem Celular Tumoral , Doxorrubicina/química
14.
Adv Drug Deliv Rev ; 191: 114614, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36347432

RESUMO

Over the past three decades, the enhanced permeability and retention (EPR) effect has been considered the basis of tumor-targeted drug delivery. Various cancer nanomedicines, including macromolecular drugs, have been designed to utilize this mechanism for preferential extravasation and accumulation in solid tumors. However, such nanomedicines have not yet achieved convincing therapeutic benefits in clinics. Increasing evidence suggests that the EPR effect is over-represented in human tumors, especially in metastatic tumors. This review covers the evolution of the concept, the heterogeneity and limitation of the EPR effect in clinical realities, and prospects for alternative strategies independent of the EPR effect.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Neoplasias/terapia , Sistemas de Liberação de Medicamentos , Antineoplásicos/uso terapêutico , Nanomedicina , Permeabilidade
15.
Adv Drug Deliv Rev ; 189: 114480, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35952830

RESUMO

Extravasation is the first step for nanomedicines in circulation to reach targeted solid tumors. Traditional nanomedicines have been designed to extravasate into tumor interstitium through the interendothelial gaps previously assumed rich in tumor blood vessels, i.e., the enhanced permeability and retention (EPR) effect. While the EPR effect has been validated in animal xenograft tumor models, accumulating evidence implies that the EPR effect is very limited and highly heterogeneous in human tumors, leading to highly unpredictable and inefficient extravasation and thus limited therapeutic efficacy of nanomedicines, including those approved in clinics. Enabling EPR-independent extravasation is the key to develop new generation of nanomedicine with enhanced efficacy. Transcytosis of tumor endothelial cells can confer nanomedicines to actively extravasate into solid tumors without relying on the EPR effect. Here, we review and prospectthe development of transcytosis-inducing nanomedicines, in hope of providing instructive insights for design of nanomedicines that can undergo selective transcellular transport across tumor endothelial cells, and thus inspiring the development of next-generation nanomedicines for clinical translation.


Assuntos
Antineoplásicos , Neoplasias , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos , Células Endoteliais , Humanos , Nanomedicina , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Transcitose
16.
J Control Release ; 348: 444-455, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35691498

RESUMO

Cancer drug delivery systems often suffer from premature drug leakage during transportation and/or inefficient drug release within cancer cells. We present here a polymeric prodrug nanoassembly that addresses these problems simultaneously. This nanoassembly comprises a polymeric prodrug with novel trivalent phenylboronate moieties for drug conjugation via ether linkages, as well as ß-lapachone (Lapa). While the ether linkage enables nearly no drug release under physiological conditions, the Lapa molecules can induce the reactive oxygen species (ROS) burst specifically in cancer cells via NAD(P)H: quinone oxidoreductase-1 catalysis, which triggers the cleavage of the ether bonds and thus cascade amplification drug release in cancer cells. As a result, the nanoassemblies exhibit much higher cytotoxicity against cancer cells than normal cells, and also increased therapeutic efficacy and reduced side effects compared to the clinically used irinotecan. We anticipate that this strategy can be applied to other drug delivery platforms to enable more precise drug release.


Assuntos
Nanopartículas , Pró-Fármacos , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Éteres , Nanopartículas/química , Polímeros/química , Pró-Fármacos/química
17.
Mater Today Bio ; 14: 100284, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35647515

RESUMO

Immune checkpoint blockade (ICB) therapies that target programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway are currently used for the treatment of various cancer types. However, low response rates of ICB remain the major issue and limit their applications in clinic. Here, we developed a ROS-responsive synergistic delivery system (pep-PAPM@PTX) by integrating physically-encapsulated paclitaxel (PTX) and surface-modified anti-PD-L1 peptide (pep) for combined chemotherapy and ICB therapy. Pep-PAPM@PTX could bind the cell surface PD-L1 and drive its recycling to lysosomal degradation, thus reverting PTX-induced PD-L1 upregulation and downregulating PD-L1 expression. As a result, pep-PAPM@PTX significantly promoted T cell infiltration and increased tumor immunoactivating factors, synergizing PTX chemotherapy to achieve enhanced anticancer potency in a triple-negative breast cancer (TNBC) model.

18.
Nano Lett ; 22(13): 5615-5625, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35749341

RESUMO

Nanocarriers have been employed extensively to enhance drug delivery efficacy and reduce the side effect. However, carrier materials for drug delivery have challenging aspects, including safety concerns, low drug content, complexity in preparation, and low reproducibility. Herein, we propose a facile, universal, and green preparation way to use natural polyphenols to build platinum nanocomplex with stable structure, proper size, and high Pt content. The nanocomplexes are constructed by metal-polyphenol coordination using natural polyphenols and 1,2-diaminocyclohexane-Pt (II), enabling dual-responsive drug release behavior. For proof of concept, we demonstrate the antitumor activity of the Pt nanocomplex using a representative tannic acid-Pt nanocomplex (denoted as PTI). PTI can induce intensive tumor cell apoptosis, trigger immunogenic cell death (ICD), remarkably promote cytotoxic T lymphocytes (CTLs) infiltration in tumors, and significantly reduce immunosuppression of the tumor microenvironments, thus stimulating potent antitumor immune responses and showing effective antitumor activity by synergizing immune checkpoint blockade (ICB) therapy.


Assuntos
Neoplasias , Platina , Linhagem Celular Tumoral , Humanos , Imunoterapia , Neoplasias/tratamento farmacológico , Platina/uso terapêutico , Polifenóis/farmacologia , Polifenóis/uso terapêutico , Reprodutibilidade dos Testes , Linfócitos T Citotóxicos , Microambiente Tumoral
19.
Sci China Chem ; 65(6): 1010-1075, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35505924

RESUMO

Biomedical polymers have been extensively developed for promising applications in a lot of biomedical fields, such as therapeutic medicine delivery, disease detection and diagnosis, biosensing, regenerative medicine, and disease treatment. In this review, we summarize the most recent advances in the synthesis and application of biomedical polymers, and discuss the comprehensive understanding of their property-function relationship for corresponding biomedical applications. In particular, a few burgeoning bioactive polymers, such as peptide/biomembrane/microorganism/cell-based biomedical polymers, are also introduced and highlighted as the emerging biomaterials for cancer precision therapy. Furthermore, the foreseeable challenges and outlook of the development of more efficient, healthier and safer biomedical polymers are discussed. We wish this systemic and comprehensive review on highlighting frontier progress of biomedical polymers could inspire and promote new breakthrough in fundamental research and clinical translation.

20.
Adv Sci (Weinh) ; 9(12): e2200173, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35187868

RESUMO

The cancer drug delivery process involves a series of biological barriers, which require the nanomedicine to exhibit different, even opposite properties for high therapeutic efficacy. The prevailing design philosophy, i.e., integrating these properties within one nanomedicine via on-demand property transitions such as PEGylation/dePEGylation, complicates nanomedicines' composition and thus impedes clinical translation. Here, polyzwitterionic micelles of poly(tertiary amine-oxide)-block-poly(ε-caprolactone) (PTAO-PCL) amphiphiles that enable all the required functions are presented. The zwitterionic nature and unique cell membrane affinity confer the PTAO micelles long blood circulation, efficient tumor accumulation and penetration, and fast cellular internalization. The mitochondrial targeting capability allows drug delivery into the mitochondria to induce mitochondrial dysfunction and overcome tumor multidrug resistance. As a result, the PTAO/drug micelles exhibit potent anticancer efficacy. This simple yet multipotent carrier system holds great promise as a generic platform for potential clinical translation.


Assuntos
Antineoplásicos , Neoplasias , Aminas/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Humanos , Micelas , Neoplasias/tratamento farmacológico , Óxidos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA