Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biomater Sci ; 11(22): 7445-7457, 2023 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-37819252

RESUMO

The topoisomerase I inhibitor, 7-ethyl-10-hydroxycamptothecin (SN38), has demonstrated potent anticancer activity. However, its clinical application is hindered by its low solubility and high crystallization propensity, which further complicates its encapsulation into nanoparticles for systemic delivery. Herein, we explore the utilization of lipid-assisted poly(ethylene glycol)-block-poly(D,L-lactide) (PEG-b-PLA) nanoparticles to achieve ultrahigh loading capability for SN38. Through the introduction of cationic, anionic, or neutral lipids, the SN38 loading efficiency and loading capacity is elevated to >90% and >10% respectively. These lipids efficiently attenuate the intermolecular π-π stacking of SN38, thereby disrupting its crystalline structure. Moreover, we assess the therapeutic activity of SN38-loaded formulations in various tumor models and identify an anionic lipid 1,2-dioleoyl-sn-glycero-3-phospho-(1'-rac-glycerol) sodium salt (DOPG)-assisted formulation that exhibits the highest anticancer activity and has favorable biosafety. Overall, our findings present a simple and robust strategy to achieve ultrahigh loading efficiency of SN38 using commonly employed PEG-b-PLA nanoparticles, opening up a new avenue for the systemic delivery of SN38.


Assuntos
Nanopartículas , Neoplasias , Humanos , Polietilenoglicóis/química , Nanopartículas/química , Álcoois Graxos , Poliésteres , Linhagem Celular Tumoral
2.
Nat Commun ; 14(1): 1993, 2023 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-37031188

RESUMO

PD-1/PD-L1 blockade therapy that eliminates T-cell inhibition signals is successful, but poor benefits are often observed. Increasing T-cell infiltration and quantity of PD-1/PD-L1 inhibitors in tumor can improve efficacy but remains challenging. Here, we devise tumor-specific gene nanomedicines to mobilize tumor cells to secrete CXCL9 (T-cell chemokine) and anti-PD-L1 scFv (αPD-L1, PD-L1 blocking agent) for enhanced immunotherapy. The tyrosinase promoter-driven NPTyr-C9AP can specifically co-express CXCL9 and αPD-L1 in melanoma cells, thereby forming a CXCL9 gradient for T-cell recruitment and high intratumoral αPD-L1 concentration for enhancing T-cell activation. As a result, NPTyr-C9AP shows strong antimelanoma effects. Moreover, specific co-expression of CXCL9 and αPD-L1 in various tumor cells is achieved by replacing the tyrosinase promoter of NPTyr-C9AP with a survivin promoter, which increases T-cell infiltration and activation and therapeutic efficacy in multiple tumors in female mice. This study provides a strategy to maximize the immunotherapeutic outcome regardless of the heterogeneous tumor microenvironment.


Assuntos
Neoplasias , Linfócitos T , Feminino , Camundongos , Animais , Receptor de Morte Celular Programada 1 , Monofenol Mono-Oxigenase , Nanomedicina , Imunoterapia , Antígeno B7-H1/genética , Microambiente Tumoral , Linhagem Celular Tumoral
3.
Pharm Res ; 40(1): 145-156, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36002611

RESUMO

PURPOSE: Hepatitis B virus (HBV) infection is such a global health problem that hundreds of millions of people are HBV carriers. Current anti-viral agents can inhibit HBV replication, but can hardly eradicate HBV. Cytosine-phosphate-guanosine (CpG) oligodeoxynucleotides (ODNs) are an adjuvant that can activate plasmacytoid dendritic cells (pDCs) and conventional dendritic cells (cDCs) to induce therapeutic immunity for HBV eradication. However, efficient delivery of CpG ODNs into pDCs and cDCs remains a challenge. In this study, we constructed a series of cationic lipid-assisted nanoparticles (CLANs) using different cationic lipids to screen an optimal nanoparticle for delivering CpG ODNs into pDCs and cDCs. METHODS: We constructed different CLANCpG using six cationic lipids and analyzed the cellular uptake of different CLANCpG by pDCs and cDCs in vitro and in vivo, and further analyzed the efficiency of different CLANCpG for activating pDCs and cDCs in both wild type mice and HBV-carrier mice. RESULTS: We found that CLAN fabricated with 1,2-Dioleoyl-3-trimethylammonium propane (DOTAP) showed the highest efficiency for delivering CpG ODNs into pDCs and cDCs, resulting in strong therapeutic immunity in HBV-carrier mice. By using CLANCpG as an immune adjuvant in combination with the injection of recombinant hepatitis B surface antigen (rHBsAg), HBV was successfully eradicated and the chronic liver inflammation in HBV-carrier mice was reduced. CONCLUSION: We screened an optimized CLAN fabricated with DOTAP for efficient delivery of CpG ODNs to pDCs and cDCs, which can act as a therapeutic vaccine adjuvant for treating HBV infection.


Assuntos
Hepatite B , Nanopartículas , Camundongos , Animais , Vírus da Hepatite B , Oligodesoxirribonucleotídeos/farmacologia , Fosfatos , Citosina , Guanosina , Hepatite B/tratamento farmacológico , Ácidos Graxos Monoinsaturados , Adjuvantes Imunológicos/uso terapêutico , Células Dendríticas
4.
J Control Release ; 345: 494-511, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35337940

RESUMO

Abnormal immune cell functions are commonly related to various diseases, including cancer, autoimmune diseases, and infectious diseases. Messenger RNA (mRNA)-based therapy can regulate the functions of immune cells or assign new functions to immune cells, thereby generating therapeutic immune responses to treat these diseases. However, mRNA is unstable in physiological environments and can hardly enter the cytoplasm of target cells; thus, effective mRNA delivery systems are critical for developing mRNA therapy. The two mRNA vaccines of Pfizer-BioNTech and Moderna have demonstrated that lipid nanoparticles (LNPs) can deliver mRNA into dendritic cells (DCs) to induce immunization against severe acute respiratory syndrome coronavirus 2, which opened the floodgates to the development of mRNA therapy. Apart from DCs, other immune cells are promising targets for mRNA therapy. This review summarized the barriers to mRNA delivery and advances in mRNA delivery for regulating the functions of different immune cells.


Assuntos
COVID-19 , Nanopartículas , COVID-19/terapia , Vacinas contra COVID-19 , Humanos , Lipossomos , RNA Mensageiro/genética , SARS-CoV-2/genética
5.
ACS Appl Mater Interfaces ; 13(25): 29424-29438, 2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-34129318

RESUMO

Efficient capture and presentation of tumor antigens by antigen-presenting cells (APCs), especially dendritic cells (DCs), are crucial for activating the anti-tumor immunity. However, APCs are immunosuppressed in the tumor microenvironment, which hinders the tumor elimination. To reprogram APCs for inducing strong anti-tumor immunity, we report here a co-delivery immunotherapeutic strategy targeting the phagocytosis checkpoint (signal regulatory protein α, SIRPα) and stimulator of interferon genes (STING) of APCs to jointly enhance their ability of capturing and presenting tumor antigens. In brief, a small interfering RNA targeting SIRPα (siSIRPα) and a STING agonist (cGAMP) were co-delivered into APCs by the encapsulation into poly(ethylene glycol)-b-poly(lactide-co-glycolide)-based polymeric nanoparticles (NPsiSIRPα/cGAMP). siSIRPα-mediated SIRPα silence promoted APCs to actively capture tumor antigens by engulfing tumor cells. The cGAMP-stimulated STING signaling pathway further enhanced the functions of APCs, thereby increased the activation and expansion of CD8+ T cells. Using ovalbumin (OVA)-expressing melanoma as a model, we demonstrated that NPsiSIRPα/cGAMP stimulated the activation of OVA-specific CD8+ T cells and induced holistic anti-tumor immune responses by reversing the immunosuppressive phenotype of APCs. Collectively, this co-delivery strategy synergistically enhanced the functions of APCs and can be extended to the treatment of tumors with poor immunogenicity.


Assuntos
Antineoplásicos , Imunoterapia/métodos , Proteínas de Membrana , Receptores Imunológicos/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Masculino , Proteínas de Membrana/agonistas , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais , Nucleotídeos Cíclicos , Fagocitose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
6.
Adv Drug Deliv Rev ; 168: 3-29, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-31759123

RESUMO

The CRISPR-Cas system initiated a revolution in genome editing when it was, for the first time, demonstrated success in the mammalian cells. Today, scientists are able to readily edit genomes, regulate gene transcription, engineer posttranscriptional events, and image nucleic acids using CRISPR-Cas-based tools. However, to efficiently transport CRISPR-Cas into target tissues/cells remains challenging due to many extra- and intra-cellular barriers, therefore largely limiting the applications of CRISPR-based therapeutics in vivo. In this review, we summarize the features of plasmid-, RNA- and ribonucleoprotein (RNP)-based CRISPR-Cas therapeutics. Then, we survey the current in vivo delivery systems. We specify the requirements for efficient in vivo delivery in clinical settings, and highlight both efficiency and safety for different CRISPR-Cas tools.


Assuntos
Sistemas CRISPR-Cas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Edição de Genes/métodos , Sistemas de Liberação de Medicamentos , Epigenoma/genética , Exossomos/metabolismo , Redes Reguladoras de Genes/fisiologia , Vetores Genéticos/metabolismo , Lipídeos/química , Nanopartículas/química , RNA/metabolismo , Transcrição Gênica/fisiologia
7.
ACS Appl Mater Interfaces ; 12(43): 48259-48271, 2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-33070614

RESUMO

Nanotechnology has shown great promise in treating diverse diseases. However, developing nanomedicines that can cure autoimmune diseases without causing systemic immunosuppression is still quite challenging. Herein, we propose an all-in-one nanomedicine comprising an autoantigen peptide and CRISPR-Cas9 to restore specific immune tolerance by engineering dendritic cells (DCs) into a tolerogenic phenotype, which can expand autoantigen-specific regulatory T (Treg) cells. In brief, we utilized cationic lipid-assisted poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PEG-PLGA) nanoparticles to simultaneously encapsulate an autoimmune diabetes-relevant peptide (2.5mi), a CRISPR-Cas9 plasmid (pCas9), and three guide RNAs (gRNAs) targeting costimulatory molecules (CD80, CD86, and CD40). We demonstrated that the all-in-one nanomedicine was able to effectively codeliver these components into DCs, followed by simultaneous disruption of the three costimulatory molecules and presentation of the 2.5mi peptide on the genome-edited DCs. The resulting tolerogenic DCs triggered the generation and expansion of autoantigen-specific Treg cells by presenting the 2.5mi peptide to CD4+ T cells in the absence of costimulatory signals. Using autoimmune type 1 diabetes (T1D) as a typical disease model, we demonstrated that our nanomedicine prevented autoimmunity to islet components and inhibited T1D development. Our all-in-one nanomedicine achieved codelivery of CRISPR-Cas9 and the peptide to DCs and could be easily applied to other autoimmune diseases by substitution of different autoantigen peptides.


Assuntos
Autoantígenos/imunologia , Sistemas CRISPR-Cas/imunologia , Nanomedicina , Peptídeos/imunologia , Animais , Engenharia Celular , Células Cultivadas , Células Dendríticas , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos NOD , Tamanho da Partícula , Propriedades de Superfície
8.
Biomater Sci ; 8(23): 6683-6694, 2020 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-33089844

RESUMO

Studies have shown that the simultaneous regulation of tumor cell proliferation and the suppressive tumor immune microenvironment (TIME) could achieve better therapeutic effects. However, the targets of the proliferation and the TIME are different, which greatly limits the development of cancer therapy. A recent study found CD155, a highly expressed poliovirus receptor in melanoma cells and melanoma-infiltrating macrophages, functions as both an oncogene and immune checkpoint. Thus, it is supposed that targeting CD155 could bring dual therapeutic effects. Herein, we propose silencing the CD155 of melanoma cells and melanoma-infiltrating macrophages by a nanoparticle-delivered small interference RNA (siRNA) targeting CD155 (siCD155). We encapsulated siCD155 into cationic lipid-assisted nanoparticles (CLANsiCD155) and demonstrated that the intravenous injection of CLANsiCD155 could efficiently deliver siCD155 into melanoma cells and melanoma-infiltrating macrophages. The downregulation of CD155 in melanoma cells directly inhibited their proliferation, and meanwhile, the downregulation of CD155 in melanoma-infiltrating macrophages increased the activation of NK cells and T cells. Owing to this dual effect, CLANsiCD155 significantly inhibited the growth of B16-F10 melanoma. Our study suggests that nanoparticle-delivered siCD155 may be a simple but effective strategy for inhibiting tumor proliferation and reprogramming TIME.


Assuntos
Melanoma , Nanopartículas , RNA Interferente Pequeno , Receptores Virais , Neoplasias Cutâneas , Animais , Proliferação de Células , Melanoma/terapia , RNA Interferente Pequeno/genética , Neoplasias Cutâneas/terapia , Microambiente Tumoral
9.
Biomater Sci ; 6(6): 1592-1603, 2018 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-29725684

RESUMO

Chronic myeloid leukemia (CML), which is characterized by the Philadelphia translocation, which fuses breakpoint cluster region (BCR) sequences from chromosome 22 upstream of the Abelson murine leukemia viral oncogene homolog (ABL) on chromosome 9, requires specific and efficient treatment. The CRISPR/Cas9 system, with its mechanism of specific DNA complementary recognition by engineered guide RNA (gRNA), allows the development of novel therapeutics for CML. To achieve targeted therapy of CML with the CRISPR/Cas9 system, we encapsulated a CRISPR/Cas9 plasmid (pCas9) expressing gRNA targeting the overhanging fusion region of the BCR-ABL gene (pCas9/gBCR-ABL) with poly(ethylene glycol)-b-poly(lactic acid-co-glycolic acid) (PEG-PLGA)-based cationic lipid-assisted polymeric nanoparticles (CLANs), which specifically disrupted the CML-related BCR-ABL gene while sparing the BCR and ABL genes in normal cells. After intravenous injection, CLANs carrying pCas9/gBCR-ABL (CLANpCas9/gBCR-ABL) efficiently knocked out the BCR-ABL fusion gene of CML cells and improved the survival of a CML mouse model, indicating that the combination of the CRISPR/Cas9 system with nanocarriers is a promising strategy for targeted treatment of CML.


Assuntos
Sistemas CRISPR-Cas , Proteínas de Fusão bcr-abl/genética , Terapia Genética/métodos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Nanopartículas/química , Poliésteres/química , Polietilenoglicóis/química , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID
10.
Biomaterials ; 172: 92-104, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29723758

RESUMO

Inflammation is closely related to the development of many diseases and is commonly characterized by abnormal infiltration of immune cells, especially neutrophils. The current therapeutics of inflammatory diseases give little attention to direct modulation of these diseases with respect to immune cells. Nanoparticles are applied for efficient drug delivery into the disease-related immune cells, but their performance is significantly affected by their surface properties. In this study, to optimize the properties of nanoparticles for modulating neutrophils-related inflammation, we prepared a library of poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PEG-b-PLGA)-based cationic lipid-assisted nanoparticles (CLANs) with different surface PEG density and surface charge. Optimized CLANs for neutrophils targeting were screened in high-fat diet (HFD)-induced type 2 diabetes (T2D) mice. Then, a CRISPR-Cas9 plasmid expressing a guide RNA (gRNA) targeting neutrophil elastase (NE) was encapsulated into the optimized CLAN and denoted as CLANpCas9/gNE. After intravenous injection, CLANpCas9/gNE successfully disrupted the NE gene of neutrophils and mitigated the insulin resistance of T2D mice via reducing the inflammation in epididymal white adipose tissue (eWAT) and in the liver. This strategy provides an example of abating the inflammatory microenvironment by directly modulating immune cells with nanoparticles carrying genome editing tools.


Assuntos
Portadores de Fármacos/química , Inflamação/tratamento farmacológico , Lipídeos/química , Nanopartículas/química , Neutrófilos/metabolismo , Animais , Sequência de Bases , Sistemas CRISPR-Cas/genética , Microambiente Celular , Diabetes Mellitus Tipo 2/induzido quimicamente , Portadores de Fármacos/administração & dosagem , Liberação Controlada de Fármacos , Edição de Genes/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética/métodos , Resistência à Insulina/genética , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais , Terapia de Alvo Molecular/métodos , Nanopartículas/administração & dosagem , Tamanho da Partícula , Poliésteres/química , Polietilenoglicóis/química
11.
J Pharm Pharmacol ; 67(5): 703-13, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25627251

RESUMO

OBJECTIVES: Desferrioxamine (DFO), deferiprone (DFP) and deferasirox (DFX) are iron chelators currently in clinical use for the treatment of iron overload. Due to difficulties with administration and associated side effects with these three molecules, the search continues for an efficient nontoxic orally active iron chelator. This communication describes the properties of one such candidate, 1-(N-acetyl-6-aminohexyl)-3-hydroxy-2-methylpyridin-4-one (CM1). METHODS: Physicochemical characterisation techniques, including partition coefficient, pKa values and logK values for iron(III). Iron scavenging assays, from iron citrate, nontransferrin bound iron and iron-loaded rats. Cytotoxicity studies using white cells, hepatocytes and cardiomyocytes. KEY FINDINGS: CM1 possesses high affinity and selectivity for iron(III) and a suitable partition coefficient to permeate membranes. CM1 forms a neutral 3 : 1 iron(III) complex under physiological conditions and so, it is predicted to be capable of entry into mammalian cells to scavenge excess intracellular iron and to efflux from cells as the neutral 3 : 1 complex. CM1 is demonstrated to be orally active and to possess a higher efficacy than DFP in rats. CM1 displays no toxicity to a range of cell types. CONCLUSION: The above promising studies will be extended to monitor the pharmacokinetics and metabolism of CM1. CM1 is an excellent candidate for phase 1 clinical trials.


Assuntos
Acetamidas/química , Acetamidas/farmacologia , Quelantes de Ferro/química , Quelantes de Ferro/farmacologia , Ferro/metabolismo , Piridonas/química , Piridonas/farmacologia , Acetamidas/administração & dosagem , Acetamidas/efeitos adversos , Administração Oral , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ferro/sangue , Quelantes de Ferro/administração & dosagem , Quelantes de Ferro/efeitos adversos , Sobrecarga de Ferro/tratamento farmacológico , Fígado/efeitos dos fármacos , Fígado/metabolismo , Testes de Mutagenicidade , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Piridonas/administração & dosagem , Piridonas/efeitos adversos , Ratos , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA