Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther Oncolytics ; 31: 100753, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38144668
2.
Signal Transduct Target Ther ; 8(1): 418, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37919282

RESUMO

Smart nanoparticles, which can respond to biological cues or be guided by them, are emerging as a promising drug delivery platform for precise cancer treatment. The field of oncology, nanotechnology, and biomedicine has witnessed rapid progress, leading to innovative developments in smart nanoparticles for safer and more effective cancer therapy. In this review, we will highlight recent advancements in smart nanoparticles, including polymeric nanoparticles, dendrimers, micelles, liposomes, protein nanoparticles, cell membrane nanoparticles, mesoporous silica nanoparticles, gold nanoparticles, iron oxide nanoparticles, quantum dots, carbon nanotubes, black phosphorus, MOF nanoparticles, and others. We will focus on their classification, structures, synthesis, and intelligent features. These smart nanoparticles possess the ability to respond to various external and internal stimuli, such as enzymes, pH, temperature, optics, and magnetism, making them intelligent systems. Additionally, this review will explore the latest studies on tumor targeting by functionalizing the surfaces of smart nanoparticles with tumor-specific ligands like antibodies, peptides, transferrin, and folic acid. We will also summarize different types of drug delivery options, including small molecules, peptides, proteins, nucleic acids, and even living cells, for their potential use in cancer therapy. While the potential of smart nanoparticles is promising, we will also acknowledge the challenges and clinical prospects associated with their use. Finally, we will propose a blueprint that involves the use of artificial intelligence-powered nanoparticles in cancer treatment applications. By harnessing the potential of smart nanoparticles, this review aims to usher in a new era of precise and personalized cancer therapy, providing patients with individualized treatment options.


Assuntos
Nanopartículas Metálicas , Nanotubos de Carbono , Neoplasias , Humanos , Ouro/uso terapêutico , Inteligência Artificial , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Peptídeos
3.
Front Immunol ; 14: 1353708, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38179059

RESUMO

[This corrects the article DOI: 10.3389/fimmu.2021.771201.].

4.
Front Immunol ; 12: 771201, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899721

RESUMO

Although breast cancer has been previously considered "cold" tumors, numerous studies are currently conducted to explore the great potentials of immunotherapies in improving breast cancer patient outcomes. In addition to the focus on stimulating adaptive immunity for antitumor responses, growing evidence showed the importance of triggering host innate immunity to eradicate established tumors and/or control tumor metastasis of breast cancer. In this review, we first briefly introduce the breast tumor immune microenvironment. We also discuss innate immune targets and pathways and mechanisms of their synergy with the adaptive antitumor response and other treatment strategies. Lastly, we review clinical trials targeting innate immune pathways for breast cancer therapies.


Assuntos
Neoplasias da Mama/terapia , Imunidade Inata/imunologia , Imunoterapia/métodos , Microambiente Tumoral/imunologia , Imunidade Adaptativa/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Feminino , Humanos , Células Matadoras Naturais/imunologia , Linfócitos do Interstício Tumoral/imunologia , Células Supressoras Mieloides/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia
7.
J Orthop Surg Res ; 15(1): 120, 2020 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-32228637

RESUMO

OBJECTIVE: Matrix-associated autologous chondrocyte implantation (MACI) achieves good clinical efficacy in young patients with focal cartilage injury; however, phenotypic de-differentiation of chondrocytes cultured in monolayer and the treatment of older OA patients are still challenges in the field of cartilage tissue engineering. This study aimed to assess the in vitro re-differentiation potential and in vivo chondrogenic capacity of human OA chondrocytes inoculated into collagen I scaffolds with different cellular phenotypes and seeding densities. METHODS: OA chondrocytes and articular chondrocyte-laden scaffolds were cultured over 2 weeks in in vitro. Reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) and histological staining were used to detect the mRNA expression profiles and extracellular matrix secretion of chondrocyte-specific markers. OA chondrocyte-laden collagen I scaffolds with different cellular phenotypes, and seeding densities were implanted into SCID mice over 4 weeks to evaluate the chondrogenic capacity in vivo. RESULTS: Increased COL2a1, ACAN, COMP, SOX9, and BMP2 expression levels and decreased COL1a1, VCAN, MMP13, and ADAMTS5 amounts were observed in OA chondrocytes seeded in collagen I scaffolds; Implantation of phenotypically superior OA chondrocytes in collagen I scaffolds at high density could improve the chondrogenic capacity of human OA chondrocytes, as confirmed by RT-qPCR assessed gene expression patterns in vitro and histological evaluation in vivo. CONCLUSIONS: Freshly isolated chondrocytes from OA patients could be a source of replacement for articular chondrocytes being commonly used in MACI. Implantation of phenotypically superior OA chondrocytes in collagen I scaffolds at high density could be a promising tool for the treatment of elderly OA patients.


Assuntos
Condrócitos/fisiologia , Condrogênese/fisiologia , Colágeno Tipo I/administração & dosagem , Osteoartrite/patologia , Fenótipo , Alicerces Teciduais , Idoso , Animais , Contagem de Células/métodos , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos SCID , Pessoa de Meia-Idade
8.
J Orthop Surg Res ; 14(1): 425, 2019 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-31829201

RESUMO

BACKGROUND: The Cadherin-11 and PI3K/Akt pathway are increasingly recognized as the potential therapeutic target of osteoarthritis (OA) synovitis. The study aimed to investigate the role of PI3K/Akt signaling pathway in the expression of Cadherin-11 and migration and invasive capacity of fibroblast-like synoviocytes (FLS) of OA patients under stimulation of TNF-α and to explore the effect of the PI3K/Akt inhibitor and Cadherin-11 antibody in the therapy of the collagenase-induced osteoarthritis (CIOA) mice. METHODS: FLS were primarily cultured from synovium of osteoarthritic patients during total knee arthroplasty. Under the simulation of TNF-α, with or without PI3K/Akt inhibitor LY294002, Cadherin-11 expression was detected by real-time PCR and Western blot, as well as the migration and invasive capacity changes of OA FLS. Cadherin-11 antibody was injected intraarticularly or LY294002 was injected intraperitoneally in CIOA mice to evaluate the changes of synovitis score, cartilage damage, and Cadherin-11 expression. RESULTS: TNF-α stimulation increased Cadherin-11 expression at mRNA and protein level in OA FLS and also increased the phosphorylation-dependent activation of Akt. PI3K inhibitor LY294002 attenuated TNF-α-induced overexpression of Cadherin-11 and decreased the invasive capacity of OA FLS. Intraperitoneal injection of PI3K inhibitor LY294002 could decrease the Cadherin-11 protein expression in synovium of CIOA mice, although it has no significant inhibitory effect on synovitis and cartilage damage. Intraarticular injection of Cadherin-11 antibody attenuated the synovitis and cartilage damage in the CIOA joints and decreased Cadherin-11 expression in the synovial lining. CONCLUSIONS: PI3K/Akt pathway was associated with TNF-α-induced activation of OA FLS, which may involve in the pathogenesis of osteoarthritis. Anti-Cadherin-11 therapy in CIOA mice could attenuate the pathological changes of OA joints.


Assuntos
Cromonas/uso terapêutico , Morfolinas/uso terapêutico , Osteoartrite/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sinoviócitos/metabolismo , Fator de Necrose Tumoral alfa/toxicidade , Idoso , Animais , Movimento Celular , Células Cultivadas , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Morfolinas/farmacologia , Osteoartrite/induzido quimicamente , Osteoartrite/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Sinoviócitos/efeitos dos fármacos
9.
BMC Musculoskelet Disord ; 19(1): 163, 2018 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-29793466

RESUMO

BACKGROUND: Angioleiomyoma is a very rare benign solitary soft tissue neoplasm originating from smooth muscle layer of blood vessels. The tumor is usually located in the subcutis or the superficial fasciae, but less often in the deep fasciae, especially rare in the knee joint cavity. Diagnosis is frequently delayed or misdiagnosed as loose body or anterior knee pain because of its rare occurrence and poor awareness of physicians. Few studies have presented intra-articular angioleiomyoma and such cases become rarer and more difficult to diagnose when it presents as loose body. CASE PRESENTATION: Two patients, a middle-aged man and an old woman, presented to our outpatient clinic with persistent anterior knee pain and both of them suffered from a solitary mass in the right knee that had slowly enlarged. One of two patients showed negative in the routine radiographic imaging and the other showed a "loose body" beside the lateral femoral condyle in the knee. MRI showed both a well-demarcated intra-articular mass of isointense signal to muscle on T1-weighted images and heterogeneous intensity on T2-weighted images. Their tumors were excised under arthroscopy finally, with the pathological results revealed vascular leiomyomas. They both recovered well with pain free after operation and no signs of recurrence were seen at the 7-year follow-up. CONCLUSIONS: This case report illustrates the atypical locations of angioleiomyoma in the knee joint should arouse our attention and be included in the differential diagnosis of nodular lesions mimicking loose bodies.


Assuntos
Angiomioma/cirurgia , Artroscopia/métodos , Corpos Livres Articulares/cirurgia , Articulação do Joelho/cirurgia , Neoplasias de Tecidos Moles/cirurgia , Adulto , Idoso , Angiomioma/diagnóstico por imagem , Diagnóstico Diferencial , Feminino , Humanos , Corpos Livres Articulares/diagnóstico por imagem , Articulação do Joelho/diagnóstico por imagem , Masculino , Neoplasias de Tecidos Moles/diagnóstico por imagem , Resultado do Tratamento
10.
Adv Mater ; 30(22): e1707112, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29656492

RESUMO

Cancer cells resist to the host immune antitumor response via multiple suppressive mechanisms, including the overexpression of PD-L1 that exhausts antigen-specific CD8+ T cells through PD-1 receptors. Checkpoint blockade antibodies against PD-1 or PD-L1 have shown unprecedented clinical responses. However, limited host response rate underlines the need to develop alternative engineering approaches. Here, engineered cellular nanovesicles (NVs) presenting PD-1 receptors on their membranes, which enhance antitumor responses by disrupting the PD-1/PD-L1 immune inhibitory axis, are reported. PD-1 NVs exhibit a long circulation and can bind to the PD-L1 on melanoma cancer cells. Furthermore, 1-methyl-tryptophan, an inhibitor of indoleamine 2,3-dioxygenase can be loaded into the PD-1 NVs to synergistically disrupt another immune tolerance pathway in the tumor microenvironment. Additionally, PD-1 NVs remarkably increase the density of CD8+ tumor infiltrating lymphocytes in the tumor margin, which directly drive tumor regression.


Assuntos
Neoplasias/terapia , Humanos , Imunoterapia , Linfócitos do Interstício Tumoral , Receptor de Morte Celular Programada 1 , Linfócitos T , Microambiente Tumoral
11.
Adv Drug Deliv Rev ; 127: 106-118, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29408182

RESUMO

The intrinsic properties of therapeutic proteins generally present a major impediment for transdermal delivery, including their relatively large molecule size and susceptibility to degradation. One solution is to utilize microneedles (MNs), which are capable of painlessly traversing the stratum corneum and directly translocating protein drugs into the systematic circulation. MNs can be designed to incorporate appropriate structural materials as well as therapeutics or formulations with tailored physicochemical properties. This platform technique has been applied to deliver drugs both locally and systemically in applications ranging from vaccination to diabetes and cancer therapy. This review surveys the current design and use of polymeric MNs for transdermal protein delivery. The clinical potential and future translation of MNs are also discussed.


Assuntos
Sistemas de Liberação de Medicamentos , Microinjeções , Agulhas , Polímeros , Proteínas/administração & dosagem , Proteínas/farmacocinética , Pele/metabolismo , Administração Cutânea , Humanos , Polímeros/química , Proteínas/metabolismo
12.
Sci Transl Med ; 10(429)2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29467299

RESUMO

Patients with low-immunogenic tumors respond poorly to immune checkpoint blockade (ICB) targeting the programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway. Conversely, patients responding to ICB can experience various side effects. We have thus engineered a therapeutic scaffold that, when formed in situ, allows the local release of gemcitabine (GEM) and an anti-PD-L1 blocking antibody (aPDL1) with distinct release kinetics. The scaffold consists of reactive oxygen species (ROS)-degradable hydrogel that releases therapeutics in a programmed manner within the tumor microenvironment (TME), which contains abundant ROS. We found that the aPDL1-GEM scaffold elicits an immunogenic tumor phenotype and promotes an immune-mediated tumor regression in the tumor-bearing mice, with prevention of tumor recurrence after primary resection.


Assuntos
Desoxicitidina/análogos & derivados , Animais , Linhagem Celular Tumoral , Terapia Combinada , Desoxicitidina/uso terapêutico , Feminino , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/metabolismo , Gencitabina
13.
Nat Biomed Eng ; 2(11): 831-840, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-31015615

RESUMO

Patients with acute myeloid leukaemia who relapse following therapy have few treatment options and face poor outcomes. Immune checkpoint inhibition, for example, by antibody-mediated programmed death-1 (PD-1) blockade, is a potent therapeutic modality that improves treatment outcomes in acute myeloid leukaemia. Here, we show that systemically delivered blood platelets decorated with anti-PD-1 antibodies (aPD-1) and conjugated to haematopoietic stem cells (HSCs) suppress the growth and recurrence of leukaemia in mice. Following intravenous injection into mice bearing leukaemia cells, the HSC-platelet-aPD-1 conjugate migrated to the bone marrow and locally released aPD-1, significantly enhancing anti-leukaemia immune responses, and increasing the number of active T cells, production of cytokines and chemokines, and survival time of the mice. This cellular conjugate also promoted resistance to re-challenge with leukaemia cells. Taking advantage of the homing capability of HSCs and in situ activation of platelets for the enhanced delivery of a checkpoint inhibitor, this cellular combination-mediated drug delivery strategy can significantly augment the therapeutic efficacy of checkpoint blockade.


Assuntos
Anticorpos/imunologia , Plaquetas/metabolismo , Transplante de Células-Tronco Hematopoéticas , Leucemia/terapia , Receptor de Morte Celular Programada 1/imunologia , Animais , Anticorpos/química , Plaquetas/química , Plaquetas/citologia , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Células-Tronco Hematopoéticas/química , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Morte Celular Programada 1/deficiência , Receptor de Morte Celular Programada 1/genética , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
14.
Sci Immunol ; 2(17)2017 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-29127106

RESUMO

Melanin is capable of transforming 99.9% of the absorbed sunlight energy into heat, reducing the risk of skin cancer. We here develop a melanin-mediated cancer immunotherapy strategy through a transdermal microneedle patch. B16F10 whole tumor lysate containing melanin is loaded into polymeric microneedles that allow sustained release of the lysate upon insertion into the skin. In combination with the near-infrared light irradiation, melanin in the patch mediates the generation of heat, which further promotes tumor-antigen uptake by dendritic cells, and leads to enhanced antitumor vaccination. We found that the spatiotemporal photoresponsive immunotherapy increases infiltration of polarized T cells and local cytokine release. These immunological effects increase the survival of mice after tumor challenge and elicited antitumor effects toward established primary tumor and distant tumor. Collectively, melanin generates local heat, boosts T cell activities by transdermal vaccines, and promotes antitumor immune responses.


Assuntos
Imunoterapia/métodos , Raios Infravermelhos , Melaninas/imunologia , Neoplasias Experimentais/terapia , Animais , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/metabolismo , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Imunoterapia/instrumentação , Estimativa de Kaplan-Meier , Melaninas/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Agulhas , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Adesivo Transdérmico , Carga Tumoral/imunologia
15.
Theranostics ; 7(14): 3504-3516, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28912891

RESUMO

The immune system protects the body against a wide range of infectious diseases and cancer by leveraging the efficiency of immune cells and lymphoid organs. Over the past decade, immune cell/organ therapies based on the manipulation, infusion, and implantation of autologous or allogeneic immune cells/organs into patients have been widely tested and have made great progress in clinical applications. Despite these advances, therapy with natural immune cells or lymphoid organs is relatively expensive and time-consuming. Alternatively, biomimetic materials and strategies have been applied to develop artificial immune cells and lymphoid organs, which have attracted considerable attentions. In this review, we survey the latest studies on engineering biomimetic materials for immunotherapy, focusing on the perspectives of bioengineering artificial antigen presenting cells and lymphoid organs. The opportunities and challenges of this field are also discussed.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Artificiais/imunologia , Tecido Linfoide/imunologia , Animais , Células Artificiais/química , Materiais Biomiméticos/química , Humanos , Tecido Linfoide/citologia
16.
Adv Mater ; 29(29)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28556553

RESUMO

Cancer immunotherapy, as a paradigm shift in cancer treatment, has recently received tremendous attention. The active cancer vaccination, immune checkpoint blockage (ICB) and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer are among these developments that have achieved a significant increase in patient survival in clinical trials. Despite these advancements, emerging research at the interdisciplinary interface of cancer biology, immunology, bioengineering, and materials science is important to further enhance the therapeutic benefits and reduce side effects. Here, an overview of the latest studies on engineering biomaterials for the enhancement of anticancer immunity is given, including the perspectives of delivery of immunomodulatory therapeutics, engineering immune cells, and constructing immune-modulating scaffolds. The opportunities and challenges in this field are also discussed.


Assuntos
Materiais Biocompatíveis/química , Imunoterapia , Neoplasias/terapia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos/genética , Antígenos/imunologia , Antígenos/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Fatores Imunológicos/metabolismo , Fatores Imunológicos/uso terapêutico , Nanoestruturas/química
17.
Nanoscale ; 9(1): 150-163, 2017 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-27910983

RESUMO

Cancer cells use autophagy to resist poor survival environmental conditions such as low PH, poor nutrients as well as chemical therapy. Nanogels have been used as efficient chemical drug carriers for cancer treatment. However, the effect of nanogels on autophagy is still unknown. Here, we used Rab proteins as the marker of multiple trafficking vesicles in endocytosis and LC3 as the marker of autophagy to investigate the intracellular trafficking network of Rhodamine B (Rho)-labeled nanogels. The nanogels were internalized by the cells through multiple protein dependent endocytosis and micropinocytosis. After inception by the cells, the nanogels were transported into multiple Rab positive vesicles including early endosomes (EEs), late endosomes (LEs), recycling endosomes (REs) and lipid droplets. Finally, these Rab positive vesicles were transported to lysosome. In addition, GLUT4 exocytosis vesicles could transport the nanogels out of the cells. Moreover, nanogels could induce autophagy and be sequestered in autophagosomes. The crosstalk between autophagosomes and Rab positive vesicles were investigated, we found that autophagosomes may receive nanogels through multiple Rab positive vesicles. Co-delivery of autophagy inhibitors such as chloroquine (CQ) and the chemotherapeutic drug doxorubicin (DOX) by nanogels blocked the autophagy induced by DOX greatly decreasing both of the volume and weight of the tumors in mice tumor models. Investigation and intervention of the autophagy pathway could provide a new method to improve the therapeutic effect of anticancer nanogels.


Assuntos
Autofagia , Sistemas de Liberação de Medicamentos , Endocitose , Géis , Nanopartículas , Neoplasias Experimentais/tratamento farmacológico , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Cloroquina/administração & dosagem , Doxorrubicina/administração & dosagem , Endossomos , Exocitose , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos SCID , Rodaminas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Hum Vaccin Immunother ; 13(1): 245-248, 2017 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-27668759

RESUMO

Immune checkpoint inhibitors (ICI) based cancer immunotherapy has recently attracted considerable interest in the field of cancer therapy. The relevant immunotherapeutic agents do not directly attack the tumor, but boost the body's immune system to recognize and kill cancer cells. In this commentary, recent efforts utilizing immunoengineering for local delivery of these immune checkpoint antibodies are introduced. Future opportunities and challenges in this research theme are also commented.


Assuntos
Anticorpos/administração & dosagem , Fatores Imunológicos/administração & dosagem , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Pesquisa Biomédica/tendências , Humanos , Terapia de Alvo Molecular/métodos
19.
ACS Nano ; 10(9): 8956-63, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27599066

RESUMO

Despite the promising efficacy of immunoregulation in cancer therapy, the clinical benefit has been restricted by inefficient infiltration of lymphocytes in the evolution of immune evasion. Also, immune-related adverse events have often occurred due to the off-target binding of therapeutics to normal tissues after systematic treatment. In light of this, we have developed a synergistic immunotherapy strategy that locally targets the immunoinhibitory receptor programmed cell death protein 1 (PD1) and immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO) for the treatment of melanoma through a microneedle-based transcutaneous delivery approach. The embedded immunotherapeutic nanocapsule loaded with anti-PD1 antibody (aPD1) is assembled from hyaluronic acid modified with 1-methyl-dl-tryptophan (1-MT), an inhibitor of IDO. This formulation method based on the combination strategy of "drug A in carriers formed by incorporation of drug B" facilitates the loading capacity of therapeutics. Moreover, the resulting delivery device elicits the sustained release and enhances retention of checkpoint inhibitors in the tumor microenvironment. Using a B16F10 mouse melanoma model, we demonstrate that this synergistic treatment has achieved potent antitumor efficacy, which is accompanied by enhanced effective T cell immunity as well as reduced immunosuppression in the local site.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Imunoterapia/métodos , Melanoma/terapia , Neoplasias Cutâneas/terapia , Animais , Terapia de Imunossupressão , Indolamina-Pirrol 2,3,-Dioxigenase/uso terapêutico , Receptor de Morte Celular Programada 1/uso terapêutico , Microambiente Tumoral
20.
Nanoscale ; 8(17): 9178-84, 2016 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-27074960

RESUMO

Drug delivery systems inspired by natural particulates hold great promise for targeted cancer therapy. An endosome formed by internalization of plasma membrane has a massive amount of membrane proteins and receptors on the surface, which is able to specifically target the homotypic cells. Herein, we describe a simple method to fabricate an internalized compartments encapsulated nanogel with endosome membrane components (EM-NG) from source cancer cells. Following intracellular uptake of methacrylated hyaluronic acid (m-HA) adsorbed SiO2/Fe3O4 nanoparticles encapsulating a crosslinker and a photoinitiator, EM-NG was readily prepared through in situ crosslinking initiated under UV irradiation after internalization. The resulting nanogels loaded with doxorubicin (DOX) displayed enhanced internalization efficiency to the source cells through a specific homotypic affinity in vitro. However, when treated with the non-source cells, the EM-NGs exhibited insignificant difference in therapeutic efficiency compared to a bare HA nanogel with DOX. This study illustrates the potential of utilizing an internalized compartments encapsulated formulation for targeted cancer therapy, and offers guidelines for developing a natural particulate-inspired drug delivery system.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas , Doxorrubicina/administração & dosagem , Compostos Ferrosos , Ácido Hialurônico , Dióxido de Silício
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA