Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Pharm ; 21(8): 3743-3763, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38953708

RESUMO

The coronavirus (COVID-19) pandemic has underscored the critical role of mRNA-based vaccines as powerful, adaptable, readily manufacturable, and safe methodologies for prophylaxis. mRNA-based treatments are emerging as a hopeful avenue for a plethora of conditions, encompassing infectious diseases, cancer, autoimmune diseases, genetic diseases, and rare disorders. Nonetheless, the in vivo delivery of mRNA faces challenges due to its instability, suboptimal delivery, and potential for triggering undesired immune reactions. In this context, the development of effective drug delivery systems, particularly nanoparticles (NPs), is paramount. Tailored with biophysical and chemical properties and susceptible to surface customization, these NPs have demonstrated enhanced mRNA delivery in vivo and led to the approval of several NPs-based formulations for clinical use. Despite these advancements, the necessity for developing a refined, targeted NP delivery system remains imperative. This review comprehensively surveys the biological, translational, and clinical progress in NPs-mediated mRNA therapeutics for both the prevention and treatment of diverse diseases. By addressing critical factors for enhancing existing methodologies, it aims to inform the future development of precise and efficacious mRNA-based therapeutic interventions.


Assuntos
COVID-19 , Sistemas de Liberação de Fármacos por Nanopartículas , RNA Mensageiro , Humanos , RNA Mensageiro/genética , RNA Mensageiro/administração & dosagem , Sistemas de Liberação de Fármacos por Nanopartículas/química , COVID-19/prevenção & controle , Nanopartículas/química , Sistemas de Liberação de Medicamentos/métodos , Animais , SARS-CoV-2/efeitos dos fármacos , Vacinas de mRNA
2.
Mater Today Bio ; 27: 101131, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39050986

RESUMO

The limitations of platinum in ovarian cancer therapy, such as poor solubility and significant side effects, often lead to suboptimal therapeutic outcome and mortality. In this study, we have developed a novel approach utilizing biodegradable polymeric nanoparticles as a drug delivery system (NDDS), loaded with advanced platinum (IV) (Pt(IV)) prodrugs. A key feature of our approach is the enhancement of nanoparticles with maleimide, a modification hypothesized to significantly boost tumor tissue accumulation. When tested in mouse models of orthotopic and peritoneal metastasis ovarian cancer, these maleimide-modified nanoparticles are anticipated to show preferential accumulation in tumor tissues, enhancing therapeutic efficiency and minimizing systemic drug exposure. Our findings demonstrate that the maleimide-modified Pt(IV)-loaded NDDSs significantly reduce tumor burden in comparison to traditional cisplatin therapy, while simultaneously reducing adverse side effects. This leads to markedly improved survival rates in models of peritoneal metastasis ovarian cancer, offering a promising new direction in the treatment of this challenging disease.

3.
Biomed Pharmacother ; 177: 117011, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38917758

RESUMO

Microglia are essential for maintaining homeostasis and responding to pathological events in the central nervous system (CNS). Their dynamic and multidimensional states in different environments are pivotal factors in various CNS disorders. However, therapeutic modulation of microglial states is challenging due to the intricate balance these cells maintain in the CNS environment and the blood-brain barrier's restriction of drug delivery. Nanomedicine presents a promising avenue for addressing these challenges, offering a method for the targeted and efficient modulation of microglial states. This review covers the challenges faced in microglial therapeutic modulation and potential use of nanoparticle-based drug delivery systems. We provide an in-depth examination of nanoparticle applications for modulating microglial states in a range of CNS disorders, encompassing neurodegenerative and autoimmune diseases, infections, traumatic injuries, stroke, tumors, chronic pain, and psychiatric conditions. This review highlights the recent advancements and future prospects in nanomedicine for microglial modulation, paving the way for future research and clinical applications of therapeutic interventions in CNS disorders.


Assuntos
Doenças do Sistema Nervoso Central , Microglia , Nanomedicina , Humanos , Microglia/efeitos dos fármacos , Nanomedicina/métodos , Doenças do Sistema Nervoso Central/tratamento farmacológico , Animais , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo
4.
Int J Biol Macromol ; 268(Pt 1): 131679, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38641274

RESUMO

Over the past few decades, significant strides have been made in understanding the pivotal roles that chemokine networks play in tumor biology. These networks, comprising chemokines and their receptors, wield substantial influence over cancer immune regulation and therapeutic outcomes. As a result, targeting these chemokine systems has emerged as a promising avenue for cancer immunotherapy. However, therapies targeting chemokines face significant challenges in solid tumor treatment, due to the complex and fragile of the chemokine networks. A nuanced comprehension of the complicacy and functions of chemokine networks, and their impact on the tumor microenvironment, is essential for optimizing their therapeutic utility in oncology. This review elucidates the ways in which chemokine networks interact with cancer immunity and tumorigenesis. We particularly elaborate on recent innovations in manipulating these networks for cancer treatment. The review also highlights future challenges and explores potential biomaterial strategies for clinical applications.


Assuntos
Quimiocinas , Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Quimiocinas/metabolismo , Animais , Imunoterapia/métodos , Portadores de Fármacos/química , Nanopartículas/química , Nanopartículas/uso terapêutico , Receptores de Quimiocinas/metabolismo
5.
Biomater Sci ; 12(9): 2381-2393, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38500446

RESUMO

The inability to systemic administration of nanoparticles, particularly cationic nanoparticles, has been a significant barrier to their clinical translation due to toxicity concerns. Understanding the in vivo behavior of cationic lipids is crucial, given their potential impact on critical biological components such as immune cells and hematopoietic stem cells (HSC). These cells are essential for maintaining the body's homeostasis, and their interaction with cationic lipids is a key factor in determining the safety and efficacy of these nanoparticles. In this study, we focused on the cytotoxic effects of cationic lipid/DNA complexes (CLN/DNA). Significantly, we observed that the most substantial cytotoxic effects, including a marked increase in numbers of long-term hematopoietic stem cells (LT-HSC), occurred 24 h post-CLN/DNA treatment in mice. Furthermore, we found that CLN/DNA-induced HSC expansion in bone marrow (BM) led to a notable decrease in the ability to reestablish blood cell production. Our study provides crucial insights into the interaction between cationic lipids and vital cellular components of the immune and hematopoietic systems.


Assuntos
Cátions , DNA , Células-Tronco Hematopoéticas , Lipídeos , Animais , DNA/química , DNA/administração & dosagem , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Cátions/química , Lipídeos/química , Nanopartículas/química , Nanopartículas/administração & dosagem , Camundongos Endogâmicos C57BL
6.
Sci Bull (Beijing) ; 69(7): 922-932, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38331707

RESUMO

Neoantigen cancer vaccines have been envisioned as one of the most promising means for cancer therapies. However, identifying neoantigens for tumor types with low tumor mutation burdens continues to limit the effectiveness of neoantigen vaccines. Herein, we proposed a "hit-and-run" vaccine strategy which primes T cells to attack tumor cells decorated with exogenous "neo-antigens". This vaccine strategy utilizes a peptide nanovaccine to elicit antigen-specific T cell responses after tumor-specific decoration with a nanocarrier containing the same peptide antigens. We demonstrated that a poly(2-oxazoline)s (POx) conjugated with OVA257-264 peptide through a matrix metalloprotease 2 (MMP-2) sensitive linker could efficiently and selectively decorate tumor cells with OVA peptides in vivo. Then, a POx-based nanovaccine containing OVA257-264 peptides to elicit OVA-specific T cell responses was designed. In combination with this hit-and-run vaccine system, an effective vaccine therapy was demonstrated across tumor types even without OVA antigen expression. This approach provides a promising and uniform vaccine strategy against tumors with a low tumor mutation burden.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Epitopos , Antígenos de Neoplasias , Neoplasias/terapia , Peptídeos
7.
Adv Mater ; 35(47): e2309667, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37807931

RESUMO

T cells play a basic and key role in immunotherapy against solid tumors, and efficiently recruiting them into neoplastic foci and sustaining long-term effector function are consistent goals that remain a critical challenge. Here, an injectable alginate-based hydrogel with abundant ß-cyclodextrin (ALG-ßCD) sites is developed and intratumorally injected to recruit CCR9+ CD8+ T cells (a subset of T cells with robust antitumor activity) via the trapped chemokine CCL25. In the meantime, an intravenously injected adamantane-decorated anti-PD1 antibody (Ad-aPD1) would hitchhike on recruited CCR9+ CD8+ T cells to achieve the improved intratumoral accumulation of Ad-aPD1. Moreover, the Ad-PD1 and Ad-PDL1 antibodies are immobilized in the ALG-ßCD hydrogel through supramolecular host-guest interactions of Ad and ßCD, which facilitate engagement between CD8+ T cells and tumor cells and reinvigorate CD8+ T cells to avoid exhaustion. Based on this treatment strategy, T cell-mediated anticancer activity is promoted at multiple levels, eventually achieving superior antitumor efficacy in both orthotopic and postsurgical B16-F10 tumor models.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Humanos , Hidrogéis/metabolismo , Imunoterapia , Neoplasias/terapia , Neoplasias/metabolismo
8.
Biomaterials ; 300: 122187, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37302279

RESUMO

Radiotherapy (IR) is capable of enhancing antitumor immune responses. However, IR treatment also aggravates the infiltration of peripheral macrophages into the tumor, resulting in reversing the therapeutic effects of antitumor immunity. Thus, a strategy to effectively prevent tumor infiltration by macrophages may further improved the therapeutic efficacy of radiotherapy. Herein, we found that PEGylated solid lipid nanoparticles with maleimide as PEG end-group (SLN-PEG-Mal) show significantly enhanced adsorption onto RBCs through reacting with reactive sulfhydryl groups on RBCs' surface both in vitro and in vivo, and caused significant changes in the surface properties and morphology of RBCs. These RBCs adsorbed by SLN-PEG-Mal were rapidly removed from circulation due to efficient engulfment by reticuloendothelial macrophages, supporting the usefulness of SLN-PEG-Mal for macrophage-targeted drug delivery. While lacking the use of radioisotope tracing (considered the gold standard for PK/BD studies), our data align with the expected pathway of host defense activation through surface-loaded RBCs. Importantly, injection of paclitaxel-loaded SLN-PEG-Mal effectively inhibited the tumor-infiltration by macrophages, and significantly improved the antitumor immune responses in tumor-bearing mice treated with low-dose irradiation. This study provides insights into the effects of maleimide as PEG end-group on enhancing the interaction between PEGylated nanoparticles and RBCs and offers an effective strategy to inhibit tumor infiltration by circulating macrophages.


Assuntos
Nanopartículas , Neoplasias , Camundongos , Animais , Polietilenoglicóis/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Eritrócitos , Nanopartículas/uso terapêutico , Macrófagos , Maleimidas
9.
APL Bioeng ; 7(1): 016116, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36968454

RESUMO

To improve the biosafety of the nanodelivery system, this study developed novel monodisperse spherical aluminum nanoparticles (Al NPs) and evaluated their cytotoxicity in vitro and distribution and biotoxicity in vivo. Compared with gold nanoparticles of the same size, Al NPs not only had low cytotoxicity in vitro but also did not cause accumulation in major organs in vivo after intravenous injections. No significant abnormalities were observed in the serum biochemical indices of mice injected with Al NPs. Additionally, no substantial changes occurred in the histopathology of major organs, and no apparent biological toxicity was measured after consecutive injections of Al NPs. These results indicate that Al NPs have a good biological safety and provide a new method for developing low-toxicity nanomedicine.

10.
Sci Adv ; 8(51): eabq3699, 2022 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-36542700

RESUMO

CD40 is an important costimulatory molecule expressed on antigen-presenting cells (APCs) and plays a critical role for APC activation, offering a promising therapeutic target for preventing allograft rejection. Here, we developed a biodegradable nanoparticle small interfering RNA delivery system (siCD40/NPs) to effectively deliver CD40 siRNA (siCD40) into hematopoietic stem cells (HSCs), myeloid progenitors, and mature dendritic cells (DCs) and macrophages. Injection of siCD40/NPs not only down-regulated CD40 expression in DCs and macrophages but also inhibited the differentiation of HSCs and/or myeloid progenitors into functional DCs and macrophages. Furthermore, siCD40/NPs treatment significantly prolonged allograft survival in mouse models of skin allotransplantation. In addition to reiteration of the role of CD40 in APC activation, our findings highlight a previously unappreciated role of CD40 in DC and macrophage differentiation from their progenitors. Furthermore, our results support the effectiveness of siCD40/NPs in suppressing alloimmune responses, providing a potential means of facilitating tolerance induction and preventing allotransplant rejection.

11.
Front Bioeng Biotechnol ; 10: 1007151, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36213072

RESUMO

Nanoparticles have been investigated as drug carriers and promising agents for cancer therapy. However, the tumor microenvironment (TME), which is formed by the tumor, is considered a barrier for nanocarriers to enter the internal tumor tissue. Therefore, the evaluation of the biological distribution of nanocarriers in TME can provide useful information on their role in tumor-targeted drug delivery. Although the tumor-bearing mouse model is commonly used to investigate the distribution of nanocarriers in the TME, there is currently a lack of a testing system to predict the distribution of nanocarriers in tumor tissues, especially in patients. This study revealed that the macrophages and dendritic cells (DCs) were more distributed in the peripheral part than the central part of the tumor, which might be an obstacle to the uniform distribution of nanoparticles in the tumor. In addition, the cellular uptake of gold nanoparticles (AuNR and AuNS) in macrophages and DCs cell lines (RAW264.7 and DC1.2) was markedly different from that in the TME. Hence, the study model of the interaction between nanoparticles and macrophages and DCs has an important impact on the accuracy of the results. The vibratome sections of tumor tissues preserved the spatial distribution of immune cells and tumor cells, and had very little effects on their morphologies and activities. More importantly, we found that the distribution of nanocarriers in vibratome sections was similar to that in tumors in vivo. In all, ex vivo analysis using vibratome sections of tumor tissues provides a more convenient and stable method for elucidating the influences of TME on the distribution of nanocarriers.

12.
Biomater Sci ; 10(23): 6755-6767, 2022 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-36301154

RESUMO

Cancer immunotherapy using immune checkpoint blockade has become an attractive treatment option for patients with different cancers. JQ1, an indirect inhibitor of MYC, enhances antitumor immune responses by regulating the expression of programmed death-ligand 1 (PD-L1) and cluster of differentiation 47 (CD47) in tumor cells; however, its role in downregulating the expression of CD47 remains elusive. The present study revealed that JQ1 failed to downregulate and, when used at high concentrations, it unexpectedly upregulated the expression of CD47 in murine B16F10 melanoma and 4T1 breast tumor cells. Hence, the combinatory use of JQ1 and CD47-specific short interfering RNA (siRNA) may lead to an improved antitumor effect. To overcome the poor water solubility of JQ1 and enhance tumor-targeted delivery, cationic lipid nanoparticles (CLNs) encapsulating both JQ1 and siCD47 simultaneously (CLN/JQ1/siCD47) or each individually (CLN/JQ1/siNC or CLN/siCD47) were prepared. CLN/JQ1/siCD47, but not CLN/JQ1/siNC or CLN/siCD47, simultaneously downregulated both PD-L1 and CD47 in vitro and in vivo. Furthermore, compared with CLN/JQ1/siNC and CLN/siCD47, CLN/JQ1/siCD47 induced a significantly enhanced antitumor effect in mice with established breast cancer. The results of this study highlight a synergistic effect of simultaneous PD-L1 and CD47 downregulation and provide a novel strategy for improving the antitumor effects of JQ1.


Assuntos
Antígeno B7-H1 , Neoplasias , Camundongos , Animais , RNA Interferente Pequeno/genética , Antígeno CD47/genética , Regulação para Baixo , Imunoterapia/métodos , Fatores Imunológicos , Lipídeos
13.
Biomaterials ; 287: 121645, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35779480

RESUMO

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor with a high mortality rate. Immunotherapy has achieved promising clinical results in multiple cancers, but shows unsatisfactory outcome in GBM patients, and poor drug delivery across the blood-brain barrier (BBB) is believed to be one of the main limitations that hinder the therapeutic efficacy of drugs. Herein, a new cationic lipid nanoparticle (LNP) that can efficiently deliver siRNA across BBB and target mouse brain is prepared for modulating the tumor microenvironment for GBM immunotherapy. By designing and screening cationic LNPs with different ionizable amine headgroups, a lipid (named as BAMPA-O16B) is identified with an optimal acid dissociation constant (pKa) that significantly enhances the cellular uptake and endosomal escape of siRNA lipoplex in mouse GBM cells. Importantly, BAMPA-O16B/siRNA lipoplex is highly effective to deliver siRNA against CD47 and PD-L1 across the BBB into cranial GBM in mice, and downregulate target gene expression in the tumor, resulting in synergistically activating a T cell-dependent antitumor immunity in orthotopic GBM. Collectively, this study offers an effective strategy for brain targeted siRNA delivery and gene silencing by optimizing the physicochemical property of LNPs. The effectiveness of modulating immune environment of GBM could further be expanded for potential treatment of other brain tumors.

14.
Front Bioeng Biotechnol ; 10: 873369, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35419357

RESUMO

Malignant tumors pose a serious threat to human health and have high fatality rates. Conventional clinical anti-tumor treatment is mainly based on traditional surgery, chemotherapy, radiotherapy, and interventional therapy, and even though these treatment methods are constantly updated, a satisfactory efficacy is yet to be obtained. Therefore, research on novel cancer treatments is being actively pursued. We review the classification of gene therapies of malignant tumors and their advantages, as well as the development of gene editing techniques. We further reveal the nano-drug delivery carrier effect in improving the efficiency of gene editing. Finally, we summarize the progress in recent years of gene editing techniques based on nano-drug delivery carriers in the treatment of various malignant tumors, and analyze the prospects of the technique and its restricting factors.

15.
Adv Mater ; 34(21): e2200449, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35291052

RESUMO

Various macro/microscopic biomaterials have been developed for controlled drug delivery in the combination therapy of malignancies. However, uncertain loading ratio, release sequence, and spatiotemporal distribution of drugs hinder their synergistic therapeutic effects and clinical applications. In this work, a tumor microenvironments-adapted composite consisting of a thermosensitive hydrogel and a reactive oxygen species (ROS)-responsive nanogel is developed for precisely sequential drug release to enhance molecularly targeted therapy and amplify immune activation. LY3200882 (LY), a selective transforming growth factor-ß (TGF-ß) inhibitor, is encapsulated in the ROS-responsive nanogel and dispersed uniformly with regorafenib (REG) in a thermosensitive hydrogel (Gel/(REG+NG/LY)). After in situ administration, REG is preferentially released from the hydrogel to inhibit tumor growth and promote ROS generation, which triggers the subsequent on-demand release of LY from the nanogel. LY contributes to preventing the epithelial-mesenchymal transition and immune escape of tumor cells induced by elevated TGF-ß. In subcutaneous and orthotopic colorectal tumor bearing mouse models, Gel/(REG+NG/LY) effectively inhibits tumor growth and liver metastases by increasing the tumor infiltration of CD8+ T cells, reducing the recruitment of tumor-associated macrophages and myeloid-derived suppressor cells, and promoting the polarization of macrophages from M2 to M1 type, indicating the significant potential in improving the prognosis of advanced cancer patients.


Assuntos
Antineoplásicos , Hidrogéis , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Humanos , Camundongos , Nanogéis , Peptídeos , Espécies Reativas de Oxigênio , Fator de Crescimento Transformador beta , Microambiente Tumoral
16.
Macromol Biosci ; 21(9): e2100171, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34169661

RESUMO

Cancer immunotherapy is to artificially stimulate the immune system against tumor cells. Effectively increasing the immunogenicity of dying tumor cells has great potential to stimulate the anticancer immune responses. Recently, a synthetic cationic anticancer polypeptide (ACPP) is prepared, which mimics the host defense peptides, to effectively inhibit tumor growth by directly inducing rapid necrosis of cancer cells through a membrane-lytic mechanism. Thus, this ACPP has the potential ability to induce immunogenic cancer cell death (ICD) and promote antitumor immunity. Herein, it is reported that ACPP successfully induces ICD in mouse colon cancer cells, resulting in effectively promoting T-cell-dependent antitumor immune responses by enhanced activation of dendritic cells. Interestingly, the level of natural killer cells, which are another kind of antitumor effector cell, in tumor microenvironment is also significantly increased by ACPP. The ratio of M1/M2 tumor-associated macrophages is further obviously increased, indicating that tumor immunosuppressive microenvironment has been effectively reprogramed. More importantly, it is found that the anticancer immunity induced by ACPP is dose dependent. Finally, 40% of the established CT26 tumors are completely eliminated by ACPP treatment with an optimized dose. This study proposes a simple and effective strategy for promoting cancer immunotherapy.


Assuntos
Peptídeos Catiônicos Antimicrobianos , Neoplasias , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Linhagem Celular Tumoral , Morte Celular Imunogênica , Imunoterapia/métodos , Camundongos , Neoplasias/terapia , Microambiente Tumoral
17.
J Phys Chem Lett ; 12(22): 5363-5370, 2021 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-34076431

RESUMO

Antifouling polymer brushes are widely used to inhibit the formation of protein corona on nanoparticles (NPs) and subsequent accumulation in the liver and spleen. Herein, we demonstrate a θ-solvent-mediated method for the preparation of gold nanoparticles with a high polyethylene glycol (PEG) grafting density. Reaching the θ-solvent by adding salt (e.g., Na2SO4) can significantly increase the grafting density of the PEG brush to 2.08 chains/nm2. The PEG polymer brush prepared in the θ-solvent possesses a double-shell structure consisting of a concentrated polymer brush (CPB) and a semidilute polymer brush (SDPB), denoted as NP@CPB@SDPB, while those prepared in a good solvent have only a SDPB shell, i.e., NP@SDPB. Compared to the NP@SDPB structure, the NP@CPB@SDPB structure decreases the liver accumulation from 34.0%ID/g to 23.1%ID/g, leading to an increase in tumor accumulation from 8.5%ID/g to 12.8%ID/g. This work provides new insights from the perspective of polymer physical chemistry into the improved stealth properties and delivery efficiency of NPs, which will accelerate the clinical translation of nanomedicine.

18.
Science ; 371(6536): 1368-1374, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33632891

RESUMO

Chiral assemblies of plasmonic nanoparticles are known for strong circular dichroism but not for high optical asymmetry, which is limited by the unfavorable combination of electrical and magnetic field components compounded by strong scattering. Here, we show that these limitations can be overcome by the long-range organization of nanoparticles in a manner similar to the liquid crystals and found in helical assemblies of gold nanorods with human islet amyloid polypeptides. A strong, polarization-dependent spectral shift and the reduced scattering of energy states with antiparallel orientation of dipoles activated in assembled helices increased optical asymmetry g-factors by a factor of more than 4600. The liquid crystal-like color variations and the nanorod-accelerated fibrillation enable drug screening in complex biological media. Improvement of long-range order can also provide structural guidance for the design of materials with high optical asymmetry.

19.
Nano Lett ; 20(8): 6191-6198, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32697585

RESUMO

Vascular disrupting agents (VDAs) are emerging anticancer agents, which show rising demand for combination with cytostatic drugs (CSDs), owing to inadequate tumor inhibition when applied singly. Nevertheless, the combination remains a challenge due to the different working sites of VDAs and CSDs and hypoxia-induced drug resistance after disrupting neovasculature by VDAs. Herein, we developed a shell-stacked nanoparticle (SNP) for coencapsulation of a VDA combretastatin A-4 phosphate (CA4P) and a proteasome inhibitor bortezomib (BTZ). The SNP could spatiotemporally deliver CA4P to tumor neovasculature and BTZ to tumor cells mediated by the site-specific stimuli-activated drug release. Moreover, the SNP also reversed the drug resistance caused by the overexpressed ABCG2 under CA4P-induced hypoxic conditions. The spatiotemporally targeted combination therapy significantly inhibited the growth of both the human A549 pulmonary adenocarcinoma xenograft model and patient-derived xenograft (PDX) model of colon cancer in mice, providing a promising strategy for treating advanced cancers.


Assuntos
Antineoplásicos , Estilbenos , Animais , Antineoplásicos/farmacologia , Resistência a Medicamentos , Hipóxia , Camundongos , Nanomedicina
20.
ACS Appl Mater Interfaces ; 12(25): 28047-28056, 2020 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-32478501

RESUMO

Immunotherapy has been successfully used in the treatment of multiple malignancies, but clinical studies revealed low response rates. Thus, the development of new effective immunotherapeutic modalities is urgently needed. Successfully inducing tumor cell death with enhanced antigenicity is important for the expansion and differentiation of tumor-specific CD8+ cytotoxic T lymphocytes. Cationic liposome/DNA complexes (CLN/DNA), which usually have obvious cytotoxic effects, may improve the antitumor immunity through enhancing the immunogenicity of dying tumor cells. Herein, we report that a plasmid DNA-encapsulated cationic lipid nanoparticle formulated with cholesterol, DOTAP, and DSPE-mPEG2000 significantly increases the tumor cell death with high antigenicity in vitro. Furthermore, the cationic liposome/DNA complex (CLN/DNA) treatment promotes the activation of dendritic cells (DCs). We also find that the intratumorally injected CLN/DNA successfully promoted the activation of DCs in the tumor-draining lymph node. Importantly, both local tumor growth and distant tumor formation were significantly inhibited by T cell-dependent antitumor immune responses after intratumoral injection of CLN/DNA. This study presents a simple and effective strategy for improving the cancer immunotherapy.


Assuntos
Cátions/química , DNA/química , Imunoterapia/métodos , Lipossomos/química , Células Dendríticas/metabolismo , Linfonodos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA