Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38500369

RESUMO

Kidney-specific nanocarriers offer a targeted approach to enhance therapeutic efficacy and reduce off-target effects in renal treatments. The nanocarriers can achieve organ or cell specificity via passive targeting and active targeting mechanisms. Passive targeting capitalizes on the unique physiological traits of the kidney, with factors like particle size, charge, shape, and material properties enhancing organ specificity. Active targeting, on the other hand, achieves renal specificity through ligand-receptor interactions, modifying nanocarriers with molecules, peptides, or antibodies for receptor-mediated delivery. Nanotechnology-enabled therapy targets diseased kidney tissue by modulating podocytes and immune cells to reduce inflammation and enhance tissue repair, or by inhibiting myofibroblast differentiation to mitigate renal fibrosis. This review summarizes the current reports of the drug delivery systems that have been tested in vivo, identifies the nanocarriers that may preferentially accumulate in the kidney, and quantitatively compares the efficacy of various cargo-carrier combinations to outline optimal strategies and future research directions. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.


Assuntos
Nefropatias , Nanopartículas , Humanos , Nanopartículas/uso terapêutico , Nanopartículas/química , Nanotecnologia , Sistemas de Liberação de Medicamentos , Nefropatias/tratamento farmacológico , Ligantes , Portadores de Fármacos/química
2.
Eur J Pharm Biopharm ; 197: 114234, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38401743

RESUMO

Nanoparticle-based delivery systems such as RNA-encapsulating lipid nanoparticles (RNA LNPs) have dramatically advanced in function and capacity over the last few decades. RNA LNPs boast of a diverse array of external and core configurations that enhance targeted delivery and prolong circulatory retention, advancing therapeutic outcomes. Particularly within the realm of cancer immunotherapies, RNA LNPs are increasingly gaining prominence. Pre-clinical in vitro and in vivo studies have laid a robust foundation for new and ongoing clinical trials that are actively enrolling patients for RNA LNP cancer immunotherapy. This review explores RNA LNPs, starting from their core composition to their external membrane formulation, set against a backdrop of recent clinical breakthroughs. We further elucidate the LNP delivery avenues, broach the prevailing challenges, and contemplate the future perspectives of RNA LNP-mediated immunotherapy.


Assuntos
Lipossomos , Nanopartículas , Neoplasias , Humanos , Neoplasias/terapia , Imunoterapia , Projetos de Pesquisa , RNA
3.
Adv Healthc Mater ; 13(2): e2302268, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37748773

RESUMO

Combination immunotherapy has emerged as a promising strategy to address the challenges associated with immune checkpoint inhibitor (ICI) therapy in breast cancer. The efficacy of combination immunotherapy hinges upon the intricate and dynamic nature of the tumor microenvironment (TME), characterized by cellular heterogeneity and molecular gradients. However, current methodologies for drug screening often fail to accurately replicate these complex conditions, resulting in limited predictive capacity for treatment outcomes. Here, a tumor-microenvironment-on-chip (TMoC), integrating a circulation system and ex vivo tissue culture with physiological oxygen and nutrient gradients, is described. This platform enables spatial infiltration of cytotoxic CD8+ T cells and their targeted attack on the tumor, while preserving the high complexity and heterogeneity of the TME. The TMoC is employed to assess the synergistic effect of five targeted therapy drugs and five chemotherapy drugs in combination with immunotherapy, demonstrating strong concordance between chip and animal model responses. The TMoC holds significant potential for advancing drug development and guiding clinical decision-making, as it offers valuable insights into the complex dynamics of the TME.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Animais , Microambiente Tumoral , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Resultado do Tratamento
5.
J Control Release ; 356: 14-25, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36805873

RESUMO

Over the past decade, immunotherapy aiming to activate an effective antitumor immune response has ushered in a new era of cancer treatment. However, the efficacy of cancer immunotherapy is limited by low response rates and high systemic toxicity. Nanotechnology is an encouraging platform for the development of next-generation cancer immunotherapy to effectively treat advanced cancer. Nanotechnology-enabled immunotherapy has remarkable advantages, ranging from the increased bioavailability and stability of immunotherapeutic agents to the enhanced activation of immune cells and favorable safety profiles. Nanotechnology-enabled immunotherapy can target solid tumors through reprogramming or stimulating immune cells (i.e., nanovaccines); modulating the immunosuppressive tumor microenvironment; or targeting tumor cells and altering their responses to immune cells to generate effective antitumor immunity. In this Oration, I introduce the advanced strategies currently being pursued by our laboratory and other groups to improve the therapeutic efficacy of cancer immunotherapy and discuss the potential challenges and future directions.


Assuntos
Neoplasias , Humanos , Neoplasias/terapia , Imunoterapia , Nanotecnologia , Imunidade , Imunossupressores , Microambiente Tumoral
6.
J Control Release ; 354: 417-428, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36627025

RESUMO

Fibrosis is an excessive accumulation of extracellular matrix (ECM) that may cause severe organ dysfunction. Nitric oxide (NO), a multifunctional gaseous signaling molecule, may inhibit fibrosis, and delivery of NO may serve as a potential antifibrotic strategy. However, major limitations in the application of NO to treat fibrotic diseases include its nonspecificity, short half-life and low availability in fibrotic tissue. Herein, we aimed to develop a stimuli-responsive drug carrier to deliver NO to halt kidney fibrosis. We manufactured a nanoparticle (NP) composed of pH-sensitive poly[2-(diisopropylamino)ethyl methacrylate (PDPA) polymers to encapsulate a NO donor, a dinitrosyl iron complex (DNIC; [Fe2(µ-SEt)2(NO)4]). The NPs were stable at physiological pH 7.4 but disintegrated at pH 4.0-6.0. The NPs showed significant cytotoxicity to cultured human myofibroblasts and were able to inhibit the activation of myofibroblasts, as indicated by a lower expression level of α-smooth muscle actin and the synthesis of a major ECM component, collagen I, in cultured human myofibroblasts. When given to mice treated with unilateral ureteral ligation/obstruction (UUO) to induce kidney fibrosis, these NPs remained in blood at a stable concentration for as long as 24 h and might enter the fibrotic kidneys to suppress myofibroblast activation and collagen I production, leading to a 70% reduction in the fibrotic area. In summary, our strategy to assemble a NO donor, the iron nitrosyl complex DNIC, into pH-responsive NPs proves effective in treating renal fibrosis and warrants further investigation for its therapeutic potential.


Assuntos
Nefropatias , Obstrução Ureteral , Camundongos , Humanos , Animais , Rim , Óxido Nítrico/metabolismo , Nefropatias/tratamento farmacológico , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Colágeno Tipo I/metabolismo , Fibrose , Concentração de Íons de Hidrogênio , Camundongos Endogâmicos C57BL
7.
Adv Mater ; 35(13): e2208966, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36609913

RESUMO

Extracellular vesicles (EVs) are released by cells to mediate intercellular communication under pathological and physiological conditions. While small EVs (sEVs; <100-200 nm, exosomes) are intensely investigated, the properties and functions of medium and large EVs (big EVs (bEVs); >200 nm, microvesicles) are less well explored. Here, bEVs and sEVs are identified as distinct EV populations, and it is determined that bEVs are released in a greater bEV:sEV ratio in the aggressive human triple-negative breast cancer (TNBC) subtype. PalmGRET, bioluminescence-resonance-energy-transfer (BRET)-based EV reporter, reveals dose-dependent EV biodistribution at nonlethal and physiological EV dosages, as compared to lipophilic fluorescent dyes. Remarkably, the bEVs and sEVs exhibit unique biodistribution profiles, yet individually promote in vivo tumor growth in a syngeneic immunocompetent TNBC breast tumor murine model. The bEVs and sEVs share mass-spectrometry-identified tumor-progression-associated EV surface membrane proteins (tpEVSurfMEMs), which include solute carrier family 29 member 1, Cd9, and Cd44. tpEVSurfMEM depletion attenuates EV lung organotropism, alters biodistribution, and reduces protumorigenic potential. This study identifies distinct in vivo property and function of bEVs and sEVs in breast cancer, which suggest the significant role of bEVs in diseases, diagnostic and therapeutic applications.


Assuntos
Exossomos , Vesículas Extracelulares , Neoplasias de Mama Triplo Negativas , Camundongos , Humanos , Animais , Distribuição Tecidual , Proteínas de Membrana/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Vesículas Extracelulares/metabolismo , Exossomos/metabolismo , Carcinogênese/metabolismo
8.
Blood Adv ; 7(1): 145-158, 2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35358998

RESUMO

In chronic lymphocytic leukemia (CLL), B-cell receptor signaling, tumor-microenvironment interactions, and somatic mutations drive disease progression. To better understand the intersection between the microenvironment and molecular events in CLL pathogenesis, we integrated bulk transcriptome profiling of paired peripheral blood (PB) and lymph node (LN) samples from 34 patients. Oncogenic processes were upregulated in LN compared with PB and in immunoglobulin heavy-chain variable (IGHV) region unmutated compared with mutated cases. Single-cell RNA sequencing (scRNA-seq) distinguished 3 major cell states: quiescent, activated, and proliferating. The activated subpopulation comprised only 2.2% to 4.3% of the total tumor bulk in LN samples. RNA velocity analysis found that CLL cell fate in LN is unidirectional, starts in the proliferating state, transitions to the activated state, and ends in the quiescent state. A 10-gene signature derived from activated tumor cells was associated with inferior treatment-free survival (TFS) and positively correlated with the proportion of activated CD4+ memory T cells and M2 macrophages in LN. Whole exome sequencing (WES) of paired PB and LN samples showed subclonal expansion in LN in approximately half of the patients. Since mouse models have implicated activation-induced cytidine deaminase in mutagenesis, we compared AICDA expression between cases with and without clonal evolution but did not find a difference. In contrast, the presence of a T-cell inflamed microenvironment in LN was associated with clonal stability. In summary, a distinct minor tumor subpopulation underlies CLL pathogenesis and drives the clinical outcome. Clonal trajectories are shaped by the LN milieu, where T-cell immunity may contribute to suppressing clonal outgrowth. The clinical study is registered at clinicaltrials.gov as NCT00923507.


Assuntos
Leucemia Linfocítica Crônica de Células B , Camundongos , Animais , Leucemia Linfocítica Crônica de Células B/patologia , Heterogeneidade Genética , Região Variável de Imunoglobulina/genética , Transdução de Sinais , Progressão da Doença , Microambiente Tumoral/genética
9.
J Control Release ; 352: 920-930, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36334859

RESUMO

While immunotherapy has emerged as a promising strategy to treat glioblastoma multiforme (GBM), the limited availability of immunotherapeutic agents in tumors due to the presence of the blood-brain barrier (BBB) and immunosuppressive tumor microenvironment dampens efficacy. Nitric oxide (NO) plays a role in modulating both the BBB and tumor vessels and could thus be delivered to disrupt the BBB and improve the delivery of immunotherapeutics into GBM tumors. Herein, we report an immunotherapeutic approach that utilizes CXCR4-targeted lipid­calcium-phosphate nanoparticles with NO donors (LCP-NO NPs). The delivery of NO resulted in enhanced BBB permeability and thus improved gene delivery across the BBB. CXCR4-targeted LCP-NO NPs delivered siRNA against the immune checkpoint ligand PD-L1 to GBM tumors, silenced PD-L1 expression, increased cytotoxic T cell infiltration and activation in GBM tumors, and suppressed GBM progression. Thus, the codelivery of NO and PD-L1 siRNA by these CXCR4-targeted NPs may serve as a potential immunotherapy for GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Nanopartículas , Humanos , Glioblastoma/tratamento farmacológico , Antígeno B7-H1 , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Óxido Nítrico/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Imunoterapia , Microambiente Tumoral , Linhagem Celular Tumoral , Receptores CXCR4/genética
10.
J Mater Chem B ; 10(46): 9590-9598, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36106522

RESUMO

Cirrhosis is a major cause of global morbidity and mortality, and significantly leads to a heightened risk of liver cancer. Despite decades of efforts in seeking for cures for cirrhosis, this disease remains irreversible. To assist in the advancement of understanding toward cirrhosis as well as therapeutic options, various disease models, each with different strengths, are developed. With the development of three-dimensional (3D) cell culture in recent years, more realistic biochemical properties are observed in 3D cell models, which have gradually taken over the responsibilities of traditional 2D cell culture, and are expected to replace some of the animal models in the near future. Here, we propose a 3D fibrotic liver model inspired by liver lobules. In the model, 3D-printed poly(glycerol sebacate) acrylate (PGSA) scaffolds facilitated the formation of 3D tissues and guided the deposition of fibrotic structures. Through the sequential seeding of hepatic stellate cells (HSCs), HepG2 and HSCs, fibrotic septum-like tissues were created on PGSA scaffolds. As albumin secretion is considered a rather important function of the liver and is found only among hepatic cells, the detection of albumin secretion up to 30 days indicates the mimicking of basic liver functions. Moreover, the in vivo fibrotic tissue shows a high similarity to fibrotic septa. Finally, via complete encapsulation of HSCs, a down-regulated albumin secretion profile was observed in the capped model, which is a metabolic indicator that is important for the prognosis for liver cirrhosis. Looking forward, the incorporation of the vasculature will further upgrade the model into a sound tool for liver research and associated treatments.


Assuntos
Células Estreladas do Fígado , Cirrose Hepática , Animais , Células Estreladas do Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Fibrose , Albuminas/metabolismo , Acrilatos , Impressão Tridimensional
11.
J Hematol Oncol ; 15(1): 85, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35794621

RESUMO

BACKGROUND: Metastasis and chemoresistance are major culprits of cancer mortality, but factors contributing to these processes are incompletely understood. METHODS: Bioinformatics methods were used to identify the relations of Smyca expression to clinicopathological features of human cancers. RNA-sequencing analysis was used to reveal Smyca-regulated transcriptome. RNA pull-down and RNA immunoprecipitation were used to examine the binding of Smyca to Smad3/4 and c-Myc/Max. Chromatin immunoprecipitation and chromatin isolation by RNA purification were used to determine the binding of transcription factors and Smyca to various gene loci, respectively. Real-time RT-PCR and luciferase assay were used to examine gene expression levels and promoter activities, respectively. Xenograft mouse models were performed to evaluate the effects of Smyca on metastasis and chemoresistance. Nanoparticle-assisted gapmer antisense oligonucleotides delivery was used to target Smyca in vivo. RESULTS: We identify lncRNA Smyca for its association with poor prognosis of many cancer types. Smyca potentiates metabolic reprogramming, migration, invasion, cancer stemness, metastasis and chemoresistance. Mechanistically, Smyca enhances TGF-ß/Smad signaling by acting as a scaffold for promoting Smad3/Smad4 association and further serves as a Smad target to amplify/prolong TGF-ß signaling. Additionally, Smyca potentiates c-Myc-mediated transcription by enhancing the recruitment of c-Myc/Max complex to a set of target promoters and c-Myc binding to TRRAP. Through potentiating TGF-ß and c-Myc pathways, Smyca synergizes the Warburg effect elicited by both pathways but evades the anti-proliferative effect of TGF-ß. Targeting Smyca prevents metastasis and overcomes chemoresistance. CONCLUSIONS: This study uncovers a lncRNA that coordinates tumor-relevant pathways to orchestra a pro-tumor program and establishes the clinical values of Smyca in cancer prognosis and therapy.


Assuntos
Neoplasias , RNA Longo não Codificante , Animais , Humanos , Camundongos , Regiões Promotoras Genéticas , RNA Longo não Codificante/genética , Fator de Crescimento Transformador beta/metabolismo
12.
J Biomed Sci ; 29(1): 29, 2022 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-35534851

RESUMO

BACKGROUND: Castration-resistant prostate cancer (CRPC) with sustained androgen receptor (AR) signaling remains a critical clinical challenge, despite androgen depletion therapy. The Jumonji C-containing histone lysine demethylase family 4 (KDM4) members, KDM4A‒KDM4C, serve as critical coactivators of AR to promote tumor growth in prostate cancer and are candidate therapeutic targets to overcome AR mutations/alterations-mediated resistance in CRPC. METHODS: In this study, using a structure-based approach, we identified a natural product, myricetin, able to block the demethylation of histone 3 lysine 9 trimethylation by KDM4 members and evaluated its effects on CRPC. A structure-based screening was employed to search for a natural product that inhibited KDM4B. Inhibition kinetics of myricetin was determined. The cytotoxic effect of myricetin on various prostate cancer cells was evaluated. The combined effect of myricetin with enzalutamide, a second-generation AR inhibitor toward C4-2B, a CRPC cell line, was assessed. To improve bioavailability, myricetin encapsulated by poly lactic-co-glycolic acid (PLGA), the US food and drug administration (FDA)-approved material as drug carriers, was synthesized and its antitumor activity alone or with enzalutamide was evaluated using in vivo C4-2B xenografts. RESULTS: Myricetin was identified as a potent α-ketoglutarate-type inhibitor that blocks the demethylation activity by KDM4s and significantly reduced the proliferation of both androgen-dependent (LNCaP) and androgen-independent CRPC (CWR22Rv1 and C4-2B). A synergistic cytotoxic effect toward C4-2B was detected for the combination of myricetin and enzalutamide. PLGA-myricetin, enzalutamide, and the combined treatment showed significantly greater antitumor activity than that of the control group in the C4-2B xenograft model. Tumor growth was significantly lower for the combination treatment than for enzalutamide or myricetin treatment alone. CONCLUSIONS: These results suggest that myricetin is a pan-KDM4 inhibitor and exhibited potent cell cytotoxicity toward CRPC cells. Importantly, the combination of PLGA-encapsulated myricetin with enzalutamide is potentially effective for CRPC.


Assuntos
Antineoplásicos , Produtos Biológicos , Flavonoides , Neoplasias de Próstata Resistentes à Castração , Androgênios/farmacologia , Androgênios/uso terapêutico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Flavonoides/farmacologia , Glicolatos , Glicóis/farmacologia , Glicóis/uso terapêutico , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/farmacologia , Masculino , Nitrilas/farmacologia , Nitrilas/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores Androgênicos/uso terapêutico
13.
J Control Release ; 346: 169-179, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35429575

RESUMO

Fibrosis is an excessive accumulation of the extracellular matrix within solid organs in response to injury and a common pathway that leads functional failure. No clinically approved agent is available to reverse or even prevent this process. Herein, we report a nanotechnology-based approach that utilizes a drug carrier to deliver a therapeutic cargo specifically to fibrotic kidneys, thereby improving the antifibrotic effect of the drug and reducing systemic toxicity. We first adopted in vitro-in vivo combinatorial phage display technology to identify peptide ligands that target myofibroblasts in mouse unilateral ureteral obstruction (UUO)-induced fibrotic kidneys. We then engineered lipid-coated poly(lactic-co-glycolic acid) nanoparticles (NPs) with fibrotic kidney-homing peptides on the surface and sorafenib, a potent antineoplastic multikinase inhibitor, encapsulated in the core. Sorafenib loaded in the myofibroblast-targeted NPs significantly reduced the infiltration of α-smooth muscle actin-expressing myofibroblasts and deposition of collagen I in UUO-treated kidneys and enhanced renal plasma flow measured by Technetium-99m mercaptoacetyltriglycine scintigraphy. This study demonstrates the therapeutic potential of the newly identified peptide fragments as anchors to target myofibroblasts and represents a strategic advance for selective delivery of sorafenib to treat renal fibrosis. SIGNIFICANCE STATEMENT: Renal fibrosis is a pathological feature accounting for the majority of issues in chronic kidney disease (CKD), which may progress to end-stage renal disease (ESRD). This manuscript describes a myofibroblast-targeting drug delivery system modified with phage-displayed fibrotic kidney-homing peptides. By loading the myofibroblast-targeting nanoparticles (NPs) with sorafenib, a multikinase inhibitor, the NPs could suppress collagen synthesis in cultured human myofibroblasts. When given intravenously to mice with UUO-induced renal fibrosis, sorafenib loaded in myofibroblast-targeting NPs significantly ameliorated renal fibrosis. This approach provides an efficient therapeutic option to renal fibrosis. The myofibroblast-targeting peptide ligands and nanoscale drug carriers may be translated into clinical application in the future.


Assuntos
Nefropatias , Nanopartículas , Obstrução Ureteral , Animais , Colágeno , Modelos Animais de Doenças , Portadores de Fármacos/uso terapêutico , Fibrose , Rim , Nefropatias/patologia , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos , Sorafenibe/uso terapêutico , Obstrução Ureteral/tratamento farmacológico , Obstrução Ureteral/patologia
15.
Vaccines (Basel) ; 10(2)2022 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-35214731

RESUMO

As of August 2021, there have been over 200 million confirmed case of coronavirus disease 2019 caused by severe acute respiratory syndrome coronavirus and more than 4 million COVID-19-related deaths globally. Although real-time polymerase chain reaction is considered to be the primary method of detection for SARS-CoV-2 infection, the use of serological assays for detecting COVID-19 antibodies has been shown to be effective in aiding with diagnosis, particularly in patients who have recovered from the disease and those in later stages of infection. Since it has a high detection rate and few limitations compared to conventional enzyme-linked immunosorbent assay protocols, we used a lateral flow immunoassay as our diagnostic tool of choice. Since lateral flow immunoassay results interpreted by the naked eye may lead to erroneous diagnoses, we developed an innovative, portable device with the capacity to capture a high-resolution reflectance spectrum as a means of promoting diagnostic accuracy. We combined this spectrum-based device with commercial lateral flow immunoassays to detect the neutralizing antibody in serum samples collected from 30 COVID-19-infected patients (26 mild cases and four severe cases). The results of our approach, lateral flow immunoassays coupled with a spectrum-based reader, demonstrated a 0.989 area under the ROC curve, 100% sensitivity, 95.7% positive predictive value, 87.5% specificity, and 100% negative predictive value. As a result, our approach exhibited great value for neutralizing antibody detection. In addition to the above tests, we also tested plasma samples from 16 AstraZeneca-vaccinated (ChAdOx1nCoV-19) patients and compared our approach and enzyme-linked immunosorbent assay results to see whether our approach could be applied to vaccinated patients. The results showed a high correlation between these two approaches, indicating that the lateral flow immunoassay coupled with a spectrum-based reader is a feasible approach for diagnosing the presence of a neutralizing antibody in both COVID-19-infected and vaccinated patients.

16.
ACS Appl Mater Interfaces ; 14(3): 3849-3863, 2022 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-35019259

RESUMO

Nitric oxide (NO) is an endogenous gasotransmitter regulating alternative physiological processes in the cardiovascular system. To achieve translational application of NO, continued efforts are made on the development of orally active NO prodrugs for long-term treatment of chronic cardiovascular diseases. Herein, immobilization of NO-delivery [Fe2(µ-SCH2CH2COOH)2(NO)4] (DNIC-2) onto MIL-88B, a metal-organic framework (MOF) consisting of biocompatible Fe3+ and 1,4-benzenedicarboxylate (BDC), was performed to prepare a DNIC@MOF microrod for enhanced oral delivery of NO. In simulated gastric fluid, protonation of the BDC linker in DNIC@MOF initiates its transformation into a DNIC@tMOF microrod, which consisted of DNIC-2 well dispersed and confined within the BDC-based framework. Moreover, subsequent deprotonation of the BDC-based framework in DNIC@tMOF under simulated intestinal conditions promotes the release of DNIC-2 and NO. Of importance, this discovery of transformer-like DNIC@MOF provides a parallel insight into its stepwise transformation into DNIC@tMOF in the stomach followed by subsequent conversion into molecular DNIC-2 in the small intestine and release of NO in the bloodstream of mice. In comparison with acid-sensitive DNIC-2, oral administration of DNIC@MOF results in a 2.2-fold increase in the oral bioavailability of NO to 65.7% in mice and an effective reduction of systolic blood pressure (SBP) to a ΔSBP of 60.9 ± 4.7 mmHg in spontaneously hypertensive rats for 12 h.


Assuntos
Materiais Biocompatíveis/farmacologia , Estruturas Metalorgânicas/farmacologia , Óxido Nítrico/química , Pró-Fármacos/farmacologia , Administração Oral , Animais , Materiais Biocompatíveis/administração & dosagem , Pressão Sanguínea/efeitos dos fármacos , Eletrodos , Concentração de Íons de Hidrogênio , Teste de Materiais , Estruturas Metalorgânicas/administração & dosagem , Camundongos , Óxido Nítrico/administração & dosagem , Tamanho da Partícula , Pró-Fármacos/química , Propriedades de Superfície
17.
Gut ; 71(9): 1843-1855, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-34921062

RESUMO

OBJECTIVE: Stromal barriers, such as the abundant desmoplastic stroma that is characteristic of pancreatic ductal adenocarcinoma (PDAC), can block the delivery and decrease the tumour-penetrating ability of therapeutics such as tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), which can selectively induce cancer cell apoptosis. This study aimed to develop a TRAIL-based nanotherapy that not only eliminated the extracellular matrix barrier to increase TRAIL delivery into tumours but also blocked antiapoptotic mechanisms to overcome TRAIL resistance in PDAC. DESIGN: Nitric oxide (NO) plays a role in preventing tissue desmoplasia and could thus be delivered to disrupt the stromal barrier and improve TRAIL delivery in PDAC. We applied an in vitro-in vivo combinatorial phage display technique to identify novel peptide ligands to target the desmoplastic stroma in both murine and human orthotopic PDAC. We then constructed a stroma-targeted nanogel modified with phage display-identified tumour stroma-targeting peptides to co-deliver NO and TRAIL to PDAC and examined the anticancer effect in three-dimensional spheroid cultures in vitro and in orthotopic PDAC models in vivo. RESULTS: The delivery of NO to the PDAC tumour stroma resulted in reprogramming of activated pancreatic stellate cells, alleviation of tumour desmoplasia and downregulation of antiapoptotic BCL-2 protein expression, thereby facilitating tumour penetration by TRAIL and substantially enhancing the antitumour efficacy of TRAIL therapy. CONCLUSION: The co-delivery of TRAIL and NO by a stroma-targeted nanogel that remodels the fibrotic tumour microenvironment and suppresses tumour growth has the potential to be translated into a safe and promising treatment for PDAC.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animais , Carcinoma Ductal Pancreático/patologia , Humanos , Camundongos , Nanogéis , Óxido Nítrico , Neoplasias Pancreáticas/patologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Microambiente Tumoral , Neoplasias Pancreáticas
18.
ACS Appl Mater Interfaces ; 13(41): 48478-48491, 2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34633791

RESUMO

Three-dimensional (3D) spheroid culture provides opportunities to model tumor growth closer to its natural context. The collagen network in the extracellular matrix supports autonomic tumor cell proliferation, but its presence and role in tumor spheroids remain unclear. In this research, we developed an in vitro 3D co-culture model in a microwell 3D (µ-well 3D) cell-culture array platform to mimic the tumor microenvironment (TME). The modular setup is used to characterize the paracrine signaling molecules and the role of the intraspheroidal collagen network in cancer drug resistance. The µ-well 3D platform is made up of poly(dimethylsiloxane) that contains 630 round wells for individual spheroid growth. Inside each well, the growth surface measured 500 µm in diameter and was functionalized with the amphiphilic copolymer. HCT-8 colon cancer cells and/or NIH3T3 fibroblasts were seeded in each well and incubated for up to 9 days for TME studies. It was observed that NIH3T3 cells promoted the kinetics of tumor organoid formation. The two types of cells self-organized into core-shell chimeric tumor spheroids (CTSs) with fibroblasts confined to the shell and cancer cells localized to the core. Confocal microscopy analysis indicated that a type-I collagen network developed inside the CTS along with increased TGF-ß1 and α-SMA proteins. The results were correlated with a significantly increased stiffness in 3D co-cultured CTS up to 52 kPa as compared to two-dimensional (2D) co-culture. CTS was more resistant to 5-FU (IC50 = 14.0 ± 3.9 µM) and Regorafenib (IC50 = 49.8 ± 9.9 µM) compared to cells grown under the 2D condition 5-FU (IC50 = 12.2 ± 3.7 µM) and Regorafenib (IC50 = 5.9 ± 1.9 µM). Targeted collagen homeostasis with Sclerotiorin led to damaged collagen structure and disrupted the type-I collagen network within CTS. Such a treatment significantly sensitized collagen-supported CTS to 5-FU (IC50 = 4.4 ± 1.3 µM) and to Regorafenib (IC50 = 0.5 ± 0.2 µM). In summary, the efficient formation of colon cancer CTSs in a µ-well 3D culture platform allows exploration of the desmoplastic TME. The novel role of intratumor collagen quality as a drug sensitization target warrants further investigation.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Esferoides Celulares/metabolismo , Microambiente Tumoral/fisiologia , Animais , Benzopiranos/farmacologia , Técnicas de Cultura de Células em Três Dimensões/métodos , Técnicas de Cocultura/métodos , Colágeno Tipo I/antagonistas & inibidores , Colágeno Tipo I/metabolismo , Neoplasias Colorretais/metabolismo , Fluoruracila/farmacologia , Humanos , Camundongos , Células NIH 3T3 , Compostos de Fenilureia/farmacologia , Poloxâmero/química , Piridinas/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Microambiente Tumoral/efeitos dos fármacos
19.
J Med Chem ; 64(19): 14513-14525, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34558909

RESUMO

Autophagy is upregulated in response to metabolic stress, a hypoxic tumor microenvironment, and therapeutic stress in various cancers and mediates tumor progression and resistance to cancer therapy. Herein, we identified a cinchona alkaloid derivative containing urea (C1), which exhibited potential cytotoxicity and inhibited autophagy in hepatocellular carcinoma (HCC) cells. We showed that C1 not only induced apoptosis but also blocked autophagy in HCC cells, as indicated by the increased expression of LC3-II and p62, inhibition of autophagosome-lysosome fusion, and suppression of the Akt/mTOR/S6k pathway in the HCC cells. Finally, to improve its solubility and efficacy, we encapsulated C1 into PEGylated lipid-poly(lactic-co-glycolic acid) (PLGA) nanoscale drug carriers. Systemic administration of nanoscale C1 significantly suppressed primary tumor growth and prevented distant metastasis while maintaining a desirable safety profile. Our findings demonstrate that C1 combines autophagy modulation and apoptosis induction in a single molecule, making it a promising therapeutic option for HCC.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Alcaloides de Cinchona/farmacologia , Neoplasias Hepáticas/patologia , Ureia/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Microambiente Tumoral/efeitos dos fármacos
20.
JACS Au ; 1(7): 998-1013, 2021 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-34467346

RESUMO

Nitric oxide (NO), a pro-neurogenic and antineuroinflammatory gasotransmitter, features the potential to develop a translational medicine against neuropathological conditions. Despite the extensive efforts made on the controlled delivery of therapeutic NO, however, an orally active NO prodrug for a treatment of chronic neuropathy was not reported yet. Inspired by the natural dinitrosyl iron unit (DNIU) [Fe(NO)2], in this study, a reversible and dynamic interaction between the biomimetic [(NO)2Fe(µ-SCH2CH2OH)2Fe(NO)2] (DNIC-1) and serum albumin (or gastrointestinal mucin) was explored to discover endogenous proteins as a vehicle for an oral delivery of NO to the brain after an oral administration of DNIC-1. On the basis of the in vitro and in vivo study, a rapid binding of DNIC-1 toward gastrointestinal mucin yielding the mucin-bound dinitrosyl iron complex (DNIC) discovers the mucoadhesive nature of DNIC-1. A reversible interconversion between mucin-bound DNIC and DNIC-1 facilitates the mucus-penetrating migration of DNIC-1 shielded in the gastrointestinal tract of the stomach and small intestine. Moreover, the NO-release reactivity of DNIC-1 induces the transient opening of the cellular tight junction and enhances its paracellular permeability across the intestinal epithelial barrier. During circulation in the bloodstream, a stoichiometric binding of DNIC-1 to the serum albumin, as another endogenous protein vehicle, stabilizes the DNIU [Fe(NO)2] for a subsequent transfer into the brain. With aging mice under a Western diet as a disease model for metabolic syndrome and cognitive impairment, an oral administration of DNIC-1 in a daily manner for 16 weeks activates the hippocampal neurogenesis and ameliorates the impaired cognitive ability. Taken together, these findings disclose the synergy between biomimetic DNIC-1 and endogenous protein vehicles for an oral delivery of therapeutic NO to the brain against chronic neuropathy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA