Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 3987, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734698

ABSTRACT

Despite advances in active drug targeting for blood-brain barrier penetration, two key challenges persist: first, attachment of a targeting ligand to the drug or drug carrier does not enhance its brain biodistribution; and second, many brain diseases are intricately linked to microcirculation disorders that significantly impede drug accumulation within brain lesions even after they cross the barrier. Inspired by the neuroprotective properties of vinpocetine, which regulates cerebral blood flow, we propose a molecular library design centered on this class of cyclic tertiary amine compounds and develop a self-enhanced brain-targeted nucleic acid delivery system. Our findings reveal that: (i) vinpocetine-derived ionizable-lipidoid nanoparticles efficiently breach the blood-brain barrier; (ii) they have high gene-loading capacity, facilitating endosomal escape and intracellular transport; (iii) their administration is safe with minimal immunogenicity even with prolonged use; and (iv) they have potent pharmacologic brain-protective activity and may synergize with treatments for brain disorders as demonstrated in male APP/PS1 mice.


Subject(s)
Blood-Brain Barrier , Brain , Cerebrovascular Circulation , Nanoparticles , Vinca Alkaloids , Animals , Vinca Alkaloids/pharmacology , Vinca Alkaloids/pharmacokinetics , Vinca Alkaloids/administration & dosage , Vinca Alkaloids/chemistry , Nanoparticles/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Mice , Cerebrovascular Circulation/drug effects , Male , Brain/metabolism , Brain/drug effects , Brain/blood supply , Humans , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Mice, Inbred C57BL , Tissue Distribution , Drug Delivery Systems , Mice, Transgenic
2.
Drug Resist Updat ; 74: 101080, 2024 May.
Article in English | MEDLINE | ID: mdl-38579635

ABSTRACT

BACKGROUND: Gastric Cancer (GC) characteristically exhibits heterogeneous responses to treatment, particularly in relation to immuno plus chemo therapy, necessitating a precision medicine approach. This study is centered around delineating the cellular and molecular underpinnings of drug resistance in this context. METHODS: We undertook a comprehensive multi-omics exploration of postoperative tissues from GC patients undergoing the chemo and immuno-treatment regimen. Concurrently, an image deep learning model was developed to predict treatment responsiveness. RESULTS: Our initial findings associate apical membrane cells with resistance to fluorouracil and oxaliplatin, critical constituents of the therapy. Further investigation into this cell population shed light on substantial interactions with resident macrophages, underscoring the role of intercellular communication in shaping treatment resistance. Subsequent ligand-receptor analysis unveiled specific molecular dialogues, most notably TGFB1-HSPB1 and LTF-S100A14, offering insights into potential signaling pathways implicated in resistance. Our SVM model, incorporating these multi-omics and spatial data, demonstrated significant predictive power, with AUC values of 0.93 and 0.84 in the exploration and validation cohorts respectively. Hence, our results underscore the utility of multi-omics and spatial data in modeling treatment response. CONCLUSION: Our integrative approach, amalgamating mIHC assays, feature extraction, and machine learning, successfully unraveled the complex cellular interplay underlying drug resistance. This robust predictive model may serve as a valuable tool for personalizing therapeutic strategies and enhancing treatment outcomes in gastric cancer.


Subject(s)
Drug Resistance, Neoplasm , Fluorouracil , Stomach Neoplasms , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Humans , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Oxaliplatin/pharmacology , Oxaliplatin/administration & dosage , Oxaliplatin/therapeutic use , Deep Learning , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precision Medicine/methods , Male , Female , Middle Aged , Immunotherapy/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects , Multiomics
3.
Medicine (Baltimore) ; 102(21): e33755, 2023 May 26.
Article in English | MEDLINE | ID: mdl-37233443

ABSTRACT

Lung adenocarcinoma (LUAD) is a common lung cancer. Although there are various treatments for LUAD, its prognosis remains poor. Therefore, it is imperative to identify new targets and develop novel therapeutic strategies. In this study, we analyze the expression of proline rich 11 (PRR11) in pan cancer based on The Cancer Genome Atlas (TCGA) database, and explore the prognostic value of PRR11 in LUAD by GEPIA2 (Gene Expression Profiling Interactive Analysis, version 2) database. In addition, the relationship between PRR11 and the clinicopathological features of LUAD was analyzed using UALCAN database. The association between PRR11 expression and immune infiltration was accessed. The PRR11 related genes were screened using LinkOmics and GEPIA2. Gene Ontology Term Enrichment (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was performed by David database. The results suggested that the expression of PRR11 in most tumor tissues was significantly higher than that in normal tissues. In LUAD patients, high expression of PRR11 was associated with shortened first progression survival (FPS), overall survival (OS) and post progression survival (PPS), and correlated with individual cancer stage, race, gender, smoking habit, and tissue subtype. Besides, the high expression of PRR11 was accompanied by a relatively higher infiltration level of cancer-associated fibroblasts (CAFs) and myeloid-derived suppressor cell (MDSC), and decreased infiltration level of CD8+ T cells in the tumor microenvironment. GO analyses showed that PRR11 participated in biological processes such as cell division and cell cycle, and was involved in protein binding and microtubule binding functions. KEGG analyses revealed that PRR11 was implicated in p53 signaling pathway. All the results indicated that PRR11 might be an independent prognostic biomarker and therapeutic target for LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Prognosis , Adenocarcinoma of Lung/genetics , Lung Neoplasms/genetics , Cell Cycle , CD8-Positive T-Lymphocytes , Tumor Microenvironment
4.
ACS Nano ; 15(11): 18100-18112, 2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34751571

ABSTRACT

Targeted delivery of nanomedicines to M2 tumor-associated macrophages (TAMs) has been proposed to reduce tumor promotion and enhance the efficacy of anticancer therapy. However, upregulated receptors on M2 TAMs are also expressed on M1 TAMs and other macrophages in normal tissues. Therefore, improving targeting specificity remains a key challenge. Here, we developed a precise M2 TAM-targeted delivery system using "eat-me" and "don't-eat-me" signals. A CD47-derived self-peptide ligand (don't-eat-me signal) and galactose ligand (eat-me signal) were introduced on liposomes. Cleavable phospholipid-polyethylene glycol was covered on the surface and could combine with the self-peptide to inhibit macrophage recognition even after immunoglobulin M adsorption and protect galactose from hepatic clearance to prolong the circulation time and promote the accumulation of liposomes in tumors. This detachable polymer can be removed by the redox microenvironment upon transcytosis through the tumor endothelium and re-expose the self-peptide and galactose. The self-peptide highly reduced M1 macrophage phagocytosis, and the galactose ligand enhanced the interaction between the liposomes and M2 macrophages. Thus, the modified liposomes enabled specific recognition of M1/M2 TAMs. In vitro evidence revealed reduced endocytosis of the liposomes by M1 macrophages. Moreover, in vivo studies demonstrated that doxorubicin-loaded liposomes efficiently eliminated M2 TAMs but did not affect M1 TAMs, enhancing the potency of the antitumor therapy. Collectively, our results demonstrate the potential of combining active escape and active targeting for precisely delivering a drug of interest to M2 macrophages and suggest its application in anticancer therapy.


Subject(s)
Liposomes , Nanomedicine , Ligands , Galactose , Cell Line, Tumor , Macrophages/pathology , Peptides , Tumor Microenvironment
5.
J Control Release ; 331: 390-403, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33485884

ABSTRACT

Effective curative therapies for spinal cord injury (SCI), which is often accompanied by intestinal complications, are lacking. Potential therapeutic targets include astrocytes and their enteric nervous system counterpart, enteric glial cells (EGCs). Based on shared biomarkers and similar functions of both cell types, we designed an orally administered targeted delivery system in which the neuropeptide apamin, stabilized by sulfur replacement with selenium, was adopted as a targeting moiety, and the liposome surface was protected with a non-covalent cross-linked chitosan oligosaccharide lactate layer. The system effectively permeated through oral absorption barriers, targeted local EGCs and astrocytes after systemic circulation, allowing for comprehensive SCI therapy. Given the involvement of the gut-organ axis in a growing number of diseases, our research may shed light on new aspects of the oral administration route as a bypass for multiple interventions and targeted therapy.


Subject(s)
Liposomes , Spinal Cord Injuries , Astrocytes , Humans , Neuroglia , Spinal Cord , Spinal Cord Injuries/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...