Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Clin Cancer Res ; 30(5): 942-958, 2024 03 01.
Article in English | MEDLINE | ID: mdl-37921739

ABSTRACT

Immune-checkpoint inhibitor (ICI) therapy has dramatically changed the clinical landscape for several cancers, and ICI use continues to expand across many cancer types. Low baseline clearance (CL) and/or a large reduction of CL during treatment correlates with better clinical response and longer survival. Similar phenomena have also been reported with other monoclonal antibodies (mAb) in cancer and other diseases, highlighting a characteristic of mAb clinical pharmacology that is potentially shared among various mAbs and diseases. Though tempting to attribute poor outcomes to low drug exposure and arguably low target engagement due to high CL, such speculation is not supported by the relatively flat exposure-response relationship of most ICIs, where a higher dose or exposure is not likely to provide additional benefit. Instead, an elevated and/or increasing CL could be a surrogate marker of the inherent resistant phenotype that cannot be reversed by maximizing drug exposure. The mechanisms connecting ICI clearance, therapeutic efficacy, and resistance are unclear and likely to be multifactorial. Therefore, to explore the potential of ICI CL as an early marker for efficacy, this review highlights the similarities and differences of CL characteristics and CL-response relationships for all FDA-approved ICIs, and we compare and contrast these to selected non-ICI mAbs. We also discuss underlying mechanisms that potentially link mAb CL with efficacy and highlight existing knowledge gaps and future directions where more clinical and preclinical investigations are warranted to clearly understand the value of baseline and/or time-varying CL in predicting response to ICI-based therapeutics.


Subject(s)
Antibodies, Monoclonal , Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Drug Elimination Routes , Kinetics , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy
2.
Pharmacol Res ; 199: 107048, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38145833

ABSTRACT

High baseline clearance of immune checkpoint inhibitors (ICIs), independent of dose or systemic exposure, is associated with cachexia and poor outcomes in cancer patients. Mechanisms linking ICI clearance, cachexia and ICI therapy failure are unknown. Here, we evaluate in four murine models and across multiple antibodies whether altered baseline catabolic clearance of administered antibody requires a tumor and/or cachexia and whether medical reversal of cachexia phenotype can alleviate altered clearance. Key findings include mild cachexia phenotype and lack of elevated pembrolizumab clearance in the MC38 tumor-bearing model. We also observed severe cachexia and decreased, instead of increased, baseline pembrolizumab clearance in the tumor-free cisplatin-induced cachexia model. Liver Fcgrt expression correlated with altered baseline catabolic clearance, though elevated clearance was still observed with antibodies having no (human IgA) or reduced (human H310Q IgG1) FcRn binding. We conclude cachexia phenotype coincides with altered antibody clearance, though tumor presence is neither sufficient nor necessary for altered clearance in immunocompetent mice. Magnitude and direction of clearance alteration correlated with hepatic Fcgrt, suggesting changes in FcRn expression and/or recycling function may be partially responsible, though factors beyond FcRn also contribute to altered clearance in cachexia.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Animals , Mice , Immune Checkpoint Inhibitors/therapeutic use , Cachexia/drug therapy , Cachexia/etiology , Cachexia/metabolism , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/metabolism , Liver/metabolism , Immunoglobulin G/metabolism
3.
Exp Cell Res ; 433(2): 113830, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37913974

ABSTRACT

Many cancer cells exhibit enhanced glycolysis, which is seen as one of the hallmark metabolic alterations, known as Warburg effect. Substantial evidence shows that upregulated glycolytic enzymes are often linked to malignant growth. Using glycolytic inhibitors for anticancer treatment has become appealing in recent years for therapeutic intervention in cancers with highly glycolytic characteristic, including non-small cell lung cancer (NSCLC). In this work, we studied the anticancer effects and the underlying mechanisms of combination of benzerazide hydrocholoride (Benz), a hexokinase 2 (HK2) inhibitor and 64, a pyruvate dehydrogenase kinase 1 (PDK1) inhibitor, in several NSCLC cell lines. We found that combination of Benz and 64 exhibited strong synergistic anticancer effects in NCI-H1975, HCC827, NCI-H1299 and SK-LU-1 cell lines. With this combination treatment, we observed changes of certain mechanistic determinants associated with metabolic stress caused by glycolysis restriction, such as mitochondrial membrane potential depolarization, overproduction of reactive oxygen species [1], activation of AMPK and down-regulation of mTOR, which contributed to enhanced apoptosis. Moreover, Benz and 64 together significantly suppressed the tumor growth in HCC827 cell mouse xenograft model. Taken together, our study may suggest that combined inhibition of HK2 and PDK1 using Benz and 64 could be a viable anticancer strategy for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Hexokinase , Lung Neoplasms , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Animals , Humans , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Glycolysis , Hexokinase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Signal Transduction
4.
Cancer Lett ; 577: 216425, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37805163

ABSTRACT

Lung adenocarcinoma (LUAD) is one of the most prevalent and aggressive types of lung cancer. Metabolic reprogramming plays a critical role in the development and progression of LUAD. Pyruvate dehydrogenase kinase 1 (PDK1) and lactate dehydrogenase A (LDHA) are two key enzymes involved in glucose metabolism, whilst their aberrant expressions are often associated with tumorigenesis. Herein, we investigated the anticancer effects of combined inhibition of PDK1 and LDHA in LUAD in vitro and in vivo and its underlying mechanisms of action. The combination of a PDK1 inhibitor, 64, and a LDHA inhibitor, NHI-Glc-2, led to a synergistic growth inhibition in 3 different LUAD cell lines and more than additively suppressed tumor growth in the LUAD xenograft H1975 model. This combination also inhibited cellular migration and colony formation, while it induced a metabolic shift from glycolysis to oxidative phosphorylation (OXPHOS) resulting in mitochondrial depolarization and apoptosis in LUAD cells. These effects were related to modulation of multiple cell signaling pathways, including AMPK, RAS/ERK, and AKT/mTOR. Our findings demonstrate that simultaneous inhibition of multiple glycolytic enzymes (PDK1 and LDHA) is a promising novel therapeutic approach for LUAD.


Subject(s)
Adenocarcinoma of Lung , Lactate Dehydrogenase 5 , Lung Neoplasms , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Humans , Adenocarcinoma of Lung/drug therapy , Cell Death , Cell Line, Tumor , Cell Proliferation , Glycolysis , L-Lactate Dehydrogenase , Lactate Dehydrogenase 5/antagonists & inhibitors , Lactate Dehydrogenase 5/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Signal Transduction
5.
Phytother Res ; 37(12): 5837-5853, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37621136

ABSTRACT

Upon prolonged use of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) in non-small-cell lung cancer (NSCLC), acquired drug resistance inevitably occurs. This study investigates the combined use of EGFR-TKIs (gefitinib or osimertinib) with epigallocatechin gallate (EGCG) to overcome acquired drug resistance in NSCLC models. The in vitro antiproliferative effects of EGFR-TKIs and EGCG combination in EGFR-mutant parental and resistant cell lines were evaluated. The in vivo efficacy of the combination was assessed in xenograft mouse models derived from EGFR-TKI-resistant NSCLC cells. We found that the combined use of EGFR-TKIs and EGCG significantly reversed the Warburg effect by suppressing glycolysis while boosting mitochondrial respiration, which was accompanied by increased cellular ROS and decreased lactate secretion. The combination effectively activated the AMPK pathway while inhibited both ERK/MAPK and AKT/mTOR pathways, leading to cell cycle arrest and apoptosis, particularly in drug-resistant NSCLC cells. The in vivo results obtained from mouse tumor xenograft model confirmed that EGCG effectively overcame osimertinib resistance. This study revealed that EGCG suppressed cancer bypass survival signaling and altered cancer metabolic profiles, which is a promising anticancer adjuvant of EGFR-TKIs to overcome acquired drug resistance in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/pathology , Proto-Oncogene Proteins c-akt/metabolism , AMP-Activated Protein Kinases , Lung Neoplasms/pathology , Cell Proliferation , Protein Kinase Inhibitors/pharmacology , Drug Resistance, Neoplasm , ErbB Receptors , Glucose/pharmacology , Cell Line, Tumor , Mutation
6.
FEBS J ; 290(19): 4792-4809, 2023 10.
Article in English | MEDLINE | ID: mdl-37410361

ABSTRACT

Lung cancer cells often show elevated levels of reactive oxygen species (ROS) and nicotinamide adenine dinucleotide phosphate (NADPH). However, the connections between deregulated redox homeostasis in different subtypes of lung cancer and acquired drug resistance in lung cancer have not yet been fully established. Herein, we analyzed different subtypes of lung cancer data reported in the Cancer Cell Line Encyclopedia (CCLE) database, the Cancer Genome Atlas program (TCGA), and the sequencing data obtained from a gefitinib-resistant non-small-cell lung cancer (NSCLC) cell line (H1975GR). Using flux balance analysis (FBA) model integrated with multiomics data and gene expression profiles, we identified cytosolic malic enzyme 1 (ME1) and glucose-6-phosphate dehydrogenase as the major contributors to the significantly upregulated NADPH flux in NSCLC tissues as compared with normal lung tissues, and gefitinib-resistant NSCLC cell line as compared with the parental cell line. Silencing the gene expression of either of these two enzymes in two osimertinib-resistant NSCLC cell lines (H1975OR and HCC827OR) exhibited strong antiproliferative effects. Our findings not only underscored the pivotal roles of cytosolic ME1 and glucose-6-phosphate dehydrogenase in regulating redox states in NSCLC cells but also provided novel insights into their potential roles in drug-resistant NSCLC cells with disturbed redox states.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Gefitinib/pharmacology , NADP/metabolism , Glucosephosphate Dehydrogenase/genetics , Drug Resistance, Neoplasm/genetics , Oxidation-Reduction , Cell Line, Tumor , Cell Proliferation
7.
Chem Biol Interact ; 378: 110467, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37004952

ABSTRACT

Pyruvate dehydrogenase kinase 1 (PDK1) is an important metabolic enzyme which is often overexpressed in many types of cancers, including non-small-cell lung cancers (NSCLC). Targeting PDK1 appears to be an attractive anticancer strategy. Based on a previously reported moderate potent anticancer PDK1 inhibitor, 64, we developed three dichloroacetophenone biphenylsulfone ethers, 30, 31 and 32, which showed strong PDK1 inhibitions of 74%, 83% and 72% at 10 µM, respectively. Then we investigated the anticancer effects of 31 in two NSCLC cell lines, namely, NCI-H1299 and NCI-H1975. It was found that 31 exhibited sub-micromolar cancer cell IC50s, suppressed colony formation, induced mitochondrial membrane potential depolarization, triggered apoptosis, altered cellular glucose metabolism, with concomitant reductions in extracellular lactate levels and enhanced the generation of reactive oxygen species in NSCLC cells. Moreover, 31 significantly suppressed the tumor growth in an NCI-H1975 mouse xenograft model, outperforming the anticancer effects of 64. Taken together our results suggested that inhibition of PDK1 via dichloroacetophenone biphenylsulfone ethers may provide a novel direction leading to an alternative treatment option in NSCLC therapy.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Protein Serine-Threonine Kinases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Ethers/pharmacology , Ethers/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cell Line, Tumor , Apoptosis , Cell Proliferation
8.
Vet Dermatol ; 34(3): 222-234, 2023 Jun.
Article in English | MEDLINE | ID: mdl-35929548

ABSTRACT

BACKGROUND: Mycophenolate mofetil (MMF) is the prodrug of mycophenolic acid (MPA) which acts as an immunosuppressive agent. During the biotransformation of MMF to MPA, additional metabolites including MPA phenol glucuronide (MPAG), MPA acyl glucuronide (AcMPAG) and MPA phenol glucoside (MPG) are formed. OBJECTIVE: To define the noncompartmental pharmacokinetic (PK) parameters of three single doses of intravenous (i.v.) MMF and its downstream metabolites in healthy horses. ANIMALS: Six healthy Standardbred mares. MATERIALS AND METHODS: Generic MMF (Par Pharmaceuticals; Chestnut Ridge, NY, USA) was reconstituted and administered as a single i.v. bolus at 1.0 mg/kg, 5.0 mg/kg and 10.0 mg/kg with an eight day washout between treatments. Blood samples were collected immediately before MMF administration and over 24 h. A liquid chromatography-tandem mass spectrometry assay was developed following FDA guidance to determine plasma MMF, MPA, MPAG, AcMPAG and MPG concentrations. Plasma concentrations were analysed independently, followed by calculation of geometric mean and coefficient of variation. RESULTS: Noncompartmental PK parameters were determined for MMF and all metabolites at all doses. MMF was rapidly converted to MPA in all horses. Each incremental dose of MMF resulted in increases in Cmax and AUCinf _obs for MPA and the three additional metabolites. Within the 10-fold dose range, the increase in Cmax and AUCinf _obs for MMF and its metabolites was nonlinear. CONCLUSIONS AND CLINICAL RELEVANCE: Horses biotransform MMF into MPA, MPAG, AcMPAG and MPG via the glucuronidation and glucosidation clearance pathways. Equine reference PK profiles for MPA and the metabolites, MPAG, AcMPAG and MPG were established.


Subject(s)
Mycophenolic Acid , Veterinary Drugs , Horses , Animals , Female , Mycophenolic Acid/therapeutic use , Glucuronides/pharmacokinetics , Immunosuppressive Agents/therapeutic use , Phenols , Area Under Curve
9.
Expert Rev Anticancer Ther ; 22(8): 861-874, 2022 08.
Article in English | MEDLINE | ID: mdl-35786142

ABSTRACT

INTRODUCTION: Immune checkpoint inhibitors (ICI) are now utilized as a standard of care treatment for multiple cancers, including in both the metastatic setting as well as in earlier stages of disease. The identification of unique immune-related adverse events (irAE) that occur during ICI treatment has led to intense research to identify potential risk factors and biomarkers that may assist in clinical decision making. Although initial studies in ICI were primarily in advanced stage disease, the use of ICI in earlier stages of disease as adjuvant therapies requires a better understanding of patient risk stratification to mitigate or prevent serious irAE. AREAS COVERED: In this review, we set out to describe the current state of research regarding potential risk factors for irAE in patients with non-small cell lung cancer, as well as explore the barriers to understanding irAE. We review data from irAE that occur in large phase 3 trials and prospective studies focusing on irAE, as well as the many retrospective studies that currently form the bulk of our understanding of irAE.


Subject(s)
Antineoplastic Agents, Immunological , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Antineoplastic Agents, Immunological/adverse effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Prospective Studies , Retrospective Studies , Risk Factors
10.
J Med Chem ; 65(14): 9955-9973, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35818137

ABSTRACT

Phenylbutyric acid (PBA) has been reported as a dual inhibitor of pyruvate dehydrogenase kinases (PDKs) and histone deacetylases (HDACs), exhibiting anticancer effects. However, the low membrane permeability and poor cellular uptake limit its access to the target organelle, resulting in weak potencies against the intended targets. Herein, we report the design and identification of a novel 4-CF3-phenyl triphenylphosphonium-based PBA conjugate (53) with improved in vitro and in vivo anticancer activities. Compound 53 exhibited an IC50 value of 2.22 µM against A375 cells, outperforming the parent drug PBA by about 4000-fold. In the A375 cell-derived xenograft mouse model, 53 reduced the tumor growth by 76% at a dose of 40 mg/kg, while PBA only reduced the tumor growth by 10% at a dose of 80 mg/kg. On the basis of these results, 53 may be considered for further preclinical evaluations for cancer therapy.


Subject(s)
Antineoplastic Agents , Prodrugs , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Histone Deacetylases , Humans , Mice , Mitochondria , Prodrugs/pharmacology , Prodrugs/therapeutic use , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
11.
Clin Lung Cancer ; 23(4): 345-355, 2022 06.
Article in English | MEDLINE | ID: mdl-35131184

ABSTRACT

BACKGROUND: Cancer cachexia exhibits decreased albumin and associates with short overall survival (OS) in patients with non-small cell lung cancer (NSCLC), but whether on-treatment albumin changes associate with OS in NSCLC patients treated with immune checkpoint inhibitors (ICIs) and combination chemoimmunotherapy has not been thoroughly evaluated. PATIENTS AND METHODS: We conducted a single-center retrospective study of patients with advanced NSCLC who received first-line ICI with or without chemotherapy between 2013 and 2020. The association of pretreatment albumin and early albumin changes with OS was evaluated using Kaplan-Meier method and Cox regression models. RESULTS: A total of 210 patients were included: 109 in ICI cohort and 101 in ICI + Chemo cohort. Within a median of 21 days from treatment initiation, patients with ≥ 10% of albumin decrease had significantly shorter OS compared to patients without albumin decrease in ICI cohort. Pretreatment albumin and albumin decrease within the first or second cycle of treatment were significantly and independently associated with OS in ICI cohort, but not in ICI + Chemo cohort. The lack of association between albumin and OS with the addition of chemotherapy was more pronounced among patients with ≥ 1% PD-L1 expression in subgroup analysis. CONCLUSION: Pretreatment serum albumin and early albumin decrease in ICI monotherapy was significantly associated with OS in advanced NSCLC. Early albumin change, as a routine lab value tested in clinic, may be combined with established biomarkers to improve outcome predictions of ICI monotherapy. The underlying mechanism of the observed association between decreased albumin and ICI resistance warrants further investigation.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Immune Checkpoint Inhibitors/therapeutic use , Prognosis , Retrospective Studies , Serum Albumin/therapeutic use
12.
Expert Opin Ther Pat ; 32(4): 441-453, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35001793

ABSTRACT

INTRODUCTION: One of the most distinctive hallmarks of cancer cells is increased glucose consumption for aerobic glycolysis, which is called the Warburg effect. In recent decades, extensive research has been carried out to exploit this famous phenomenon, trying to detect promising targetable vulnerabilities in altered metabolism to fight cancer. Targeting aberrant glucose metabolism can perturb cancer malignant proliferation and even induce programmed cell death. AREAS COVERED: This review covered the recent patents which focused on targeting key glycolytic enzymes, including hexokinase, pyruvate dehydrogenase kinases, and lactate dehydrogenase for cancer treatment. EXPERT OPINION: Compared with the conventional cancer treatment, specifically targeting the well-known Achilles heel, the Warburg effect has attracted considerable attention. Although there is still no single glycolytic agent for clinical cancer treatment, the combination of glycolytic inhibitor with conventional anticancer drugs or the combined use of multiple glycolytic inhibitors are being investigated extensively in recent years, which could emerge as attractive anticancer strategies.


Subject(s)
Neoplasms , Patents as Topic , Glucose/metabolism , Glycolysis , Humans , Neoplasms/pathology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
13.
JCSM Rapid Commun ; 4(2): 232-244, 2021.
Article in English | MEDLINE | ID: mdl-34514376

ABSTRACT

BACKGROUND: Monoclonal antibody (mAb) immune checkpoint inhibitor (ICI) therapies have dramatically impacted oncology this past decade. However, only about one-third of patients respond to treatment, and biomarkers to predict responders are lacking. Recent ICI clinical pharmacology data demonstrate high baseline drug clearance (CL0) significantly associates with shorter overall survival, independent of ICI exposure, in patients receiving ICI mAb therapies. This suggests CL0 may predict outcomes from ICI therapy, and cachectic signalling may link elevated CL0 and poor response. Our aim was to determine if mouse models of cancer cachexia will be useful for studying these phenomena and their underlying mechanisms. METHODS: We evaluated pembrolizumab CL in the C26 and Lewis lung carcinoma mouse models of cancer cachexia. A single treatment of vehicle or pembrolizumab, at a dose of 2 or 10 mg/kg, was administered intravenously by tail vein injection. Pembrolizumab was quantified by an ELISA in serial plasma samples, and FcRn gene (Fcgrt) expression was assessed in liver using real-time quantitative reverse transcription PCR. Non-compartmental and mixed-effects pharmacokinetics analyses were performed. RESULTS: We observed higher pembrolizumab CL0 and decreased Fcgrt expression in whole liver tissue from tumour-bearing vs. tumour-free mice. In multivariate analysis, presence of tumour, total murine IgG, muscle weight and Fcgrt expression were significant covariates on CL, and total murine IgG was a significant covariate on V1 and Q. CONCLUSIONS: These data demonstrate increases in catabolic clearance of monoclonal antibodies observed in humans can be replicated in cachectic mice, in which Fcgrt expression is also reduced. Notably, FcRn activity is essential for proper antigen presentation and antitumour immunity, which may permit the study of cachexia's impact on FcRn-mediated clearance and efficacy of ICI therapies.

14.
Drug Discov Today ; 26(3): 836-844, 2021 03.
Article in English | MEDLINE | ID: mdl-33450176

ABSTRACT

Cancer cells metabolize glucose via anaerobic glycolysis, with lactate formed in the cytosol as the end-product. To avoid intercellular acidification, excessive lactate and proton are excreted by monocarboxylate transporters (MCTs), which are often overexpressed in different malignant cancers. Targeting the MCT-mediated lactate/proton efflux makes MCTs a potentially interesting anticancer target. Although X-ray co-crystal structures of human MCTs with inhibitors are not yet available, homology models have been established, which helped to rationalize the binding modes and the design of new MCT inhibitors. In this review, we discuss the structures and functions of MCTs as well as recently reported small-molecule MCTs inhibitors. We assess the current development of MCT inhibitors and highlight possible directions for future development.


Subject(s)
Antineoplastic Agents/pharmacology , Monocarboxylic Acid Transporters/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Drug Design , Drug Development/methods , Glucose/metabolism , Glycolysis/physiology , Humans , Monocarboxylic Acid Transporters/metabolism , Neoplasms/pathology
15.
Virus Res ; 286: 198066, 2020 09.
Article in English | MEDLINE | ID: mdl-32553609

ABSTRACT

Primary Effusion Lymphoma (PEL) is a B-cell lymphoma associated with Kaposi's sarcoma herpesvirus (KSHV) infection. However, the mechanism of oncogenesis of PEL is still unclear. Studies have shown that the cellular transcriptional coactivator p300 regulates the interaction between host and virus, which plays a vital role in viral replication. In this study, we investigated the role of p300 in BCBL1 cells during the KSHV life cycle. We found that p300 knockout resulted in an overall increase for the early lytic genes and changed the expression of genes associated with tumor development, proliferation, and the immune response in the KSHV infected B cells. However, knockout of p300 significantly inhibited the expression of the immediate-early gene RTA and the late lytic gene K8 after KSHV lytic activation. Additionally, the intracellular KSHV genome copy number and the virion production were reduced. These results demonstrated that p300 plays a crucial role in suppressing KSHV viral replication in BCBL1. Furthermore, we observed that the growth of BCBL1 was inhibited by knockout of p300, which confirmed our findings that p300 promotes cell proliferation. This study further provided evidence that p300 plays an important role in the pathogenesis of BCBL1, which might lead to the oncogenesis of PEL caused by KSHV infection.


Subject(s)
B-Lymphocytes/virology , Cell Proliferation/genetics , E1A-Associated p300 Protein/genetics , Herpesvirus 8, Human/physiology , Lymphoma, Primary Effusion/virology , Virus Activation , B-Lymphocytes/pathology , Cell Line, Tumor , Gene Expression , Herpesvirus 8, Human/genetics , Humans , Lymphoma, Primary Effusion/pathology , RNA-Seq , Virus Latency
16.
Nanomicro Lett ; 12(1): 129, 2020 Jun 16.
Article in English | MEDLINE | ID: mdl-34138128

ABSTRACT

Liposomes hold great potential in anti-cancer drug delivery and the targeting treatment of tumors. However, the clinical therapeutic efficacy of liposomes is still limited by the complexity of tumor microenvironment (TME) and the insufficient accumulation in tumor sites. Meanwhile, the application of cholesterol and polyethylene glycol (PEG), which are usually used to prolong the blood circulation and stabilize the structure of liposomes respectively, has been questioned due to various disadvantages. Herein, we developed a ginsenoside Rh2-based multifunctional liposome system (Rh2-lipo) to effectively address these challenges once for all. Different with the conventional 'wooden' liposomes, Rh2-lipo is a much more brilliant carrier with multiple functions. In Rh2-lipo, both cholesterol and PEG were substituted by Rh2, which works as membrane stabilizer, long-circulating stealther, active targeting ligand, and chemotherapy adjuvant at the same time. Firstly, Rh2 could keep the stability of liposomes and avoid the shortcomings caused by cholesterol. Secondly, Rh2-lipo showed a specifically prolonged circulation behavior in the blood. Thirdly, the accumulation of the liposomes in the tumor was significantly enhanced by the interaction of glucose transporter of tumor cells with Rh2. Fourth, Rh2-lipo could remodel the structure and reverse the immunosuppressive environment in TME. When tested in a 4T1 breast carcinoma xenograft model, the paclitaxel-loaded Rh2-lipo realized high efficient tumor growth suppression. Therefore, Rh2-lipo not only innovatively challenges the position of cholesterol as a liposome component, but also provides another innovative potential system with multiple functions for anti-cancer drug delivery.

17.
Theranostics ; 9(15): 4437-4449, 2019.
Article in English | MEDLINE | ID: mdl-31285771

ABSTRACT

The clinical treatment of gastric cancer (GC) is hampered by the development of anticancer drug resistance and the unfavorable pharmacokinetics, off-target toxicity, and inadequate intratumoral accumulation of the current chemotherapy treatments. Ginsenosides combined with paclitaxel (PTX) have been shown to exert synergistic inhibition of human GC cell proliferation. In the present study, we developed a novel multifunctional liposome system, in which ginsenosides functioned as the chemotherapy adjuvant and membrane stabilizer. These had long blood circulation times and active targeting abilities, thus creating multifunctionality of the liposomes and facilitating drug administration to the GC cells. Methods: Three ginsenosides with different structures were used to formulate the unique nanocarrier, which was prepared using the thin-film hydration method. The stability of the ginsenoside liposomes was determined by particle size analysis using dynamic light scattering. The long circulation time of ginsenoside liposomes was compared with that of conventional liposome and polyethylene glycosylated liposomes in vivo. The active targeting effect of ginsenoside liposomes was examined with a GC xenograft model using an in vivo imaging system. To examine the antitumor activity of ginsenoside liposomes against GC, MTT, cell cycle, and apoptosis assays were performed on BGC-823 cells in vitro and PTX-loaded ginsenoside liposomes were prepared to evaluate the therapeutic efficacy on GC in vivo. Results: The ginsenosides stabilized the liposomes in a manner similar to cholesterol. We confirmed the successful delivery of the bioactive combination drugs and internalization into GC cells via analysis of the glucose-related transporter recognition and longer blood circulation time. PTX was encapsulated in different liposomal formulations for use as a combination therapy, in which ginsenosides were found to exert their inherent anticancer activity, as well as act synergistically with PTX. The combination therapy using these targeted liposomes significantly suppressed GC tumor growth and outperformed most reported PTX formulations, including Lipusu® and Abraxane®. Conclusion: We established novel ginsenoside-based liposomes as a tumor-targeting therapy, in which ginsenoside functioned not only as a chemotherapy adjuvant, but also as a functional membrane material. Ginsenoside-based liposomes offer a novel platform for anticancer drug delivery and may lead to a new era of nanocarrier treatments for cancer.


Subject(s)
Drug Delivery Systems , Ginsenosides/therapeutic use , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Combined Modality Therapy , Endocytosis , Ginsenosides/chemistry , Ginsenosides/pharmacology , Humans , Liposomes , Mice, Nude , Paclitaxel/therapeutic use
18.
Molecules ; 23(9)2018 Aug 22.
Article in English | MEDLINE | ID: mdl-30135411

ABSTRACT

Background: Ginsenoside Rg5 has been proved to have a wide range of pharmacological activities. However, the in vitro and in vivo metabolism pathways of ginsenosides are still unclear, which impedes the understanding of their in vivo fate. In this paper, the possible metabolic process of Rg5 was studied and the metabolites are identified. Methods: Samples from rat liver microsomes (RLMs) in vitro and from rat urine, plasma and feces in vivo were collected for analysis after oral administration of Rg5. A rapid analysis technique using ultra-performance liquid chromatography (UPLC)/quadrupole-time-of-flight mass spectrometry (QTOF-MS) was applied for detecting metabolites of Rg5 both in vitro and in vivo. Results: A feasible metabolic pathway was proposed and described for ginsenoside Rg5. A total of 17 metabolic products were detected in biological samples, including the RLMs (four), rat urine (two), feces (13) and plasma (four). Fifteen of them have never been reported before. Oxidation, deglycosylation, deoxidation, glucuronidation, demethylation and dehydration were found to be the major metabolic reactions of Rg5. Conclusions: The present study utilized a reliable and quick analytical tool to explore the metabolism of Rg5 in rats and provided significant insights into the understanding of the metabolic pathways of Rg5 in vitro and in vivo. The results could be used to not only evaluate the efficacy and safety of Rg5, but also identify potential active drug candidates from the metabolites.


Subject(s)
Chromatography, Liquid , Ginsenosides/metabolism , Ginsenosides/pharmacokinetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Animals , Ginsenosides/administration & dosage , Ginsenosides/chemistry , Metabolic Networks and Pathways , Metabolomics/methods , Molecular Structure , Rats , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
19.
Spectrochim Acta A Mol Biomol Spectrosc ; 192: 93-100, 2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29126014

ABSTRACT

As a kind of common prescriptions, Shaoyao-Gancao-Tang (SGT) contains two Chinese herbs with four different proportions which have different clinical efficacy because of their various components. In order to investigate the herb-herb interaction mechanisms, we used the method of tri-level infrared macro-fingerprint spectroscopy to evaluate the concentration change of active components of four SGTs in this research. Fourier transform infrared spectroscopy (FT-IR) and Second derivative infrared spectroscopy (SD-IR) can recognize the multiple prescriptions directly and simultaneously. 2D-IR spectra enhance the spectral resolution and obtain much new information for discriminating the similar complicated samples of SGT. Furthermore, the whole analysis method from the analysis of the main components to the specific components and the relative content of the components may evaluate the quality of TCM better. Then we concluded that paeoniflorin and glycyrrhizic acid were the highest proportion in active ingredients in SGT-12:1 and the lowest one in SGT-12:12, which matched the HPLC-DAD results. It is demonstrated that the method composed by the tri-level infrared macro-fingerprint spectroscopy and the whole analysis can be applicable for effective, visual and accurate analysis and identification of very complicated and similar mixture systems of traditional Chinese medicine.


Subject(s)
Drug Compounding , Drugs, Chinese Herbal/chemistry , Glucosides/analysis , Glycyrrhizic Acid/analysis , Monoterpenes/analysis , Spectroscopy, Fourier Transform Infrared/methods , 1-Butanol/chemistry
20.
Theranostics ; 7(13): 3260-3275, 2017.
Article in English | MEDLINE | ID: mdl-28900508

ABSTRACT

Delivery of macromolecular drugs to the brain is impeded by the blood brain barrier. The recruitment of leukocytes to lesions in the brain, a typical feature of neuroinflammation response which occurs in cerebral ischemia, offers a unique opportunity to deliver drugs to inflammation sites in the brain. In the present study, cross-linked dendrigraft poly-L-lysine (DGL) nanoparticles containing cis-aconitic anhydride-modified catalase and modified with PGP, an endogenous tripeptide that acts as a ligand with high affinity to neutrophils, were developed to form the cl PGP-PEG-DGL/CAT-Aco system. Significant binding efficiency to neutrophils, efficient protection of catalase enzymatic activity from degradation and effective transport to receiver cells were revealed in the delivery system. Delivery of catalase to ischemic subregions and cerebral neurocytes in MCAO mice was significantly enhanced, which obviously reducing infarct volume in MCAO mice. Thus, the therapeutic outcome of cerebral ischemia was greatly improved. The underlying mechanism was found to be related to the inhibition of ROS-mediated apoptosis. Considering that neuroinflammation occurs in many neurological disorders, the strategy developed here is not only promising for treatment of cerebral ischemia but also an effective approach for various CNS diseases related to inflammation.


Subject(s)
Brain Ischemia/drug therapy , Drug Delivery Systems , Macromolecular Substances/therapeutic use , Nanoparticles/chemistry , Neutrophils/metabolism , Aconitic Acid/analogs & derivatives , Aconitic Acid/chemistry , Animals , Brain/pathology , Brain Ischemia/complications , Brain Ischemia/pathology , Catalase/metabolism , Cell Communication , Cell Death , Cell Differentiation , Dendrimers/chemistry , Endocytosis , Exosomes/metabolism , HL-60 Cells , Humans , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Mice, Nude , Nanoparticles/ultrastructure , Peptides/metabolism , Polymers/chemical synthesis , Polymers/chemistry , Proton Magnetic Resonance Spectroscopy , Reperfusion Injury/complications , Reperfusion Injury/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...