Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Chemistry ; 29(42): e202301014, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37195142

ABSTRACT

O3-type Fe/Mn-based layered oxide cathode materials with abundant reserves have a promising prospect in sodium-ion batteries. However, the electrochemical reversibility of most O3-type Fe/Mn-based oxide cathode materials is still not high enough. Herein, the effect of different Cu contents on the electrochemical properties of O3-NaFe0.50 Mn0.50 O2 materials is systematically investigated. The as-prepared NaFe0.30 Mn0.50 Cu0.20 O2 cathode achieves the synergistic optimization of the interface and bulk phase. It shows superior electrochemical performance, with an initial discharge specific capacity of 114 mAh g-1 at 0.1 C, a capacity retention rate of 94 % after 100 cycles at 0.5 C, and excellent chemical stability in air and water. In addition, the sodium ion full battery based on NaFe0.30 Mn0.50 Cu0.20 O2 cathode and hard carbon anode has a capacity retention rate of 81 % after 100 cycles. This research provides a useful approach for the preparation of low-cost and high-performance O3-type layered cathode materials.

2.
Molecules ; 27(20)2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36296726

ABSTRACT

Metastasis is an important cause of cancer-related death. Previous studies in our laboratory found that pregnane alkaloids from Pachysandra terminalis had antimetastatic activity against breast cancer cells. In the current study, we demonstrated that treatment with one of the alkaloid derivatives, (Z)-3ß-ethylamino-pregn-17(20)-en (1), led to the downregulation of the HIF-1α/VEGF/VEGFR2 pathway, suppressed the phosphorylation of downstream molecules Akt, mTOR, FAK, and inhibited breast cancer metastasis and angiogenesis both in vitro and in vivo. Furthermore, the antimetastasis and antiangiogenesis effects of 1 treatment (40 mg/kg) were more effective than that of Sorafenib (50 mg/kg). Surface plasmon resonance (SPR) analysis was performed and the result suggested that HSP90α was a direct target of 1. Taken together, our results suggested that compound 1 might represent a candidate antitumor agent for metastatic breast cancer.


Subject(s)
Alkaloids , Antineoplastic Agents , Triple Negative Breast Neoplasms , Humans , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism , Triple Negative Breast Neoplasms/drug therapy , Sorafenib/therapeutic use , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Antineoplastic Agents/pharmacology , TOR Serine-Threonine Kinases/metabolism , Alkaloids/pharmacology , Alkaloids/therapeutic use , Pregnanes/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit
3.
Bioorg Chem ; 115: 105201, 2021 10.
Article in English | MEDLINE | ID: mdl-34329994

ABSTRACT

Breast cancer has become the number one killer of women. In our previous study, an active compound, ION-31a, with potential anti-metastasis activity against breast cancer was identified through the synthesis of ionone alkaloid derivatives. In the present study, we aimed to identify the therapeutic target of ION-31a. We used a fluorescence tag labeled probe, molecular docking simulation, and surface plasmon resonance (SPR) analysis to identify the target of ION-31a. The main target of ION-31a was identified as heat shock protein 90 (HSP90). Thus, ION-31a is a novel HSP90 inhibiter that could suppress the metastasis of breast cancer and angiogenesis significantly in vitro and in vivo. ION-31a acts via inhibiting the HSP90/hypoxia inducible factor 1 alpha (HIF-1α)/vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) pathway and downregulating downstream signal pathways, including protein kinase B (AKT)/mammalian target of rapamycin (mTOR), AKT2/protein kinase C epsilon (PKCζ), extracellular regulated kinase 1/2 (ERK1/2), focal adhesion kinase (FAK), and mitogen-activated protein kinase 14 (p38MAPK) pathways. ION-31a affects multiple effectors implicated in tumor metastasis and has the potential to be developed as an anti-metastatic agent to treat patients with breast cancer.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Norisoprenoids/pharmacology , Alkaloids/chemical synthesis , Alkaloids/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Structure , Norisoprenoids/chemical synthesis , Norisoprenoids/chemistry , Structure-Activity Relationship , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
4.
Bioconjug Chem ; 29(2): 437-444, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29300459

ABSTRACT

Small molecule modified anticancer drug conjugates (SMMDCs) can self-assemble into nanoparticles (NPs) as therapeutic NP platforms for cancer treatment. Here we demonstrate that the XlogP and Hansen solubility parameters of paclitaxel (PTX) SMMDCs is essential for SMMDCs self-assembling into NPs. The amorphous state of PTX SMMDCs will also affect SMMDCs self-assembling into NPs. However, the antitumor activity of these PTX SMMDCs NPs decreased along with their XlogP values, indicating that a suitable XlogP value for designing the SMMDCs is important for self-assembling into NPs and for possessing antitumor activity. For higher level XlogP SMMDCs, a degradable linker should be considered in the design of SMMDCs to overcome the problem of lower antitumor activity. It is preferable that the hydrophilic groups in the SMMDCs should be present on the surface of self-assembling NPs.


Subject(s)
Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Paclitaxel/analogs & derivatives , Small Molecule Libraries/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Survival/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Paclitaxel/pharmacology , Small Molecule Libraries/pharmacology , Solubility
5.
AAPS PharmSciTech ; 19(2): 934-940, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29079988

ABSTRACT

Here, the mesoporous silica (Sylysia 350) was selected as mesoporous material, hydroxypropyl methylcellulose (HPMC) was selected as crystallization inhibitor, and febuxostat (FBT) was selected as model drug, respectively. The FBT-Sylysia-HPMC nanomatrix (FBT@SHN) was prepared. The characteristics of FBT@SHN were investigated in vitro and in vivo. Our results indicated that the FBT in FBT@SHN was in amorphous form. The solubility and dissolution of FBT in FBT@SHN were significantly increased. The oral bioavailability of FBT in FBT@SHN was greatly improved 5.8-fold compared with that in FBT suspension. This nanomatrix could be used as a drug delivery platform for improving the oral bioavailability.


Subject(s)
Febuxostat/chemistry , Febuxostat/metabolism , Nanostructures/chemistry , Polymers/chemistry , Polymers/metabolism , Administration, Oral , Animals , Biological Availability , Crystallization , Drug Delivery Systems/methods , Febuxostat/administration & dosage , Gout Suppressants/administration & dosage , Gout Suppressants/chemistry , Gout Suppressants/metabolism , Hypromellose Derivatives/administration & dosage , Hypromellose Derivatives/chemistry , Hypromellose Derivatives/metabolism , Male , Methylcellulose/chemistry , Nanostructures/administration & dosage , Polymers/administration & dosage , Rats , Rats, Sprague-Dawley , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Silicon Dioxide/metabolism , Solubility
6.
Eur J Pharmacol ; 965: 176330, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38220139

ABSTRACT

Pancreatic cancer (PC) is the most frequently occurring cancer, with few effective treatments and a 5-year survival rate of only about 11%. It is characterized by stiff interstitium and pressure on blood vessels, leading to an increased glycolytic metabolism. PFKFB3 plays an important role in glycolysis, and its products (fructose-2,6-bisphosphate), which are allosteric PFK1 activators, limit the glycolytic rate. In this study, 14 PFKFB3 inhibitors were obtained by virtually screening the FDA-approved compound library. Subsequently, the in-vitro investigations confirmed that Lomitapide and Cabozantinib S-malate exhibit the excellent potential to inhibit PFKFB3. The combined administration of Lomitapide and Gemcitabine at a certain molar ratio indicated an enhanced anti-tumor effect in Orthotopic Pancreatic Cancer (OPC) models. This investigation provides a new treatment strategy for PC therapy.


Subject(s)
Pancreatic Neoplasms , Phosphofructokinase-2 , Humans , Phosphofructokinase-2/metabolism , Drug Repositioning , Early Detection of Cancer , Phosphoric Monoester Hydrolases/metabolism , Pancreatic Neoplasms/drug therapy , Glycolysis
7.
Int J Nanomedicine ; 18: 1281-1298, 2023.
Article in English | MEDLINE | ID: mdl-36945256

ABSTRACT

Purpose: Chemotherapy is a significant and effective therapeutic strategy that is frequently utilized in the treatment of cancer. Small molecular prodrug-based nanoassemblies (SMPDNAs) combine the benefits of both prodrugs and nanomedicine into a single nanoassembly with high drug loading, increased stability, and improved biocompatibility. Methods: In this study, a disulfide bond inserted 7-ethyl-10-hydroxycamptothecin (SN38) prodrug was rationally designed and then used to prepare nanoassemblies (SNSS NAs) that were selectively activated by rich glutathione (GSH) in the tumor site. The characterization of SNSS NAs and the in vitro and in vivo evaluation of their antitumor effect on a pancreatic cancer model were performed. Results: In vitro findings demonstrated that SNSS NAs exhibited GSH-induced SN38 release and cytotoxicity. SNSS NAs have demonstrated a passive targeting effect on tumor tissues, a superior antitumor effect compared to irinotecan (CPT-11), and satisfactory biocompatibility with double dosage treatment. Conclusion: The SNSS NAs developed in this study provide a new method for the preparation of SN38-based nano-delivery systems with improved antitumor effect and biosafety.


Subject(s)
Nanoparticles , Pancreatic Neoplasms , Prodrugs , Humans , Prodrugs/chemistry , Drug Liberation , Irinotecan/pharmacology , Drug Delivery Systems/methods , Pancreatic Neoplasms/drug therapy , Disulfides/chemistry , Cell Line, Tumor , Nanoparticles/chemistry , Camptothecin , Pancreatic Neoplasms
8.
Drug Deliv ; 28(1): 800-813, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33866918

ABSTRACT

Cancer immunotherapy is a strategy that is moving to the frontier of cancer treatment in the current decade. In this study, we show evidence that 3-(2-nitrophenyl) propionic acid-paclitaxel nanoparticles (NPPA-PTX NPs), act as immunogenic cell death (ICD) inducers, stimulating an antitumor response which results in synergistic antitumor activity by combining anti-PD-L1 antibody (aPD-L1) in vivo. To investigate the antitumor immunity induced by NPPA-PTX NPs, the expression of both ICD marker calreticulin (CRT) and high mobility group box 1 (HMGB1) were analyzed. In addition, the antitumor activity of NPPA-PTX NPs combined with aPD-L1 in vivo was also investigated. The immune response was also measured through quantitation of the infiltration of T cells and the secretion of pro-inflammatory cytokines. The results demonstrate that NPPA-PTX NPs induce ICD of MDA-MB-231 and 4T1 cells through upregulation of CRT and HMGB1, reactivating the antitumor immunity via recruitment of infiltrating CD3+, CD4+, CD8+ T cells, secreting IFN-γ, TNF-α, and the enhanced antitumor activity by combining with aPD-L1. These data suggest that the combined therapy has a synergistic antitumor activity and has the potential to be developed into a novel therapeutic regimen for cancer patients.


Subject(s)
Albumins/pharmacology , Antineoplastic Agents/pharmacology , Immunogenic Cell Death/drug effects , Inflammation Mediators/metabolism , Nanoparticles/chemistry , Paclitaxel/pharmacology , Albumins/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , B7-H1 Antigen/immunology , Calreticulin/drug effects , Cell Line, Tumor , Female , HMGB1 Protein/drug effects , Humans , Mice , Mice, Inbred BALB C , Paclitaxel/administration & dosage , Propionates/chemistry , Up-Regulation , Xenograft Model Antitumor Assays
9.
Int J Nanomedicine ; 15: 1809-1821, 2020.
Article in English | MEDLINE | ID: mdl-32214813

ABSTRACT

INTRODUCTION: Because tumor-associated inflammation is a hallmark of cancer treatment, in the present study, sorafenib mesoporous silica nanomatrix (MSNM@SFN) co-administrated with flufenamic acid (FFA, a non-steroidal anti-inflammatory drug (NSAID)) was investigated to enhance the anti-tumor activity of MSNM@SFN. METHODS: Metastatic breast tumor 4T1/luc cells and hepatocellular carcinoma HepG2 cells were selected as cell models. The effects of FFA in vitro on cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in 4T1/luc and HepG2 cells were investigated. The in vivo anti-tumor activity of MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) was evaluated in a 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model, respectively. RESULTS: The results indicated that FFA could markedly decrease cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in both 4T1/luc and HepG2 cells. The enhanced anti-tumor activity of MSNM@SFN+FFA compared with that of MSNM@SFN was confirmed in the 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model in vivo, respectively. DISCUSSION: MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) developed in this study is an alternative strategy for improving the therapeutic efficacy of MSNM@SFN via co-administration with NSAIDs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , 20-Hydroxysteroid Dehydrogenases/metabolism , Aldo-Keto Reductase Family 1 Member C3/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Dinoprostone/metabolism , Female , Flufenamic Acid/administration & dosage , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Nanostructures/administration & dosage , Nanostructures/chemistry , Silicon Dioxide/chemistry , Sorafenib/administration & dosage , Xenograft Model Antitumor Assays
10.
Int J Nanomedicine ; 14: 195-204, 2019.
Article in English | MEDLINE | ID: mdl-30636872

ABSTRACT

BACKGROUND: 3-(2-Nitrophenyl) propionic acid-paclitaxel (NPPA-PTX) is a paclitaxel (PTX) bioreductive prodrug synthesized by our lab. We hypothesize that NPPA-PTX can self-assemble to form nanoparticles (NPs). MATERIALS AND METHODS: In the present research, the theoretical partition coefficient (XlogP) and Hansen solubility parameters of NPPA-PTX were calculated. NPPA-PTX nanoparticles prepared by NPPA-PTX and DSPE-PEG (NPPA-PTX:DSPE-PEG =1:0.1, w/w) (NPPA-PTX@PEG NPs) were prepared and characterized. The cellular uptake, in vitro antitumor activity, in vivo targeting effect, tumor distribution, in vivo antitumor activity, and safety of NPPA-PTX@PEG NPs were investigated. RESULTS: Our results indicate that NPPA-PTX can self-assemble to form NPPA-PTX@PEG NPs. Both the cellular uptake and safety of NPPA-PTX@PEG NPs were higher than those of Taxol. NPPA-PTX@PEG NPs could target tumor tissues by a passive targeting effect. In tumor tissues, NPPA-PTX@PEG NPs could completely transform into active PTX. The in vivo antitumor activity of NPPA-PTX@PEG NPs was confirmed in MDA-MB-231 tumor-bearing nude mice. CONCLUSION: The bioreductive prodrug NPPA-PTX could self-assemble to form NPs. The safety and antitumor activity of NPPA-PTX@PEG were confirmed in our in vitro and in vivo experiments. The NPPA-PTX@PEG NPs developed in this study could offer a new way of preparing bioreductive prodrug, self-assembled NPs suitable for antitumor therapy.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation/drug effects , Nanoparticles/administration & dosage , Paclitaxel/analogs & derivatives , Phenylpropionates/pharmacology , Prodrugs/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Female , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Phenylpropionates/administration & dosage , Prodrugs/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
J Control Release ; 295: 102-117, 2019 02 10.
Article in English | MEDLINE | ID: mdl-30582952

ABSTRACT

Nucleus-targeting drug delivery systems (NTDDs) deliver chemotherapeutic agents to nuclei in order to improve the efficacy of anti-tumour therapy. Histone H1 (H1) plays a key role in establishing and maintaining higher order chromatin structures and could bind to cell membranes. In the present study, we selected H1 as a target to prepare a novel H1-mediated NTDD. Low molecular weight heparin (LMHP) and doxorubicin (DOX) were combined to form LMHP-DOX. Then, a novel NTDD consisting of LMHP-DOX nanoparticles (LMHP-DOX NPs) was prepared by self-assembly. The characteristics of LMHP-DOX and LMHP-DOX NPs were investigated. Histone H1 high-expressive prostate cancer PC-3M cell line was selected as the cell model. Cellular uptake, and the in vitro and in vivo anti-tumour activity of LMHP-DOX NPs were evaluated on H1 high-expressive human prostate cancer PC-3M cells. Our results indicated that intact LMHP-DOX NPs mediated by H1 could be absorbed by H1 high-expressive PC-3M cells, escape from the lysosomes to the cytoplasm, and localize in the perinuclear region via H1-mediated, whereby DOX could directly enter the cell nucleus and quickly increase the concentration of DOX in the nuclei of H1 high-expressive PC-3M cells to enhance the apoptotic activity of cancer cells. The anti-coagulant activity of LMHP-DOX NPs was almost completely diminished in rat blood compared with that of LMHP, indicating the safety of LMHP-DOX NPs. Compared to traditional NTDD strategies, LMHP-DOX NPs avoid the complicated modification of nucleus-targeting ligands and provide a compelling solution for the substantially enhanced nuclear uptake of chemotherapeutic agents for the development of more intelligent NTDDs.


Subject(s)
Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Heparin, Low-Molecular-Weight/administration & dosage , Histones/analysis , Prostatic Neoplasms/drug therapy , Animals , Anticoagulants/administration & dosage , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Delivery Systems , Drug Liberation , Heparin, Low-Molecular-Weight/pharmacokinetics , Heparin, Low-Molecular-Weight/pharmacology , Humans , Male , Nanoparticles/ultrastructure , PC-3 Cells , Prostatic Neoplasms/pathology , Rats, Sprague-Dawley
12.
Int J Nanomedicine ; 13: 1495-1504, 2018.
Article in English | MEDLINE | ID: mdl-29559778

ABSTRACT

BACKGROUND: In the present study, the tumor-specific, pH-responsive peptide H7K(R2)2-modified, theranostic liposome-containing paclitaxel (PTX) and superparamagnetic iron oxide nanoparticles (SPIO NPs), PTX/SPIO-SSL-H7K(R2)2, was prepared by using H7K(R2)2 as the targeting ligand, SPIO NPs as the magnetic resonance imaging (MRI) agent, PTX as antitumor drug. METHODS: The PTX/SPIO-SSL-H7K(R2)2 was prepared by a thin film hydration method. The characteristics of PTX/SPIO-SSL-H7K(R2)2 were evaluated. The targeting effect, MRI, and antitumor activity of PTX/SPIO-SSL-H7K(R2)2 were investigated detail in vitro and in vivo in human breast carcinoma MDA-MB-231 cell models. RESULTS: Our results of in vitro flow cytometry, in vivo imaging, and in vivo MR imaging confirmed the pH-responsive characteristic of H7K(R2)2 in MDA-MB-231 cell line in vitro and in vivo. The results of in vivo MRI and in vivo antitumor activity confirmed the theranostic effect of PTX/SPIO-SSL-H7K(R2)2 in MDA-MB-231 tumor-bearing model. CONCLUSION: Considering all our in vitro and in vivo results, we conclude that we developed targeting modified theranostic liposome which could achieve both role of antitumor and MRI.


Subject(s)
Ferric Compounds/chemistry , Magnetite Nanoparticles/chemistry , Neoplasms/drug therapy , Paclitaxel/therapeutic use , Peptides/chemistry , Theranostic Nanomedicine/methods , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Drug Liberation , Female , Flow Cytometry , Humans , Hydrogen-Ion Concentration , Liposomes , Magnetic Resonance Imaging , Mice, Inbred BALB C , Mice, Nude , Neoplasms/pathology , Paclitaxel/pharmacology , Tissue Distribution/drug effects
13.
Drug Deliv ; 24(1): 270-277, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28165798

ABSTRACT

In the present study, we select the Sylysia 350 (Sylysia) as mesoporous material, distearoylphosphatidylethanolamine-poly(ethylene glycol)2000 (DSPE-PEG) as absorption enhancer and hydroxy propyl methyl cellulose (HPMC) as crystallization inhibitor to prepare sorafenib tosylate (SFN) nanomitrix (MSNM@SFN) for improving the anti-tumor activity of SFN. The MSNM@SFN was prepared by solvent evaporation method. The solubility, dissolution, and bioavailability of SFN in MSNM@SFN were also investigated. The anti-tumor activity of MSNM@SFN was evaluated in vitro and in vivo. Our results indicated that the solubility and dissolution of SFN in MSNM@SFN were significantly increased. The oral bioavailability of SFN in MSNM@SFN was greatly improved 7.7-fold compared with that in SFN suspension. The enhanced anti-tumor activity of MSNM@SFN was confirmed in vitro and in vivo experiments. This nanomatrix developed in this study could be a promising drug delivery platform for improving the therapeutic efficacy of poorly water-soluble drugs.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Nanoparticles/administration & dosage , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Polymers/administration & dosage , Tumor Burden/drug effects , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Female , Humans , Lactose/administration & dosage , Lactose/analogs & derivatives , Lactose/chemistry , Male , Methylcellulose/administration & dosage , Methylcellulose/analogs & derivatives , Methylcellulose/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Niacinamide/administration & dosage , Niacinamide/chemistry , Phenylurea Compounds/chemistry , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polymers/chemistry , Random Allocation , Rats , Rats, Sprague-Dawley , Sorafenib , Tumor Burden/physiology
14.
Oncotarget ; 7(30): 48467-48480, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27366947

ABSTRACT

Hypoxia is an important microenvironmental pressure present in the majority of solid tumors and, so, tumor hypoxia might be considered an attractive target for tumor therapy. One strategy for targeting hypoxia is to develop bioreductive prodrugs. In the present research, we synthesized a bioreductive paclitaxel prodrug, 3-(2-Nitrophenyl) propionic acid-paclitaxel (NPPA-PTX). The stability of NPPA-PTX in PBS and rat plasma was investigated. The anti-tumor activity of NPPA-PTX was also evaluated in vitro and in vivo. The results of our stability study indicated that NPPA-PTX was stable in PBS and rat plasma as well as in the blood circulation. The in vitro and in vivo anti-tumor activity of NPPA-PTX was confirmed in both KB cells and MDA-MB-231 cells. Our results also indicated that NPPA-PTX could completely convert to active PTX in tumor tissues and produced the anti-tumor activity in both KB and MDA-MB-231 tumor-bearing nude mice. We suggest that the dissociated PTX which converted from NPPA-PTX in tumor tissues played a key role in producing anti-tumor activity. Considering all our results, we suggest that NPPA-PTX is a novel bioreductive PTX prodrug which could undergo further evaluation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Phenylpropionates/pharmacology , Prodrugs/pharmacology , Tumor Hypoxia/drug effects , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Stability , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Paclitaxel/chemical synthesis , Paclitaxel/therapeutic use , Phenylpropionates/chemical synthesis , Phenylpropionates/therapeutic use , Prodrugs/chemical synthesis , Prodrugs/therapeutic use , Propionates/chemical synthesis , Propionates/pharmacology , Propionates/therapeutic use , Rats , Rats, Sprague-Dawley
15.
Oncotarget ; 7(28): 43616-43628, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27283987

ABSTRACT

Like the anti-angiogenic strategy, anti-vascular mimicry is considered as a novel targeting strategy for glioma. In the present study, we used NGR as a targeting ligand and prepared NGR-modified liposomes containing combretastatin A4 (NGR-SSL-CA4) in order to evaluate their potential targeting of glioma tumor cells and vasculogenic mimicry (VM) formed by glioma cells as well as their anti-VM activity in mice with glioma tumor cells. NGR-SSL-CA4 was prepared by a thin-film hydration method. The in vitro targeting of U87-MG (human glioma tumor cells) by NGR-modified liposomes was evaluated. The in vivo targeting activity of NGR-modified liposomes was tested in U87-MG orthotopic tumor-bearing nude mice. The anti-VM activity of NGR-SSL-CA4 was also investigated in vitro and in vivo. The targeting activity of the NGR-modified liposomes was demonstrated by in vitro flow cytometry and in vivo biodistribution. The in vitro anti-VM activity of NGR-SSL-CA4 was indicated in a series of cell migration and VM channel experiments. NGR-SSL-CA4 produced very marked anti-tumor and anti-VM activity in U87-MG orthotopic tumor-bearing mice in vivo. Overall, the NGR-SSL-CA4 has great potential in the multi-targeting therapy of glioma involving U87-MG cells, and the VM formed by U87-MG cells as well as endothelial cells producing anti-U87-MG cells, and anti-VM formed by U87-MG cells as well as anti-endothelial cell activity.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Liposomes/chemistry , Stilbenes/therapeutic use , Animals , Apoptosis/drug effects , Brain/blood supply , Brain/diagnostic imaging , Brain/pathology , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Flow Cytometry , Glioma/diagnostic imaging , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Multiple Chronic Conditions , Neovascularization, Pathologic/drug therapy , Oligopeptides/chemistry , Optical Imaging , Tissue Distribution , Xenograft Model Antitumor Assays
16.
Sci Rep ; 6: 36614, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27812039

ABSTRACT

The main objective of this study was to demonstrate the proof-of-principle for the hypothesis that conjugated linoleic acid-paclitaxel conjugate (CLA-PTX), a novel fatty acid modified anti-cancer drug conjugate, could self-assemble forming nanoparticles. The results indicated that a novel self-assembling nanomedicine, CLA-PTX@PEG NPs (about 105 nm), with Cremophor EL (CrEL)-free and organic solvent-free characteristics, was prepared by a simple precipitation method. Being the ratio of CLA-PTX:DSPE-PEG was only 1:0.1 (w/w), the higher drug loading CLA-PTX@PEG NPs (about 90%) possessed carrier-free characteristic. The stability results indicated that CLA-PTX@PEG NPs could be stored for at least 9 months. The safety of CLA-PTX@PEG NPs was demonstrated by the MTD results. The anti-tumor activity and cellular uptake were also confirmed in the in vitro experiments. The lower crystallinity, polarity and solubility of CLA-PTX compared with that of paclitaxel (PTX) might be the possible reason for CLA-PTX self-assembling forming nanoparticles, indicating a relationship between PTX modification and nanoparticles self-assembly. Overall, the data presented here confirm that this drug self-delivery strategy based on self-assembly of a CLA-PTX conjugate may offer a new way to prepare nanomedicine products for cancer therapy involving the relationship between anticancer drug modification and self-assembly into nanoparticles.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Linoleic Acids, Conjugated/chemistry , Nanomedicine , Paclitaxel/chemistry , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Carriers , Drug Screening Assays, Antitumor , Female , Linoleic Acids, Conjugated/administration & dosage , Male , Mice , Mice, Inbred ICR , Microscopy, Electron, Transmission , Paclitaxel/administration & dosage , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL