Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Mol Diagn Ther ; 27(6): 769-779, 2023 11.
Article in English | MEDLINE | ID: mdl-37689607

ABSTRACT

OBJECTIVE: We aimed to evaluate the clinical performance of expanded noninvasive prenatal testing (NIPT-Plus) for the detection of aneuploidies and microdeletion/microduplication syndromes. METHODS: A total of 7177 pregnant women were enrolled in the study from June 2020 to March 2022 at Xijing Hospital, China. Cases with NIPT-Plus-positive results were further confirmed by chromosomal karyotyping and a chromosomal microarray analysis. RESULTS: A total of 112 positive cases (1.56%) were identified by NIPT-Plus, including 60 chromosome aneuploidies and 52 microdeletion/microduplication syndromes. Ninety-five cases were validated by amniocentesis, and 57 were confirmed with true-positive results, comprising 18 trisomy 21, 4 trisomy 18, 1 trisomy 13, 17 sex chromosome aneuploidies, 1 other aneuploidy, and 16 microdeletion/microduplication syndromes. The positive predictive value of total chromosomal abnormalities was 60% (57/95). For trisomy 21, trisomy 18, trisomy 13, sex chromosome aneuploidies, other aneuploidies and microdeletion/microduplication syndromes, the sensitivity was all 100%, the specificity was 100, 99.986, 100, 99.888, 99.958, and 99.636%, and the positive predictive value was 100, 80, 100, 68, 25, and 38.10%, respectively. For all clinical characteristics, the abnormal maternal serum screening group was found to have the highest prevalence of chromosomal abnormalities (1.54%), and the ultrasound abnormality group presented the highest positive predictive value (73.33%). CONCLUSIONS: NIPT-Plus has great potential for the detection of aneuploidies and microdeletion/microduplication syndromes owing to its high sensitivity, safety, and specificity, which greatly reduces unnecessary invasive procedures and the risk of miscarriage and allows informed maternal choice.


Subject(s)
Down Syndrome , Noninvasive Prenatal Testing , Female , Pregnancy , Humans , Down Syndrome/diagnosis , Down Syndrome/genetics , Trisomy 18 Syndrome/diagnosis , Trisomy 18 Syndrome/genetics , Prenatal Diagnosis/methods , Trisomy 13 Syndrome/diagnosis , Trisomy 13 Syndrome/genetics , Aneuploidy , Chromosome Aberrations
2.
Theranostics ; 13(9): 2800-2810, 2023.
Article in English | MEDLINE | ID: mdl-37284456

ABSTRACT

Rationale: Broad-spectrum oncolytic peptides (Olps) constitute potential therapeutic options for treating heterogeneous triple-negative breast cancer (TNBC); however, their clinical application is limited owing to high toxicity. Methods: A nanoblock-mediated strategy was developed to induce selective anticancer activity of synthetic Olps. A synthetic Olp, C12-PButLG-CA, was conjugated to the hydrophobic or hydrophilic terminal of a poly(ethylene oxide)-b-poly(propylene oxide) nanoparticle or a hydrophilic poly(ethylene oxide) polymer. A nanoblocker, that can significantly reduce the toxicity of Olp, was screened out through hemolytic assay, and then Olps were conjugated to the nanoblock via a tumor acidity-cleavable bond to obtain the selective RNolp ((mPEO-PPO-CDM)2-Olp). The tumor acidity responsive membranolytic activity, in vivo toxicity and anti-tumor efficacy of RNolp were determined. Results: We found that the conjugation of Olps to the hydrophobic core of a nanoparticle but not the hydrophilic terminal or a hydrophilic polymer restricts their motion and drastically reduces their hemolytic activity. We then covalently conjugated Olps to such a nanoblock via a cleavable bond that can be hydrolyzed in the acidic tumor environment, yielding a selective RNolp molecule. At physiological pH (pH 7.4), RNolp remained stable with the Olps shielded by nanoblocks and exhibited low membranolytic activity. At the acidic tumor environment (pH 6.8), Olps could be released from the nanoparticles via the hydrolysis of the tumor acidity-cleavable bonds and exerted membranolytic activity against TNBC cells. RNolp is well tolerated in mice and demonstrated high antitumor efficacy in orthotopic and metastatic mouse models of TNBC. Conclusion: We developed a simple nanoblock-mediated strategy to induce a selective cancer therapy of Olps for TNBC.


Subject(s)
Nanoparticles , Triple Negative Breast Neoplasms , Humans , Mice , Animals , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Ethylene Oxide/therapeutic use , Peptides/chemistry , Nanoparticles/chemistry , Polymers/chemistry
3.
ACS Appl Mater Interfaces ; 15(18): 21781-21791, 2023 May 10.
Article in English | MEDLINE | ID: mdl-37115169

ABSTRACT

Antimicrobial polymers exhibit great potential for treating drug-resistant bacteria; however, designing antimicrobial polymers that can selectively kill bacteria and cause relatively low toxicity to normal tissues/cells remains a key challenge. Here, we report a pH window for ionizable polymers that exhibit high selectivity toward bacteria. Ionizable polymer PC6A showed the greatest selectivity (131.6) at pH 7.4, exhibiting low hemolytic activity and high antimicrobial activity against bacteria, whereas a very high or low protonation degree (PD) produced relatively low selectivity (≤35.6). Bactericidal mechanism of PC6A primarily comprised membrane lysis without inducing drug resistance even after consecutive incubation for 32 passages. Furthermore, PC6A demonstrated synergistic effects in combination with antibiotics at pH 7.4. Hence, this study provides a strategy for designing selective antimicrobial polymers.


Subject(s)
Anti-Bacterial Agents , Hydrogen-Ion Concentration , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Polymers/chemistry , Microbial Sensitivity Tests , Bacteria/drug effects
4.
Biomater Sci ; 11(4): 1451-1458, 2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36602031

ABSTRACT

Oncolytic peptides (OLPs) with membranolytic activity show great potential to combat multidrug-resistant cancer cells. Herein, we report a cationic helical oncolytic polypeptide (OLPP) with potent membranolytic activity for cancer therapy. The OLPP was synthesized by ring-opening polymerization of N-carboxyanhydrides (NCAs) and thiol-ene reaction. The OLPP was resistant to protease, showed high cytotoxicity to a series of cancer cells and caused cancer cell necrosis by quickly lysing cancer cell membrane independent of classic death-related intracellular pathways. Intra-tumoral injection of the OLPP effectively suppressed tumor growth in mice through the direct oncolytic effect. The OLPP represents a potential oncolytic chemotherapeutics for cancer therapy.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Animals , Mice , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Peptides , Neoplasms/therapy , Necrosis , Cell Line, Tumor
5.
Sci China Chem ; 65(6): 1010-1075, 2022.
Article in English | MEDLINE | ID: mdl-35505924

ABSTRACT

Biomedical polymers have been extensively developed for promising applications in a lot of biomedical fields, such as therapeutic medicine delivery, disease detection and diagnosis, biosensing, regenerative medicine, and disease treatment. In this review, we summarize the most recent advances in the synthesis and application of biomedical polymers, and discuss the comprehensive understanding of their property-function relationship for corresponding biomedical applications. In particular, a few burgeoning bioactive polymers, such as peptide/biomembrane/microorganism/cell-based biomedical polymers, are also introduced and highlighted as the emerging biomaterials for cancer precision therapy. Furthermore, the foreseeable challenges and outlook of the development of more efficient, healthier and safer biomedical polymers are discussed. We wish this systemic and comprehensive review on highlighting frontier progress of biomedical polymers could inspire and promote new breakthrough in fundamental research and clinical translation.

7.
Nat Nanotechnol ; 17(5): 541-551, 2022 05.
Article in English | MEDLINE | ID: mdl-35332294

ABSTRACT

Plasma membrane rupture is a promising strategy for drug-resistant cancer treatment, but its application is limited by the low tumour selectivity of membranolytic molecules. Here we report the design of 'proton transistor' nanodetergents that can convert the subtle pH perturbation signals of tumour tissues into sharp transition signals of membranolytic activity for selective cancer therapy. Our top-performing 'proton transistor' nanodetergent, P(C6-Bn20), can achieve a >32-fold change in cytotoxicity with a 0.1 pH input signal. At physiological pH, P(C6-Bn20) self-assembles into neutral nanoparticles with inactive membranolytic blocks shielded by poly(ethylene glycol) shells, exhibiting low toxicity. At tumour acidity, a sharp transition in its protonation state induces a morphological transformation and an activation of the membranolytic blocks, and the cation-π interaction facilitates the insertion of benzyl groups-containing hydrophobic domains into the cell membranes, resulting in potent membranolytic activity. P(C6-Bn20) is well tolerated in mice and shows high anti-tumour efficacy in various mouse tumour models.


Subject(s)
Nanoparticles , Neoplasms , Animals , Hydrogen-Ion Concentration , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Polyethylene Glycols/chemistry , Protons
8.
Nat Commun ; 12(1): 1359, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33649336

ABSTRACT

Modulating effector immune cells via monoclonal antibodies (mAbs) and facilitating the co-engagement of T cells and tumor cells via chimeric antigen receptor- T cells or bispecific T cell-engaging antibodies are two typical cancer immunotherapy approaches. We speculated that immobilizing two types of mAbs against effector cells and tumor cells on a single nanoparticle could integrate the functions of these two approaches, as the engineered formulation (immunomodulating nano-adaptor, imNA) could potentially associate with both cells and bridge them together like an 'adaptor' while maintaining the immunomodulatory properties of the parental mAbs. However, existing mAbs-immobilization strategies mainly rely on a chemical reaction, a process that is rough and difficult to control. Here, we build up a versatile antibody immobilization platform by conjugating anti-IgG (Fc specific) antibody (αFc) onto the nanoparticle surface (αFc-NP), and confirm that αFc-NP could conveniently and efficiently immobilize two types of mAbs through Fc-specific noncovalent interactions to form imNAs. Finally, we validate the superiority of imNAs over the mixture of parental mAbs in T cell-, natural killer cell- and macrophage-mediated antitumor immune responses in multiple murine tumor models.


Subject(s)
Antibodies, Monoclonal/metabolism , Immunomodulation , Immunotherapy , Nanoparticles/chemistry , Neoplasms/immunology , Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Immobilized Proteins/metabolism , Immunity , Killer Cells, Natural/immunology , Male , Mice, Inbred C57BL , Nanoparticles/ultrastructure , T-Lymphocytes/immunology
9.
Biomater Sci ; 8(24): 6858-6866, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32815940

ABSTRACT

Since the development of bacterial resistance, the decreasing effectiveness of antibiotics is becoming one of the most critical problems worldwide. Novel antibacterial agents are urgently needed to prevent humanity from falling back into the "post-antibiotic era". As an important part of the innate immune system, antimicrobial peptides (AMPs) are one of the most promising antibacterial agents showing broad-spectrum activity against bacteria and low propensity for drug resistance. However, the shortcomings of AMPs, such as high toxicity and easy digestion by proteases, limit their clinical application. This review mainly focuses on the effect of the secondary structure on the antimicrobial activity and cytotoxicity of AMPs and the strategies of designing conformationally transitionable AMPs with improved selectivity towards bacteria.


Subject(s)
Antimicrobial Cationic Peptides , Bacterial Infections , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bacteria , Humans , Pore Forming Cytotoxic Proteins
10.
ACS Nano ; 14(3): 3563-3575, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32053346

ABSTRACT

Polymeric nanoparticles (NPs) are an important category of drug delivery systems, and their in vivo fate is closely associated with delivery efficacy. Analysis of the protein corona on the surface of NPs to understand the in vivo fate of different NPs has been shown to be reliable but complicated and time-consuming. In this work, we establish a simple approach for predicting the in vivo fate of polymeric NPs. We prepared a series of poly(ethylene glycol)-block-poly(d,l-lactide) (PEG-b-PLA) NPs with different protein binding behaviors by adjusting their PEG densities, which were determined by analyzing the serum protein adsorption. We further determined the protein binding affinity, denoted as the equilibrium association constant (KA), to correlate with in vivo fate of NPs. The in vivo fate, including blood clearance and Kupffer cell uptake, was studied, and the maximum concentration (Cmax), the area under the plasma concentration-time curve (AUC), and the mean residence time (MRT) were negatively linearly dependent, while Kupffer cell uptake was positively linearly dependent on KA. Subsequently, we verified the reliability of the approach for in vivo fate prediction using poly(methoxyethyl ethylene phosphate)-block-poly(d,l-lactide) (PEEP-b-PLA) and poly(vinylpyrrolidone)-block-poly(d,l-lactide) (PVP-b-PLA) NPs, and the linear relationship between the KA value and their PK parameters further suggests that the protein binding affinity of polymeric NPs can be a direct indicator of their pharmacokinetics.


Subject(s)
Blood Proteins/chemistry , Nanoparticles/chemistry , Polymers/pharmacokinetics , Adsorption , Animals , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Nanoparticles/administration & dosage , Particle Size , Polymers/administration & dosage , Polymers/chemistry , Protein Binding , RAW 264.7 Cells , Surface Properties
11.
Biosens Bioelectron ; 142: 111531, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31401228

ABSTRACT

The use of new functional two-dimensional nanomaterials for construction of advanced biosensors has attracted great attention. Herein, we report an electrochemical DNA (E-DNA) biosensor to detect gliotoxin based on DNA nanostructure-modified MXene (Ti3C2) nanosheets. Tetrahedral DNA nanostructures (TDNs) were facilely immobilized onto the surface of MXene nanosheets through coordination interactions between the phosphate groups on DNA and titanium, which avoids cumbersome and expensive modification of DNA probes. MXene nanosheets possess large surface area to modify a large number of DNA probes and excellent conductivity to facilitate the electron transfer between electrochemical species and the underlying electrode surface. Meanwhile, the unique configuration of TDN enables efficient and rapid binding of target molecules onto electrode surface, thereby producing amplified electrochemical signals. Through combining the merits of the two nanomaterials, the proposed sensor exhibits a wide detection range from 5 pM to 10 nM with a low limit of detection (LOD) of 5 pM. We believe that this work opens a new avenue for development of MXene-based E-DNA biosensors and could be further extended to detect other mycotoxins.


Subject(s)
Biosensing Techniques/methods , DNA/chemistry , Gliotoxin/analysis , Nanostructures/chemistry , Titanium/chemistry , Electrochemical Techniques/methods , Limit of Detection , Nanostructures/ultrastructure
12.
Small ; 15(36): e1902022, 2019 09.
Article in English | MEDLINE | ID: mdl-31318147

ABSTRACT

The cell nucleus-targeted delivery of therapeutic agents plays a critical role in cancer therapy, since the biological target of many anticancer therapeutics is the cell nucleus. However, multiple physiological barriers limit the delivery efficiency of free drugs, resulting in unsatisfactory therapeutic effects. Herein, thioketal crosslinked polyphosphoester-based nanoparticles with a tumor acidity (pHe )-sensitive transactivator of transcription (TAT) peptide (DA-masked TAT-decorating reactive oxygen species (ROS)-sensitive Ce6/DOX-loaded hyperbranched nanoparticles (D TRCD)) are explored for cascade nucleus-targeted drug delivery. Following administration, D TRCD experiences prolonged circulation by masking the targeting effect of its TAT peptide and then achieves enhanced tumor cell uptake and improved translocation into the perinuclear region by reactivating the TAT targeting capability in tumor tissue. Subsequently, ROS generated by D TRCD under 660 nm laser not only disrupts the nuclear membrane to allow entry into the nuclei but also triggers intracellular release of the payload in the nuclei. As evidenced by in vivo experiments, such pHe /photo dual-sensitive polymeric nanocarriers offer remarkable therapeutic effects, efficiently suppressing tumor growth. This multistage cascade nucleus-targeted drug delivery concept provides new avenues to develop nucleus-targeted drug delivery systems.


Subject(s)
Cell Nucleus/metabolism , Nanoparticles/chemistry , Neoplasms/drug therapy , Polymers/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Drug Delivery Systems/methods , Humans , Hydrogen-Ion Concentration , Reactive Oxygen Species
13.
Colloids Surf B Biointerfaces ; 180: 313-318, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31071571

ABSTRACT

Photodynamic therapy (PDT) is a promising procedure for breast cancer therapy. Curcumin (Cur), a hydrophobic polyphenol derived from the spice turmeric, has been considered as a potential photosensitizer for PDT with evoked immune response, excellent safety, and low cost. However, the translation of curcumin in clinical cancer therapy suffers from an insufficient therapeutic dose in tumor tissues due to its poor solubility and low bioavailability. In this study, carrier-free curcumin nanodrugs (Cur NDs) were prepared without using any toxic solvents through a facile and green reprecipitation method. Cur NDs exhibited distinct optical properties, light-sensitive drug release behavior, resulting in increased reactive oxygen species (ROS) generation and PDT efficacy on breast cancer cells compared with free Cur. Furthermore, cell apoptosis during Cur-based PDT was concomitant with the activation of the ROS-mediated JNK/caspase-3 signaling pathway. Overall, our carrier-free Cur nanodrugs may be promising candidates for facilitating the efficacy and safety of PDT against breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Curcumin/therapeutic use , Drug Carriers/chemistry , Green Chemistry Technology/methods , Light , Nanoparticles/chemistry , Photochemotherapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Curcumin/pharmacology , Drug Liberation , Female , Mice , Nanoparticles/ultrastructure , Reactive Oxygen Species/metabolism
14.
Nano Lett ; 19(4): 2688-2693, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30844291

ABSTRACT

The inherent features of small interfering RNA (siRNA), including a relatively high molecular weight, negative charge, and hydrophilic nature, lead to the widespread use of cationic polymers and lipid-based nanocarriers, which might induce potential cytotoxicity, thus limiting their clinical application. Here, we report a facile strategy for changing the inherent features of siRNA molecules by achieving hydrophobization. We found that the simple mixing of siRNA and doxorubicin hydrochloride (DOX·HCl) could form a hydrophobic complex, which was readily encapsulated into noncationic PEG- b-PLA micelles for systemic delivery. In addition to delivering DOX·HCl, this strategy could be extended to deliver other hydrochloride forms of anticancer drugs with large hydrophobic domains. This facile strategy efficiently avoids the use of cationic nanocarriers, providing a new avenue for siRNA delivery.


Subject(s)
Drug Carriers/chemistry , Drug Delivery Systems , Neoplasms/drug therapy , RNA, Small Interfering/pharmacology , Antineoplastic Agents/chemistry , Cations/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Carriers/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Lactates/chemistry , Micelles , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , RNA, Small Interfering/chemistry
15.
ACS Biomater Sci Eng ; 5(2): 1034-1044, 2019 Feb 11.
Article in English | MEDLINE | ID: mdl-33405794

ABSTRACT

The use of antimicrobial peptides (AMPs)-functionalized titanium implants is an efficient method for preventing bacterial infection. However, the attachment of AMPs to the surface of titanium implants remains a challenge. In this study, a "clickable" titanium surface was developed by using a silane coupling agent with an alkynyl group. The antimicrobial titanium implant was then constructed through the reaction between the "clickable" surface and azido-AMPs (PEG-HHC36:N3-PEG12-KRWWKWWRR) via click chemistry of Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC). Such an antimicrobial titanium implant, with an AMP density of 897.4 ± 67.3 ng/cm2 (2.5 ± 0.2 molecules per nm2) on the surface, exhibited good and stable antimicrobial activity, inhibited 90.2% of Staphylococcus aureus and 88.1% of Escherichia coli after 2.5 h of incubation, and even inhibited 69.5% of Staphylococcus aureus after 4 days of degradation. The CCK-8 assay indicated that the antimicrobial titanium implant exhibited negligible cytotoxicity to mouse bone mesenchymal stem cells. In vivo assay illustrated that this implant could kill 78.8% of Staphylococcus aureus after 7 days. This method has great potential for the preparation of antimicrobial titanium implants and the prevention of infections in the clinic.

16.
Proc Natl Acad Sci U S A ; 114(48): 12675-12680, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133389

ABSTRACT

Current clinical treatment of Helicobacter pylori infection, the main etiological factor in the development of gastritis, gastric ulcers, and gastric carcinoma, requires a combination of at least two antibiotics and one proton pump inhibitor. However, such triple therapy suffers from progressively decreased therapeutic efficacy due to the drug resistance and undesired killing of the commensal bacteria due to poor selectivity. Here, we report the development of antimicrobial polypeptide-based monotherapy, which can specifically kill H. pylori under acidic pH in the stomach while inducing minimal toxicity to commensal bacteria under physiological pH. Specifically, we designed a class of pH-sensitive, helix-coil conformation transitionable antimicrobial polypeptides (HCT-AMPs) (PGA)m-r-(PHLG-MHH)n, bearing randomly distributed negatively charged glutamic acid and positively charged poly(γ-6-N-(methyldihexylammonium)hexyl-l-glutamate) (PHLG-MHH) residues. The HCT-AMPs showed unappreciable toxicity at physiological pH when they adopted random coiled conformation. Under acidic condition in the stomach, they transformed to the helical structure and exhibited potent antibacterial activity against H. pylori, including clinically isolated drug-resistant strains. After oral gavage, the HCT-AMPs afforded comparable H. pylori killing efficacy to the triple-therapy approach while inducing minimal toxicity against normal tissues and commensal bacteria, in comparison with the remarkable killing of commensal bacteria by 65% and 86% in the ileal contents and feces, respectively, following triple therapy. This strategy renders an effective approach to specifically target and kill H. pylori in the stomach while not harming the commensal bacteria/normal tissues.


Subject(s)
Amines/chemistry , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Glutamic Acid/pharmacology , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Animals , Anti-Bacterial Agents/chemical synthesis , Antimicrobial Cationic Peptides/chemical synthesis , Disease Models, Animal , Female , Glutamic Acid/analogs & derivatives , Glutamic Acid/chemical synthesis , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Helicobacter pylori/physiology , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Organ Specificity , Protein Conformation, alpha-Helical , Static Electricity , Stomach/drug effects , Stomach/microbiology , Stomach/pathology
17.
J Microencapsul ; 34(6): 513-521, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28705043

ABSTRACT

Mannose receptor (MR) is a highly effective endocytic receptor. It is closely related to tumour immune escape and metastasis. We found that MR was highly expressed in some colon cancer cell lines such as CT26 and HCT116 cells. Therefore, MR might be a potential target in colon cancer therapy. In this study, we aimed to develop mannosylated liposomes containing anticancer drug paclitaxel and investigate the potential effects on targeted therapy for colon cancer. Mannosylated liposomes were prepared by film dispersion method. Characterisation, drug release behaviour, cytotoxicity, cellular uptake, anti-tumour efficacy and safety profiles of liposomes were investigated. The results showed that mannosylated liposomes had a higher CT26 cells uptake efficiency and tumour inhibition rate, which might be due to the target effect to MR. And no notable toxicity was observed. Taken together, these data demonstrated that mannosylated liposomes could target colon cancer and improve the efficacy of chemotherapy.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Colonic Neoplasms/drug therapy , Lectins, C-Type/metabolism , Liposomes/chemistry , Mannose-Binding Lectins/metabolism , Mannose/metabolism , Paclitaxel/administration & dosage , Receptors, Cell Surface/metabolism , Animals , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Mannose Receptor , Mice , Mice, Inbred BALB C
18.
Angew Chem Int Ed Engl ; 56(36): 10826-10829, 2017 08 28.
Article in English | MEDLINE | ID: mdl-28660713

ABSTRACT

The application of antimicrobial peptides (AMPs) is largely hindered by their non-specific toxicity against mammalian cells, which is usually associated with helical structure, hydrophobicity, and charge density. A random coil-to-helix transition mechanism has now been introduced into the design of AMPs, minimizing the toxicity against mammalian cells while maintaining high antimicrobial activity. By incorporating anionic phosphorylated tyrosine into the cationic polypeptide, the helical structure of AMPs was distorted owing to the side-chain charge interaction. Together with the decreased charge density, the AMPs exhibited inhibited toxicity against mammalian cells. At the infectious site, the AMPs can be activated by bacterial phosphatase to restore the helical structure, thus contributing to strong membrane disruptive capability and potent antimicrobial activity. This bacteria-activated system is an effective strategy to enhance the therapeutic selectivity of AMPs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Bacillus cereus/drug effects , Staphylococcus aureus/drug effects , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Bacillus cereus/metabolism , Cell Line , HEK293 Cells , Humans , Mice , Microbial Sensitivity Tests , Molecular Conformation , Particle Size , RAW 264.7 Cells , Staphylococcus aureus/metabolism
20.
Sci Rep ; 6: 20864, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26868841

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is a candidate oncogenic driver due to its prevalent overexpression and aberrant repression of tumor suppressor genes in diverse cancers. Therefore, blocking EZH2 enzyme activity may present a valid therapeutic strategy for the treatment of cancers with EZH2 overexpression including breast cancers. Here, we described ZLD1039 a potent, highly selective, and orally bioavailable small molecule inhibitor of EZH2, which inhibited breast tumor growth and metastasis. ZLD1039 considerably inhibited EZH2 methyltransferase activity with nanomolar potency, decreased global histone-3 lysine-27 (H3K27) methylation, and reactivated silenced tumor suppressors connected to increased survival of patients with breast cancer. Comparable to conditional silencing of EZH2, its inhibition by ZLD1039 decreased cell proliferation, cell cycle arrest, and induced apoptosis. Comparably, treatment of xenograft-bearing mice with ZLD1039 led to tumor growth regression and metastasis inhibition. These data confirmed the dependency of breast cancer progression on EZH2 activity and the usefulness of ZLD1039 as a promising treatment for breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Breast Neoplasms/metabolism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histones/metabolism , Pyridones/pharmacology , Quinolones/pharmacology , Animals , Apoptosis/drug effects , Benzamides/chemistry , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Humans , Intracellular Space/metabolism , Methylation/drug effects , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Polycomb Repressive Complex 2/metabolism , Pyridones/chemistry , Quinolones/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL