Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Anal Chim Acta ; 1316: 342860, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38969429

ABSTRACT

BACKGROUND: Glutathione (GSH), a highly abundant thiol compound within cells, plays a critical role in physiological processes and exhibits close correlation with cancer. Among molecular imaging technologies, most probes have relatively short emission wavelengths and lack photoacoustic imaging (PA) capability, resulting in the inability to obtain tissue images with high penetration depth. The presence of GSH in the tumor microenvironment neutralizes ROS, diminishing the therapeutic effect of PDT, thus resulting in often unsatisfactory therapeutic efficacy. Therefore, it is imperative to develop a dual-modal probe for the detection of GSH and the diagnosis and treatment of cancer. RESULTS: In this study, we synthesized a novel dual-modal probe, Cy-Bio-GSH, utilizing near-infrared fluorescence (NIRF) and photoacoustic (PA) imaging techniques for GSH detection. The probe integrates cyanine dye as the fluorophore, nitroazobenzene as the recognition moiety, and biotin as the tumor-targeting moiety. Upon reacting with GSH, the probe emits NIR fluorescence at 820 nm and generates a PA signal. Significantly, this reaction activates the photodynamic and photothermal properties of the probe. By depleting GSH and employing a synergistic photothermal therapy (PTT) treatment, the therapeutic efficacy of photodynamic therapy (PDT) is remarkably enhanced. In-vivo experiments confirm the capability of the probe to detect GSH via NIRF and PA imaging. Notably, the combined tumor-targeting ability and PDT/PTT synergistic therapy enhance therapeutic outcomes for tumors and facilitate their ablation. SIGNIFICANCE: A novel tumor-targeting and dual-modal imaging probe (Cy-Bio-GSH) is synthesized, exhibiting remarkable sensitivity and selectivity to GSH, enabling the visualization of GSH in cells and the differentiation between normal and cancer cells. Cy-Bio-GSH enhances PDT/PTT with effective killing of cancer cells and makes the ablation of tumors in mice. This work represents the first tumor-targeting probe for GSH detection, and provides crucial tool for cancer diagnosis and treatment by dual-modal imaging with improved PDT/PTT synergistic therapy.


Subject(s)
Biotin , Glutathione , Photoacoustic Techniques , Photochemotherapy , Glutathione/chemistry , Glutathione/metabolism , Animals , Humans , Mice , Biotin/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Optical Imaging , Female , Photothermal Therapy , Mice, Nude , Mice, Inbred BALB C , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/therapeutic use
2.
Bioorg Chem ; 149: 107531, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38850779

ABSTRACT

Nitroreductase (NTR) overexpression often occurs in tumors, highlighting the significance of effective NTR detection. Despite the utilization of various optical methods for this purpose, the absence of an efficient tumor-targeting optical probe for NTR detection remains a challenge. In this research, a novel tumor-targeting probe (Cy-Bio-NO2) is developed to perform dual-modal NTR detection using near-infrared fluorescence and photoacoustic techniques. This probe exhibits exceptional sensitivity and selectivity to NTR. Upon the reaction with NTR, Cy-Bio-NO2 demonstrates a distinct fluorescence "off-on" response at 800 nm, with an impressive detection limit of 12 ng/mL. Furthermore, the probe shows on-off photoacoustic signal with NTR. Cy-Bio-NO2 has been successfully employed for dual-modal NTR detection in living cells, specifically targeting biotin receptor-positive cancer cells for imaging purposes. Notably, this probe effectively detects tumor hypoxia through dual-modal imaging in tumor-bearing mice. The strategy of biotin incorporation markedly enhances the probe's tumor-targeting capability, facilitating its engagement in dual-modal imaging at tumor sites. This imaging capacity holds substantial promise as an accurate tool for cancer diagnosis.


Subject(s)
Fluorescent Dyes , Nitroreductases , Optical Imaging , Animals , Humans , Mice , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms/diagnostic imaging , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Nitroreductases/metabolism , Nitroreductases/analysis , Photoacoustic Techniques , Nitrogen Dioxide/chemical synthesis , Nitrogen Dioxide/chemistry
3.
Food Chem ; 450: 139296, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38636381

ABSTRACT

Advanced targeted nanoparticles (NPs) were designed to enhance the targeted delivery of resveratrol (RES) and quercetin (QUE) by utilizing carboxymethyl chitosan (CTS) and Jiuzao glutelin isolate (JGI) conjugates. Briefly, RES and QUE were encapsuled within CTS-JGI-2 (CTS/JGI, m/m, 2:1). The carrier's targeting properties were further improved through the incorporation of folic acid (FA) and polyethylenimine (PEI). Moreover, the stability against digestion was enhanced by incorporating baker yeast cell walls (BYCWs) to construct RES-QUE/FA-PEI/CTS-JGI-2/MAT/BYCW NPs. The results demonstrated that FA-PEI/CTS-JGI-2/MAT/BYCW NPs could improve cellular uptake and targeting property of RES and QUE through endocytosis of folic acid receptors (FOLRs). Additionally, RES-QUE successfully alleviated LPS- and DSS-induced inflammation by regulating NF-κB/IkBa/AP-1 and AMPK/SIRT1signaling pathways and reducing the secretion of inflammatory mediators and factors. These findings indicate FA-PEI/CTS-JGI-2/MAT/BYCW NPs hold promise as an oral drug delivery system with targeted delivery capacities for functional substances prone to instability in dietary supplements.


Subject(s)
Chitosan , Folic Acid , Nanoparticles , Quercetin , Resveratrol , Chitosan/chemistry , Chitosan/pharmacology , Chitosan/analogs & derivatives , Folic Acid/chemistry , Folic Acid/pharmacology , Quercetin/chemistry , Quercetin/analogs & derivatives , Quercetin/pharmacology , Quercetin/administration & dosage , Nanoparticles/chemistry , Resveratrol/chemistry , Resveratrol/pharmacology , Resveratrol/administration & dosage , Animals , Mice , Humans , Drug Carriers/chemistry , Drug Delivery Systems , RAW 264.7 Cells
4.
ACS Nano ; 17(15): 14555-14571, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37350440

ABSTRACT

Persistent inflammation within atherosclerotic plaques is a crucial factor contributing to plaque vulnerability and rupture. It has become increasingly evident that the proinflammatory microenvironment of the plaque, characterized by heightened monocyte recruitment, oxidative stress, and impaired clearance of apoptotic cells, plays a significant role in perpetuating inflammation and impeding its resolution. Consequently, targeting and eliminating these proinflammatory features within the plaque microenvironment have emerged as a promising therapeutic approach to restore inflammation resolution and mitigate the progression of atherosclerosis. While recent advancements in nanotherapeutics have demonstrated promising results in targeting individual proinflammatory characteristics, the development of an effective therapeutic strategy capable of simultaneously addressing multiple proinflammatory features remains a challenge. In this study, we developed a multifunctional nanozyme based on Prussian blue, termed PBNZ@PP-Man, to simultaneously target and eliminate various proinflammatory factors within the plaque microenvironment. Through systematic investigations, we have elucidated the antiatherosclerotic mechanisms of PBNZ@PP-Man. Our results demonstrate that PBNZ@PP-Man possesses the ability to accumulate within atherosclerotic plaques and effectively eliminate multiple proinflammatory factors, leading to inflammation resolution. Specifically, PBNZ@PP-Man suppresses monocyte recruitment, scavenges reactive oxygen species, and enhances efferocytosis. Notably, PBNZ@PP-Man exhibits a much stronger efficacy to resolve the proinflammatory plaque microenvironment and attenuate atherosclerosis in comparison to the approach that merely eliminates one single risky factor in the plaque. It significantly enhances the inflammation resolution capabilities of macrophages and attenuates atherosclerosis. These results collectively underscore the importance of modulating the proinflammatory plaque microenvironment as a complementary strategy for resolving inflammation in atherosclerosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Humans , Plaque, Atherosclerotic/drug therapy , Atherosclerosis/drug therapy , Macrophages , Phagocytosis , Inflammation/drug therapy
5.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-932410

ABSTRACT

Objective:To prepare a phase-change lipid nanoparticle modified by tumor homing membrane-penetrating peptide (tLyP-1) and carrying paclitaxel (PTX) engineered by metal polyphenol network (TA-Fe 3+ ), and evaluate the therapeutic effects of tumor targeting, ultrasound/photoacoustic imaging and photothermal combined chemotherapy in vitro. Methods:Phase-change lipid nanoparticles (t-P@TFP) with TA-Fe 3+ engineered PTX mediated by tLyP-1 were prepared by solvent replacement method, thin film hydration method and double emulsification method. Its detection and characterization, in vitro targeting ability, photothermal conversion ability, in vitro photoacoustic and ultrasonic imaging ability, CCK-8 method, cell live and death double staining method and flow cytometry method were used to detect the safety of nanoparticles and the killing effects of different nanoparticles on 4T1 cells. Results:t-P@TFP nanoparticles were successfully prepared. Transmission electron microscopy showed that the nanoparticles were spherical with uniform shape and size, with a particle size of (209.8±1.56)nm and a potential of (-25.9±1.36)mV. Laser confocal scanning microscopy showed that t-P@TFP nanoparticles could gather around 4T1 cells in a targeted manner. It had an efficient photothermal conversion effect, and nanoparticles could quickly become microbubbles after being irradiated by near-infrared laser, which enhanced the in vitro ultrasonic imaging effect; The photoacoustic signal of nanoparticles increased with the increase of concentration. CCK-8 method, double staining of living and dead cells and flow cytometry showed that t-P@TFP combined photothermal chemotherapy had the best anti-tumor effect. Conclusions:t-P@TFP nanoparticles are successfully prepared. The nanoparticles have good targeting ability for photoacoustic and ultrasonic imaging and have good photothermal effect, killing breast cancer cells, which is expected to realize the integration of diagnosis and treatment.

6.
Int J Nanomedicine ; 16: 8037-8048, 2021.
Article in English | MEDLINE | ID: mdl-34934312

ABSTRACT

BACKGROUND: The construction of tumor-targeting carriers with favorable transfection efficiency was of great significance to achieve the tumor gene therapy. The phenylboronic acid-modified polyamidoamine (namely PP) was employed as a carrier for the delivery of Polo-like kinase-1 siRNA (siPlk-1), inducing an obvious anti-tumor response. MATERIALS AND METHODS: The interaction between PP and siPlk-1 was evaluated by gel retardation assay. The transfection efficiency and tumor-targeting ability were analyzed by flow cytometry and confocal laser scanning microscopy, using hepatocarcinoma cell line HepG2 as a model. The anti-proliferation effect of PP/siPlk-1 and related mechanism were studied using the strategies of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, cell apoptosis and cell cycle arrest. The anti-migration effect induced by PP/siPlk-1 delivery was assayed by wound healing and Transwell migration techniques. Finally, quantitative real-time PCR and Western blotting were performed to measure the expression level of Plk-1 and other key targets. RESULTS: The derivative PP could achieve the condensation of siPlk-1 into stable nanoparticles at nitrogen/phosphate groups ratio (N/P ratio) of >3.0, and it could facilitate the transfection of siPk-1 in a phenylboronic acid-dependent manner. The PP/siPlk-1 nanoparticles exhibited obvious anti-proliferation effect owing to the gene silence of Plk-1, which was identified to be associated with the cell apoptosis and cell cycle arrest at G2 phase. Meanwhile, PP/siPlk-1 transfection could efficiently suppress the migration and invasion of tumor cells. CONCLUSION: The derivative PP has been demonstrated to be an ideal tumor-targeting carrier for the delivery of Plk-1 siRNA, exhibiting great potential in the gene therapy of malignant tumors.


Subject(s)
Apoptosis , Neoplasms , Boronic Acids , Cell Line, Tumor , Cell Proliferation , Humans , Polyamines , RNA, Small Interfering/genetics , Transfection
7.
Nanomedicine (Lond) ; 16(24): 2207-2242, 2021 10.
Article in English | MEDLINE | ID: mdl-34533048

ABSTRACT

With the excellent ability to transform near-infrared light to localized visible or UV light, thereby achieving deep tissue penetration, lanthanide ion-doped upconversion nanoparticles (UCNP) have emerged as one of the most striking nanoscale materials for more effective and safer cancer treatment. Up to now, UCNPs combined with photosensitive components have been widely used in the delivery of chemotherapy drugs, photodynamic therapy and photothermal therapy. Applications in these directions are reviewed in this article. We also highlight microenvironmental tumor monitoring and precise targeted therapies. Then we briefly summarize some new trends and the existing challenges for UCNPs. We hope this review can provide new ideas for future cancer treatment based on UCNPs.


Subject(s)
Lanthanoid Series Elements , Nanoparticles , Neoplasms , Photochemotherapy , Humans , Infrared Rays , Lanthanoid Series Elements/therapeutic use , Neoplasms/drug therapy
8.
Cancer Med ; 10(14): 4677-4696, 2021 07.
Article in English | MEDLINE | ID: mdl-34165267

ABSTRACT

INTRODUCTION: Traditional cancer therapy has many disadvantages such as low selectivity and high toxicity of chemotherapy, as well as insufficient efficacy of targeted therapy. To enhance the cytotoxic effect and targeting ability, while reducing the toxicity of antitumor drugs, an antibody drug conjugate (ADC) was developed to deliver small molecular cytotoxic payloads directly to tumor cells by binding to specific antibodies via linkers. METHOD: By reviewing published literature and the current progress of ADCs, we aimed to summarize the basic characteristics, clinical progress, and challenges of ADCs to provide a reference for clinical practice and further research. RESULTS: ADC is a conjugate composed of three fundamental components, including monoclonal antibodies, cytotoxic payloads, and stable linkers. The mechanisms of ADC including the classical internalization pathway, antitumor activity of antibodies, bystander effect, and non-internalizing mechanism. With the development of new drugs and advances in technology, various ADCs have achieved clinical efficacy. To date, nine ADCs have received US Food and Drug Administration (FDA) approval in the field of hematologic tumors and solid tumors, which have become routine clinical treatments. CONCLUSION: ADC has changed traditional treatment patterns for cancer patients, which enable the same treatment for pancreatic cancer patients and promote individualized precision treatment. Further exploration of indications could focus on early-stage cancer patients and combined therapy settings. Besides, the mechanisms of drug resistance, manufacturing techniques, optimized treatment regimens, and appropriate patient selection remain the major topics.


Subject(s)
Immunoconjugates/therapeutic use , Neoplasms/therapy , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Bystander Effect , Clinical Trials as Topic , Drug Approval , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Humans , Immunoconjugates/immunology , Molecular Targeted Therapy/methods , Neoplasms/immunology , Pancreatic Neoplasms/therapy
9.
Biomaterials ; 274: 120888, 2021 07.
Article in English | MEDLINE | ID: mdl-34029915

ABSTRACT

The modification of targeting ligands on nanoparticles (NPs) is anticipated to enhance the delivery of therapeutics to diseased tissues. However, once exposed to the blood stream, NPs can immediately adsorb proteins to form the "protein corona," which may greatly hinder the targeting ligand from binding to its receptor. For brain-targeting delivery, nanotherapeutics must traverse the blood-brain barrier (BBB) to enter the brain parenchyma and then target the diseased cells. However, it remains elusive whether, apart from receptor recognition, the protein corona can affect other processes involved in BBB transcytosis, such as endocytosis, intracellular trafficking, and exocytosis. Furthermore, the targeting ability of NPs toward diseased cells after transcytosis remains unclear. Herein, transferrin (Tf), a brain-targeting ligand, was coupled to NPs to evaluate BBB transcytosis and brain tumor targeting ability. Different impacts of the in vitro and in vivo protein corona on receptor targeting, lysosomal escape, and BBB transcytosis were found. The in vitro protein corona abolished the Tf-mediated effects of the abovementioned processes, whereas the in vivo protein corona attenuated these effects. After crossing the BBB, Tf retained its targeting specificity towards brain tumor cells. Together, these results revealed that several bound apolipoproteins, especially apolipoprotein A-I, may help NPs traverse the BBB, thereby providing novel insights into the development of brain-targeted delivery.


Subject(s)
Brain Neoplasms , Nanoparticles , Protein Corona , Blood-Brain Barrier/metabolism , Brain/metabolism , Humans , Transcytosis , Transferrin
10.
Angew Chem Int Ed Engl ; 60(28): 15459-15465, 2021 07 05.
Article in English | MEDLINE | ID: mdl-33904236

ABSTRACT

A tumor-targeting enhanced chemotherapy, enabled by aptamer-drug conjugate nanomicelles, is reported that boosts antitumor immune responses. Multivalent aptamer drug conjugate (ApMDC), an amphiphilic telodendrimer consisting of a hydrophilic aptamer and a hydrophobic monodendron anchored with four anticancer drugs by acid-labile linkers, was designed and synthesized. By co-self-assembly with an ApMDC analogue, in which aptamer is replaced with polyethylene glycol, the surface aptamer density of these nanomicelles can be screened to reach an optimal complementation between blood circulation and tumor-targeting ability. Optimized nanomicelles can enhance immunogenic cell death of tumor cells, which strikingly augments the tumor-specific immune responses of the checkpoint blockade in immunocompetent tumor-bearing mice. ApMDC nanomicelles represent a robust platform for structure-function optimization of drug conjugates and nanomedicines.


Subject(s)
Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/therapeutic use , Immunotherapy , Nanoparticles/chemistry , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/chemistry , Micelles , Nanomedicine , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology
11.
AAPS PharmSciTech ; 22(3): 96, 2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33694067

ABSTRACT

Increasing the drug tumor-specific accumulation and controlling their release is considered one of the most effective ways to increase the efficacy of drugs. Here, we developed a vesicle system that can target hepatoma and release drugs rapidly within tumor cells. This non-ionic surfactant vesicle is biodegradable. Galactosylated stearate has been used to glycosylate the vesicles to achieve liver targeting; replacement of a portion (Chol:CHEMS = 1:1) of cholesterol by cholesteryl hemisuccinate (CHEMS) allows for a rapid release of drugs in an acidic environment. In vitro release experiments confirmed that galactose-modified pH-sensitive niosomes loaded with tanshinone IIA had excellent drug release performance in acid medium. In vitro experiments using ovarian cancer cells (A2780), colon cancer cells (HCT8), and hepatoma cell (Huh7, HepG2) confirmed that the preparation had specific targeting ability to hepatoma cells compared with free drugs, and this ability was dependent on the galactose content. Furthermore, the preparation also had a more substantial inhibitory effect on tumor cells, and subsequent apoptosis assays and cell cycle analyses further confirmed its enhanced anti-tumor effect. Results of pharmacokinetic experiments confirmed that the vesicle system could significantly extend the blood circulation time of tanshinone IIA, and the larger area under the curve indicated that the preparation had a better drug effect. Thus, the results of biodistribution experiments confirmed the in vivo liver targeting ability of this preparation. Niosomes designed in this manner are expected to be a safe and effective drug delivery system for liver cancer therapy.


Subject(s)
Abietanes/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Carcinoma, Hepatocellular/metabolism , Drug Delivery Systems/methods , Galactose/administration & dosage , Liver Neoplasms/metabolism , Abietanes/pharmacokinetics , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Liberation/physiology , Galactose/pharmacokinetics , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Liposomes , Liver Neoplasms/drug therapy , Male , Mice , Random Allocation , Rats , Rats, Sprague-Dawley
12.
Nanomedicine ; 32: 102342, 2021 02.
Article in English | MEDLINE | ID: mdl-33253922

ABSTRACT

Acute kidney injury (AKI) is a life-threatening disease without effective treatment. The utilization of curcumin (Cur) for the treatment of AKI is still facing challenges due to its poor water-solubility and low bioavailability. Herein, kidney-targeted octenyl succinic anhydride-grafted fucoidan loaded with Cur (OSA-Fucoidan/Cur) was fabricated for synergistic treatment of AKI. It was found that OSA-Fucoidan/Cur micelles had a sustained drug release behavior and excellent physicochemical stability. Cellular uptake studies demonstrated that the specific binding between fucoidan and P-selectin overexpressed on H2O2-stimulated HUVECs contributed to the higher internalization of OSA-Fucoidan/Cur micelles by the cells. In addition, OSA-Fucoidan micelles exhibited an ideal kidney-targeted characteristic in lipopolysaccharide (LPS)-induced AKI mice. In vivo studies showed that the combination of Cur and OSA-Fucoidan endowed the OSA-Fucoidan/Cur micelles with synergistically anti-inflammatory and antioxidant abilities, thereby largely enhancing the therapeutic efficacy of AKI. Therefore, OSA-Fucoidan/Cur micelles may represent a potential kidney-targeted nanomedicine for effective treatment of AKI.


Subject(s)
Acute Kidney Injury/drug therapy , Drug Carriers/chemistry , Micelles , P-Selectin/antagonists & inhibitors , Polysaccharides/chemistry , Acute Kidney Injury/pathology , Animals , Antioxidants/pharmacology , Curcumin/pharmacology , Curcumin/therapeutic use , Drug Liberation , Endocytosis/drug effects , Half-Life , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred ICR , Succinic Anhydrides/chemistry , Tissue Distribution/drug effects
13.
Anticancer Agents Med Chem ; 21(7): 910-918, 2021.
Article in English | MEDLINE | ID: mdl-32698755

ABSTRACT

OBJECTIVE: To explore the targeting effect of PLGA-NP and iRGD co-administration with PTXPLGA NP (PTX-PLGA + iRGD) on colorectal cancer. METHODS: Whether PLGA-NP co-administration with iRGD peptide could show effective tumor-targeting ability in contrast to with PLGA-NP in colorectal cancer mice models was evaluated. Moreover, the chemotherapeutics Paclitaxel (PTX) was loaded into the PLGA-NP to impart anti-tumor efficiency to the PTX-PLGA. Whether iRGD co-administration with PTX-PLGA NP (PTX-PLGA + iRGD) in colorectal cancer models enabled PTX to achieve better anti-tumor efficiency and biocompatibility was further assessed. RESULTS: The targeting ability of PLGA-NP was enhanced in cell experiment and colorectal cancer mice models by co-administration of iRGD. As a result, PTX-PLGA + iRGD achieved better anti-tumor efficacy than PTX and PTX-PLGA. Conlusion: The nanocarrier based on PLGA with specific targeting ability could promote the clinical application of various chemotherapeutics similar to PTX. The combination of drug-loaded nanoparticles and iRGD could develop into a promising drug delivery system.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/drug therapy , Nanoparticles/chemistry , Oligopeptides/chemistry , Paclitaxel/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/pathology , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Nude , Nanoparticles/administration & dosage , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Oligopeptides/administration & dosage , Paclitaxel/administration & dosage , Paclitaxel/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Tumor Cells, Cultured
14.
Colloids Surf B Biointerfaces ; 189: 110829, 2020 May.
Article in English | MEDLINE | ID: mdl-32036332

ABSTRACT

Chlorin e6 (Ce6), with its high phototoxic potential, has wide applications in photodynamic therapy (PDT) for many human diseases. However, poor cancer cell localization of Ce6 has limited its direct application for PDT. Here, we developed cancer-targeting peptide p 18-4/chlorin e6 (Ce6)-conjugated polyhedral oligomeric silsesquioxane (PPC) nanoparticles for improving the targeting ability of Ce6 to breast cancer cells, thereby enhancing PDT efficacy. The synthesized PPC nanoparticles exhibited a spherical shape with an average diameter of 127.2 ± 11.3 nm in aqueous solution. Compared with free Ce6, the immobilization of p 18-4 enhanced the in vitro cellular uptake and targeting ability of PPC nanoparticles in breast cancer cell line MDA-MB-231. In addition, the intracellular uptake of PPC nanoparticles in MDA-MB-231 cells was dramatically increased compared with other cancer cells, indicating an obvious targeting ability of PPC nanoparticles on breast cancer cells. Upon light irradiation, PPC nanoparticles revealed significantly improved phototoxicity to MDA-MB-231 cells, mainly due to apoptotic cell death. In vivo PDT study suggested that PPC nanoparticles exhibited increased retention in tumor tissues and effectively inhibited the growth of MDA-MB-231 tumors in a target-specific manner. Overall, these results indicate that PPC nanoparticles are highly effective PDT agents for breast cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Nanoparticles/chemistry , Organosilicon Compounds/pharmacology , Peptides/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Chlorophyllides , Drug Screening Assays, Antitumor , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Organosilicon Compounds/chemistry , Particle Size , Peptides/chemistry , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Surface Properties , Tumor Cells, Cultured
15.
AAPS PharmSciTech ; 20(7): 270, 2019 Jul 30.
Article in English | MEDLINE | ID: mdl-31363872

ABSTRACT

Currently, there is no specific treatment for acute lung injury (ALI). E-selectin-binding peptide (Esbp), a high-affinity peptide that delivers drugs targeting inflammatory vascular endothelial cells, can bind to E-selectin and act as a targeting ligand for selective drug delivery. In this study, we coupled the thiol groups of Esbp to the amino groups on the surface of bovine serum albumin (BSA) using succinimidyl iodoacetic acid to make Esbp-modified BSA nanoparticles (BSANPs) at the average ratio of 19.3 µg Esbp to 1 mg BSA. The Esbp-modified BSANPs were spherical in shape and had a particle size of 266.7 ± 2.7 nm, polydispersity index of 0.165 ± 0.02, zeta potential of - 33.64 ± 1.23 mV, encapsulation efficiency of 84.3 ± 2.3%, and drug loading of 6.7 ± 0.32%. The cumulative release rate of dexamethasone-loaded Esbp-modified BSANPs was 51.2% within 12 h, significantly lower than that of 88.2% of free drugs. Moreover, Esbp-modified BSANPs could be uptaken in vitro by activated human umbilical vein endothelial cells and in vivo by the lungs of the established ALI mouse model. These results indicated that our Esbp-modified BSANPs delivery system has characteristics of good targeting ability and biocompatibility and is able to inhibit inflammation. Overall, our Esbp-modified BSANPs delivery system has therapeutic potentials as a new targeting drug system for the treatment of ALI in the future.


Subject(s)
Acute Lung Injury/drug therapy , Dexamethasone/administration & dosage , E-Selectin/administration & dosage , HLA-D Antigens/administration & dosage , Nanoparticles/administration & dosage , Serum Albumin, Bovine/administration & dosage , Acute Lung Injury/metabolism , Animals , Cattle , Cell Line, Tumor , Dexamethasone/metabolism , Dose-Response Relationship, Drug , Drug Carriers/administration & dosage , Drug Carriers/metabolism , Drug Delivery Systems/methods , E-Selectin/metabolism , HLA-D Antigens/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Nanoparticles/metabolism , Particle Size , Serum Albumin, Bovine/metabolism , Treatment Outcome
16.
AAPS PharmSciTech ; 20(7): 292, 2019 Aug 19.
Article in English | MEDLINE | ID: mdl-31428888

ABSTRACT

Mitoxantrone (MTO) is used to treat certain types of cancer, mostly metastatic cancer. While the drug has poor aqueous solubility and high side effects. Self-assembly nanocrystal is a novel lymphatic targeting delivery system. In our study, MTO self-assembly nanocrystal (MTO NC) was successfully prepared to improve lymphatic targeting ability and reduce its toxicity. MTO NCs had small size, stable potential, and uniform distribution. The average particle size of MTO NCs was less than 100 nm with the 0.218 PDI and - 6.6 mV the Zeta potential value. TEM images showed that MTO NCs had a sphere-like morphology with smooth surface and uniform distribution; Atomic force microscopy (AFM) images gave a 3D surface of MTO NCs. Polarizing microscope micrograph (PLM) of MTO NCs in lymph nodes demonstrated the crystal structure of MTO NCs when it was exposed to physiological condition. Transmission electron microscopy showed the presence of MTO NCs in mice lymph nodes. Pharmacokinetic parameters of MTO strongly demonstrated that MTO NCs could target the lymph nodes after subcutaneous injection. Moreover, tissue distribution results indicated that MTO NCs were mainly absorbed by the lymphatics and reduced system toxicity. Finally, a lymphatic metastasis mice model was established to precede the pharmacodynamics of MTO NCs, and using MTO liposomes as a reference preparation, the inhibitory effect of MTO NCs on lymphatic metastasis was markedly higher. Briefly, MTO NCs, as a novel self-assembled lymphatic targeting system, can accumulate in the metastatic lymph nodes and lead anticancer drug to kill cancer cells and control lymphatic metastasis with extremely low systemic toxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Lymph Nodes/drug effects , Lymphatic Metastasis , Mitoxantrone/pharmacology , Nanoparticles , Animals , Antineoplastic Agents/chemistry , Liposomes/metabolism , Mice , Mitoxantrone/chemistry , Solubility , Tissue Distribution
17.
Mol Pharm ; 16(5): 1982-1998, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30892898

ABSTRACT

Locating nanomedicines at the active sites plays a pivotal role in the nanoparticle-based cancer therapy field. Herein, a multifunctional nanotherapeutic is designed by using graphene oxide (GO) nanosheets with rich carboxyl groups as the supporter for hyaluronic acid (HA)-methotrexate (MTX) prodrug modification via an adipicdihydrazide cross-linker, achieving synergistic multistage tumor-targeting and combined chemo-photothermal therapy. As a tumor-targeting biomaterial, HA can increase affinity of the nanocarrier toward CD44 receptor for enhanced cellular uptake. MTX, a chemotherapeutic agent, can also serve as a tumor-targeting enhancer toward folate receptor based on its similar structure with folic acid. The prepared nanosystems possess a sheet shape with a dynamic size of approximately 200 nm and pH-responsive drug release. Unexpectedly, the physiological stability of HA-MTX prodrug-decorated GO nanosystems in PBS, serum, and even plasma is more excellent than that of HA-decorated GO nanosystems, while both of them exhibit an enhanced photothermal effect than GO nanosheets. More importantly, because of good blood compatibility as well as reduced undesired interactions with blood components, HA-MTX prodrug-decorated GO nanosystems exhibited remarkably superior accumulation at the tumor sites by passive and active targeting mechanisms, achieving highly effective synergistic chemo-photothermal therapeutic effect upon near-infrared laser irradiation, efficient ablation of tumors, and negligible systemic toxicity. Hence, the HA-MTX prodrug-decorated hybrid nanosystems have a promising potential for synergistic multistage tumor-targeting therapy.


Subject(s)
Drug Delivery Systems/methods , Graphite/chemistry , Nanoconjugates/chemistry , Photochemotherapy/methods , Uterine Cervical Neoplasms/therapy , Adipates/chemistry , Adipates/metabolism , Animals , Cell Survival/drug effects , Drug Liberation , Drug Stability , Drug Synergism , Female , Graphite/metabolism , HeLa Cells , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , MCF-7 Cells , Methotrexate/chemistry , Methotrexate/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NIH 3T3 Cells , Prodrugs/chemistry , Prodrugs/metabolism , Tissue Distribution , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
18.
Int J Nanomedicine ; 13: 4961-4975, 2018.
Article in English | MEDLINE | ID: mdl-30214200

ABSTRACT

BACKGROUND: Red blood cell membrane-coated nanoparticle (RBCm-NP) platform, which consist of natural RBCm and synthetic polymeric core, can extend circulation time in vivo with an improved biocompatibility and stability of this biomimetic nanocarrier. To achieve better bioavailability of antitumor drugs that were loaded in RBCm-NPs, the functionalization of coated RBCm with specific targeting ability is essential. Bispecific recombinant protein anti-EGFR-iRGD, containing both tumor penetrating peptide (internalizing RGD peptide) and EGFR single-domain antibody (sdAb), seems to be an optimal targeting ligand for RBCm-NPs in the treatment of multiple tumors, especially colorectal cancer with high EGFR expression. MATERIALS AND METHODS: We modified the anti-EGFR-iRGD recombinant protein on the surface of RBCm-NPs by lipid insertion method to construct iE-RBCm-PLGA NPs and confirmed the presentation of active tumor-targeting ability in colorectal cancer models with high EGFR expression when compared with RBCm-PLGA NPs. In addition, potential anti-tumor drug gambogic acid (GA) was loaded into the NPs to endow the antitumor efficiency of iE-RBCm-GA/PLGA NPs. It was simultaneously evaluated whether GA can reach better biocompatibility benefiting from the improved antitumor efficiency of iE-RBCm-GA/PLGA NPs in colorectal cancer models. RESULTS: We successfully modified anti-EGFR-iRGD proteins on the surface of biomimetic NPs with integrated and stable "shell-core" structure. iE-RBCm-PLGA NPs showed its improved targeting ability in vitro (multicellular spheroids [MCS]) and in vivo (nude mice bearing tumors). Besides, no matter on short-term cell apoptosis at tumor site (terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling [TUNEL]) and long-term tumor inhibition, iE-RBCm-GA/PLGA NPs achieved better antitumor efficacy than free GA in spite of the similar effects of cytotoxicity and apoptosis to GA in vitro. CONCLUSION: We expect that the bispecific biomimetic nanocarrier can extend the clinical application of many other potential antitumor drugs similar to GA and become a novel drug carrier in the colorectal cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomimetic Materials/chemistry , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Nanoparticles/chemistry , Oligopeptides/chemistry , Recombinant Proteins/chemistry , Xanthones/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Drug Carriers/chemistry , ErbB Receptors/metabolism , Humans , Lactic Acid/chemistry , Lipids , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Xanthones/pharmacology
19.
Colloids Surf B Biointerfaces ; 161: 508-518, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29128837

ABSTRACT

It has been shown that multivalent ligands could significantly enhance the binding avidity compared with the monovalent ones; therefore, once incorporated into nanoparticles, they promote superior targeting ability without increasing the ligand density. Although ligand valency and density play a key role on the targeting ability of corresponding nanoparticles, these facotrs remain largely unexplored and detailed studies are lacking. Herein, a series of multivalent ligands with certain valencies (FAn, n indicates the valency of ligand: n=3, 5, 7) has been conveniently synthesized by conjugating different copies of folate ligands with poly(acrylic acid) (PAA). Negatively charged chitosan nanoparticles (CTS-SA NPs) have been utilized as proper multivalent platforms because they can strongly suppress non-specific protein adsorption and cellular uptake without interfering with the targeting ability of multivalent ligands. Subsequently, the structure of CTS-SA NPs has been modified using different amounts of FAn to form multivalent nanoparticles (FAn-CTS-SA NPs) with various valencies and densities. A series of specific investigations of them suggested that the cellular uptake of multivalent nanoparticles has largely varied with the ligand valency variation even at similar ligand densities; and also largely varied with ligand density variation even at the same ligand valencies. The intermediate valency and density values determined in the current study (ie., 5 and 2.4wt%, respectively) have provided the best cellular uptake, facilitating superior targeting ability at relatively low ligand valency and density. Unexpectedly, no conspicuous difference has been observed during endocytotic inhibition assays with single inhibitors, which may be attributed to the synergetic endocytotic mechanism with multiple pathways of multivalent nanoparticles. The optimal multivalent nanoparticles have also exhibited excellent biocompatibility, long-term stability in vitro and enhanced circulation time in vivo, thus demonstrating their potential for targeted drug delivery.


Subject(s)
Chitosan/chemistry , Drug Carriers/chemistry , Drug Delivery Systems/methods , Nanoparticles/chemistry , Animals , Cell Line, Tumor , Cell Survival/drug effects , Endocytosis , Female , Folic Acid/administration & dosage , Folic Acid/chemistry , Folic Acid/pharmacokinetics , Hep G2 Cells , Humans , Ligands , Metabolic Clearance Rate , Nanoparticles/ultrastructure , Rats, Sprague-Dawley , Vitamin B Complex/administration & dosage , Vitamin B Complex/chemistry , Vitamin B Complex/pharmacokinetics
20.
Acta Pharmaceutica Sinica ; (12): 865-877, 2018.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-779946

ABSTRACT

Anti-tumor antibiotics exhibit great application potential in the anti-tumor therapy. Some drugs have become the first-line medication clinically. However, there are always various problems associated with anti-tumor antibiotics, such as poor solubility and instability as well as severe systemic side effects. It is important to choose suitable delivery carriers for a reasonable delivery system for a good targeting ability, enhanced anti-tumor efficacy and reduced adverse effects of the anti-tumor antibiotics, especially in the smart delivery systems. This review summarizes the carriers and the advances in the delivery systems of anti-tumor antibiotics, including anti-tumor antibiotic drugs currently on the market, in the clinical research stage and in the basic research stage.

SELECTION OF CITATIONS
SEARCH DETAIL
...