Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.767
Filter
1.
AAPS PharmSciTech ; 25(7): 216, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289249

ABSTRACT

This study aimed to develop paclitaxel (PTX)-loaded PEGylated (PEG)-pH-sensitive (SpH) liposomes to enhance drug delivery efficiency and cytotoxicity against MCF-7 breast cancer cells. PTX-loaded PEG-SpH liposomes were prepared using the thin film hydration method. ATR-FTIR compatibility studies revealed no significant interactions among liposome formulation components. TEM images confirmed spherical morphology, stability, and an ideal size range (180-200 nm) for improved blood circulation. At pH 5.5, liposomes exhibited increased size and positive zeta potential, indicating pH-sensitive properties due to CHEMS response to the acidic tumor microenvironment. Conversely, at pH 7.4, liposomes showed a slightly larger size (199.25 ± 1.64 nm) and a more negative zeta potential (-36.94 ± 0.32 mV), suggesting successful PEG-SpH surface modification, enhancing stability, and reducing aggregation. PTX-loaded PEG-SpH liposomes demonstrated high encapsulation efficiency (84.57 ± 0.92% w/w) and drug loading capacity (4.12 ± 0.26% w/w). In-vitro drug release studies revealed accelerated first-order PTX release at pH 5.5 and a controlled zero-order release at pH 7.4. Cellular uptake studies on MCF-7 cells demonstrated enhanced PTX uptake, attributed to mPEG-PCL incorporation prolonging circulation time and CHEMS facilitating PTX release in the tumor microenvironment. Furthermore, PTX-loaded PEG-SpH liposomes exhibited significantly improved cytotoxicity with an IC50 value of 1.107 µM after 72-h incubation, approximately 90% lower than plain PTX solution. Stability studies confirmed the robustness of the liposomal formulation under various storage conditions. These findings highlight the potential of PEGylated pH-responsive liposomes as effective nanocarriers for enhancing PTX therapy against breast cancer.


Subject(s)
Breast Neoplasms , Drug Liberation , Liposomes , Paclitaxel , Polyethylene Glycols , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Paclitaxel/pharmacokinetics , Paclitaxel/chemistry , Humans , Liposomes/chemistry , MCF-7 Cells , Hydrogen-Ion Concentration , Polyethylene Glycols/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/chemistry , Particle Size , Cell Survival/drug effects , Drug Delivery Systems/methods
2.
Carbohydr Polym ; 346: 122645, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245506

ABSTRACT

Melanoma either intrinsically possesses resistance or rapidly acquires resistance to anti-tumor therapy, which often leads to local recurrence or distant metastasis after resection. In this study, we found histone 3 lysine 27 (H3K27) demethylated by an inhibitor of histone methyltransferase EZH2 could epigenetically reverse the resistance to chemo-drug paclitaxel (PTX), or enhance the efficacy of immune checkpoint inhibitor anti-TIGIT via downregulating TIGIT ligand CD155. Next, to address the complexity in the combination of multiple bioactive molecules with distinct therapeutic properties, we developed a polysaccharides-based organohydrogel (OHG) configured with a heterogenous network. Therein, hydroxypropyl chitosan (HPC)-stabilized emulsions for hydrophobic drug entrapment were crosslinked with oxidized dextran (Odex) to form a hydrophilic gel matrix to facilitate antibody accommodation, which demonstrated a tunable sustained release profile by optimizing emulsion/gel volume ratios. As results, local injection of OHG loaded with EZH2 inhibitor UNC1999, PTX and anti-TIGIT did not only synergistically enhance the cytotoxicity of PTX, but also reprogrammed the immune resistance via bi-directionally blocking TIGIT/CD155 axis, leading to the recruitment of cytotoxic effector cells into tumor and conferring a systemic immune memory to prevent lung metastasis. Hence, this polysaccharides-based OHG represents a potential in-situ epigenetic-, chemo- and immunotherapy platform to treat unresectable metastatic melanoma.


Subject(s)
Chitosan , Dextrans , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Melanoma , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Chitosan/chemistry , Chitosan/analogs & derivatives , Dextrans/chemistry , Animals , Melanoma/drug therapy , Melanoma/pathology , Melanoma/immunology , Mice , Humans , Epigenesis, Genetic/drug effects , Paclitaxel/pharmacology , Paclitaxel/chemistry , Paclitaxel/therapeutic use , Drug Resistance, Neoplasm/drug effects , Hydrogels/chemistry , Cell Line, Tumor , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
3.
Carbohydr Polym ; 346: 122596, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245487

ABSTRACT

Drug treatment of glioblastoma, the most aggressive and widespread form of brain cancer, is complicated due to the difficulty of penetration of chemotherapeutic drugs through the blood-brain barrier (BBB). Moreover, with surgical removal of tumors, in 90 % of cases they reappear near the original focus. To solve this problem, we propose to use hydrogel based on cellulose nanocrystals grafted with poly(N-isopropylacrylamide) (CNC-g-PNIPAM) as a promising material for filling postoperative cavities in the brain with the release of antitumor drugs. The CNC-g-PNIPAM is formed by "grafting to" method for precise control of molecular weight and grafting density. This colloidal system is liquid under injection conditions (at r. t.) and turns into a gel at human body temperature (when filling the postoperative area). It was shown for the first time that due to the rod-shaped of CNC, the gel has a fibrillar structure and, thus, mechanical properties similar to those of brain tissue, including nonlinear mechanics (strain-stiffening and compression softening). The biocompatibility of the hydrogel with primary brain cells is demonstrated. In addition, the release of the antitumor drug paclitaxel from the hydrogel and its antitumor activity is shown. The resulting nanocolloid system provides an innovative alternative approach to filling postoperative cavities and can be used for postoperative treatment due to the programmable release of drugs, as well as for in vitro modeling of tumor interaction with the BBB affecting drug transport in the brain.


Subject(s)
Acrylic Resins , Biocompatible Materials , Cellulose , Hydrogels , Nanoparticles , Cellulose/chemistry , Nanoparticles/chemistry , Acrylic Resins/chemistry , Humans , Animals , Biocompatible Materials/chemistry , Hydrogels/chemistry , Brain/drug effects , Brain/metabolism , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Temperature , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioblastoma/pathology , Drug Liberation , Blood-Brain Barrier/metabolism
4.
Carbohydr Polym ; 345: 122569, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39227105

ABSTRACT

Breast cancer is a malignant tumor that poses a significant threat to women's health and single therapy fails to play a good oncological therapeutic effect. Synergistic treatment with multiple strategies may make up for the deficiencies and has gained widespread attention. In this study, sulfhydryl-modified hyaluronic acid (HA-SH) was covalently crosslinked with polydopamine (PDA) via a Michael addition reaction to develop an injectable hydrogel, in which PDA can be used not only as a matrix but also as a photothermal agent. After HSA and paclitaxel were spontaneously organized into nanoparticles via hydrophobic interaction, hyaluronic acid with low molecular weight was covalently linked to HSA, thus conferring effectively delivery. This photothermal injectable hydrogel incorporates PTX@HSA-HA nanoparticles, thereby initiating a thermochemotherapeutic response to target malignancy. Our results demonstrated that this injectable hydrogel possesses consistent drug delivery capability in a murine breast cancer model, collaborating with photothermal therapy to effectively suppress tumor growth, represented by low expression of Ki-67 and increasing apoptosis. Photothermal therapy (PTT) can effectively stimulate immune response by increasing IL-6 and TNF-α. Notably, the treatment did not elicit any indications of toxicity. This injectable hydrogel holds significant promise as a multifaceted therapeutic agent that integrates photothermal and chemotherapeutic modalities.


Subject(s)
Breast Neoplasms , Hyaluronic Acid , Hydrogels , Paclitaxel , Photothermal Therapy , Animals , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Female , Breast Neoplasms/therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Mice , Photothermal Therapy/methods , Paclitaxel/pharmacology , Paclitaxel/chemistry , Paclitaxel/administration & dosage , Humans , Indoles/chemistry , Indoles/pharmacology , Mice, Inbred BALB C , Polymers/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Nanoparticles/chemistry , Drug Carriers/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/administration & dosage , Drug Delivery Systems/methods , Phototherapy/methods
5.
ACS Biomater Sci Eng ; 10(9): 5666-5674, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39166920

ABSTRACT

Microtubules (MTs) are widely recognized as targets for cancer therapies. They are directly related to unique mechanical properties, closely dependent on MT architecture and tubulin molecular features. Taxol is known to affect tubulin interactions resulting in the stabilization of the MT lattice, and thus the hierarchical organization stability, mechanics, and function. A deeper understanding of the molecular mechanisms through which taxol modulates intertubulin interactions in the MT lattice, and consequently, its stability and mechanical response is crucial to characterize how MT properties are regulated by environmental factors, such as interacting ligands. In this study, a computational analysis of the effect of taxol on the MT was performed at different scales, combining molecular dynamics simulation, dynamical network analysis, and elastic network modeling. The results show that the taxol-induced conformational differences at the M-loop region increase the stability of the lateral interactions and the amount of surface in contact between laterally coupled tubulins. Moreover, the conformational rearrangements in the taxane binding site result in a different structural communication pattern. Finally, the different conformation of the tubulin heterodimers and the stabilized lateral interactions resulted in a tendency toward higher deformation of the vibrating MT in the presence of taxol. Overall, this work provides additional insights into taxol-induced stabilization and relates the conformational changes at the tubulin level to the MT mechanics. Besides providing useful insights into taxol effect on MT mechanics, a methodological framework that could be used to characterize the effects of other MT stabilizing agents is presented.


Subject(s)
Microtubules , Molecular Dynamics Simulation , Paclitaxel , Tubulin , Paclitaxel/pharmacology , Paclitaxel/chemistry , Microtubules/metabolism , Microtubules/drug effects , Tubulin/metabolism , Tubulin/chemistry , Binding Sites , Humans
6.
Int J Pharm ; 663: 124570, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39134291

ABSTRACT

A multi-component paclitaxel (PTX) -loaded ß-elemene nanoemulsion by transferrin modification (Tf-PE-MEs) was developed to enhance non-small-cell lung cancer (NSCLC) treatment. After transferrin modification, the particle size of Tf-PE-MEs was (14.87 ± 1.84) nm, and the zeta potential was (-10.19 ± 0.870) mV, respectively. In vitro experiments showed that Tf-PE-MEs induced massive apoptosis in A549 cells, indicating that it had significant cytotoxicity to A549 cells. Through transferrin modification, Tf-PE-MEs accumulated at the tumor site efficiently with overexpressed transferrin receptor (TfR) on the surface of A549 cells. This will allow increasing PTX and ß-elemene concentration in the target cells, enhancing the therapeutic effect. Compared to PTX alone, Tf-PE-MEs displayed good anti-tumor efficacy and diminished systemic toxicity in vivo studies. With favourable therapeutic potential, this study provides a new strategy for the combined anticancer treatment of non-small cell lung cancer.


Subject(s)
Antineoplastic Agents, Phytogenic , Apoptosis , Carcinoma, Non-Small-Cell Lung , Emulsions , Lung Neoplasms , Nanoparticles , Paclitaxel , Sesquiterpenes , Transferrin , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Paclitaxel/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Transferrin/chemistry , Transferrin/administration & dosage , Lung Neoplasms/drug therapy , Animals , A549 Cells , Sesquiterpenes/pharmacology , Sesquiterpenes/chemistry , Sesquiterpenes/administration & dosage , Apoptosis/drug effects , Nanoparticles/chemistry , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Mice, Inbred BALB C , Mice, Nude , Receptors, Transferrin/metabolism , Particle Size , Mice , Cell Line, Tumor , Male , Drug Liberation , Cell Survival/drug effects
7.
Colloids Surf B Biointerfaces ; 244: 114136, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39116602

ABSTRACT

Aggregation-induced emission (AIE) materials are attracting great attention in biomedical fields such as sensors, bioimaging, and cancer treatment, et al. due to their strong fluorescence emission in the aggregated state. In this contribution, a series of tetraphenylene-acetonitrile AIE compounds with D-A-D' structures were synthesized by Suzuki coupling reaction and Knoevenagel condensation, and their relationship of chemical structure and fluorescence properties was investigated in detail, among which TPPA compound was selected as the monomer owing to the longest emission wavelength at about 530 nm with low energy band gap ΔE 3.09 eV of neutral TPPA and 1.43 eV of protonated TPPA. Novel amphiphilic AIE PEG-TA copolymers were prepared by RAFT polymerization of TPPA and PEGMA with about 1.44×104 Mw and narrow PDI, and the molar ratio of TPPA in the PEG-TA1 and PEG-TA2 copolymers was about 23.4 % and 29.6 %. The as-prepared PEG-TA copolymers would self-assembled in aqueous solution to form core-shell structures with a diameter of 150-200 nm, and their emission wavelength could reversibly convert from 545 nm to 650 nm with excellent pH sensitivity. The CLSM images showed that the PEG-TA FONs and PTX drugs-loaded PTX-TA FONs could be endocytosed by cells and mainly enriched in the cytoplasm, and CCK-8 results showed that the PEG-TA FONs had excellent biocompatibility but PTX-TA FONs had high inhibition ratio for A549 cells, moreover, the flow cytometry also showed that PTX-TA FONs could result in the apoptosis of A549 cells with some extent anti-tumor effect.


Subject(s)
Acetonitriles , Drug Delivery Systems , Paclitaxel , Humans , Structure-Activity Relationship , Paclitaxel/pharmacology , Paclitaxel/chemistry , Acetonitriles/chemistry , Acetonitriles/pharmacology , A549 Cells , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Molecular Structure , Cell Proliferation/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/chemical synthesis , Particle Size , Stilbenes/chemistry , Stilbenes/pharmacology , Stilbenes/chemical synthesis , Drug Liberation
8.
ACS Appl Mater Interfaces ; 16(36): 47325-47336, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39190919

ABSTRACT

Herein, we developed a paclitaxel prodrug (PSFc) through the conjugation of paclitaxel (PTX) and ferrocene via a redox-responsive disulfide bond. PSFc displays acid-enhanced catalytic activity of Fenton reaction and is capable of forming stable nanoparticles (PSFc NPs) through the assembly with distearoyl phosphoethanolamine-PEG2000. After being endocytosed, PSFc NPs could release PTX to promote cell apoptosis in response to overexpressed redox-active species of tumor cells. Meanwhile, the ferrocene-mediated Fenton reaction promotes intracellular accumulation of hydroxyl radicals and depletion of glutathione, thus leading to ferroptosis. Compared with the clinically used Taxol, PSFc NPs exhibited more potent in vivo antitumor outcomes through the combined effect of chemotherapy and ferroptosis. This study may offer insight into a facile design of a prodrug integrating different tumor treatment methods for combating malignant tumors.


Subject(s)
Ferroptosis , Ferrous Compounds , Metallocenes , Paclitaxel , Prodrugs , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Metallocenes/chemistry , Ferrous Compounds/chemistry , Ferrous Compounds/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Humans , Animals , Mice , Ferroptosis/drug effects , Cell Line, Tumor , Female , Mice, Inbred BALB C , Nanoparticles/chemistry , Apoptosis/drug effects , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology
9.
ACS Appl Mater Interfaces ; 16(33): 43350-43363, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39106360

ABSTRACT

We present a modular strategy to synthesize nanoparticle sensors equipped with dithiomaleimide-based, fluorescent molecular reporters capable of discerning minute changes in interparticle chemical environments based on fluorescence lifetime analysis. Three types of nanoparticles were synthesized with the aid of tailor-made molecular reporters, and it was found that protein nanoparticles exhibited greater sensitivity to changes in the core environment than polymer nanogels and block copolymer micelles. Encapsulation of the hydrophobic small-molecule drug paclitaxel (PTX) in self-reporting protein nanoparticles induced characteristic changes in fluorescence lifetime profiles, detected via time-resolved fluorescence spectroscopy. Depending on the mode of drug encapsulation, self-reporting protein nanoparticles revealed pronounced differences in their fluorescence lifetime signatures, which correlated with burst- vs diffusion-controlled release profiles observed in previous reports. Self-reporting nanoparticles, such as the ones developed here, will be critical for unraveling nanoparticle stability and nanoparticle-drug interactions, informing the future development of rationally engineered nanoparticle-based drug carriers.


Subject(s)
Nanoparticles , Paclitaxel , Nanoparticles/chemistry , Paclitaxel/chemistry , Paclitaxel/pharmacology , Drug Carriers/chemistry , Spectrometry, Fluorescence , Micelles , Proteins/chemistry
10.
ACS Appl Mater Interfaces ; 16(33): 43257-43271, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39119624

ABSTRACT

Prostate cancer presents as a challenging disease, as it is often characterized as an immunologically "cold" tumor, leading to suboptimal outcomes with current immunotherapeutic approaches in clinical settings. Photodynamic therapy (PDT) harnesses reactive oxygen species generated by photosensitizers (PSs) to disrupt the intracellular redox equilibrium. This process induces DNA damage in both the mitochondria and nucleus, activating the process of immunogenic cell death (ICD) and the cGAS-STING pathway. Ultimately, this cascade of events leads to the initiation of antitumor immune responses. Nevertheless, existing PSs face challenges, including suboptimal tumor targeting, aggregation-induced quenching, and insufficient oxygen levels in the tumor regions. To this end, a versatile bionic nanoplatform has been designed for the simultaneous delivery of the aggregation-induced emission PS TPAQ-Py-PF6 and paclitaxel (PTX). The cell membrane camouflage of the nanoplatform leads to its remarkable abilities in tumor targeting and cellular internalization. Upon laser irradiation, the utilization of TPAQ-Py-PF6 in conjunction with PTX showcases a notable and enhanced synergistic antitumor impact. Additionally, the nanoplatform has the capability of initiating the cGAS-STING pathway, leading to the generation of cytokines. The presence of damage-associated molecular patterns induced by ICD collaborates with these aforementioned cytokines lead to the recruitment and facilitation of dendritic cell maturation. Consequently, this elicits a systemic immune response against tumors. In summary, this promising strategy highlights the use of a multifunctional biomimetic nanoplatform, combining chemotherapy, PDT, and immunotherapy to enhance the effectiveness of antitumor treatment.


Subject(s)
Immunogenic Cell Death , Immunotherapy , Membrane Proteins , Nucleotidyltransferases , Photochemotherapy , Photosensitizing Agents , Prostatic Neoplasms , Humans , Nucleotidyltransferases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Immunogenic Cell Death/drug effects , Immunogenic Cell Death/radiation effects , Membrane Proteins/metabolism , Male , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Animals , Mice , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Cell Line, Tumor , Drug Delivery Systems , Porphyrins/chemistry , Porphyrins/pharmacology
11.
Int J Nanomedicine ; 19: 8237-8251, 2024.
Article in English | MEDLINE | ID: mdl-39157735

ABSTRACT

Background: Breast cancer presents significant challenges due to the limited effectiveness of available treatments and the high likelihood of recurrence. iRGD possesses both RGD sequence and C-terminal sequence and has dual functions of targeting and membrane penetration. iRGD-modified nanocarriers can enhance drug targeting of tumor vascular endothelial cells and penetration of new microvessels, increasing drug concentration in tumor tissues. Methods: The amidation reaction was carried out between SiO2/AuNCs and iRGD/PTX, yielding a conjugated drug delivery system (SiO2/AuNCs-iRGD/PTX, SAIP@NPs). The assessment encompassed the characterization of the morphology, particle size distribution, physicochemical properties, in vitro release profile, cytotoxicity, and cellular uptake of SAIP@NPs. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed using a small animal in vivo imaging system and a tumor-bearing nude mice model, respectively. The tumor targeting and anti-tumor efficacy of SAIP@NPs were assessed utilizing a small animal in vivo imaging system and an in situ nude mice breast cancer xenograft model, respectively. Results: The prepared SAIP@NPs exhibited decent stability and a certain slow-release effect in phosphate buffer (PBS, pH 7.4). In vitro studies had shown that, due to the dual functions of transmembrane and targeting of iRGD peptide, SAIP@NPs exhibited strong binding to integrin αvß3, which was highly expressed on the membrane of MDA-MB-231 cells, improving the uptake capacity of tumor cells, inhibiting the rapid growth of tumor cells, and promoting tumor cell apoptosis. The results of animal experiments further proved that SAIP@NPs had longer residence time in tumor sites, stronger anti-tumor effect, and no obvious toxicity to major organs of experimental animals. Conclusion: The engineered SAIP@NPs exhibited superior functionalities including efficient membrane permeability, precise tumor targeting, and imaging, thereby significantly augmenting the therapeutic efficacy against breast cancer with a favorable safety profile.


Subject(s)
Breast Neoplasms , Gold , Metal Nanoparticles , Mice, Nude , Oligopeptides , Silicon Dioxide , Animals , Silicon Dioxide/chemistry , Female , Breast Neoplasms/drug therapy , Humans , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Mice , Cell Line, Tumor , Metal Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Mice, Inbred BALB C , Paclitaxel/chemistry , Paclitaxel/pharmacology , Paclitaxel/pharmacokinetics , Paclitaxel/administration & dosage , Drug Delivery Systems/methods , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Particle Size , MCF-7 Cells
12.
Molecules ; 29(16)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39202850

ABSTRACT

A ZnO-Graphene oxide nanocomposite (Z-G) was prepared in order to exploit the biomedical features of each component in a single anticancer material. This was achieved by means of an environmentally friendly synthesis, taking place at a low temperature and without the involvement of toxic reagents. The product was physicochemically characterized. The ZnO-to-GO ratio was determined through thermogravimetric analysis, while scanning electron microscopy and transmission electron microscopy were used to provide insight into the morphology of the nanocomposite. Using energy-dispersive X-ray spectroscopy, it was possible to confirm that the graphene flakes were homogeneously coated with ZnO. The crystallite size of the ZnO nanoparticles in the new composite was determined using X-ray powder diffraction. The capacity of Z-G to enhance the toxicity of the anticancer drug Paclitaxel towards breast cancer cells was assessed via a cell viability study, showing the remarkable anticancer activity of the obtained system. Such results support the potential use of Z-G as an anticancer agent in combination with a common chemotherapeutic like Paclitaxel, leading to new chemotherapeutic formulations.


Subject(s)
Breast Neoplasms , Cell Survival , Graphite , Nanocomposites , Paclitaxel , Zinc Oxide , Graphite/chemistry , Humans , Zinc Oxide/chemistry , Zinc Oxide/pharmacology , Paclitaxel/pharmacology , Paclitaxel/chemistry , Nanocomposites/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Survival/drug effects , Female , Cell Line, Tumor , Drug Carriers/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Delivery Systems , MCF-7 Cells , X-Ray Diffraction
13.
Eur J Med Chem ; 277: 116761, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39151276

ABSTRACT

The P-glycoprotein (ABCB1)-mediated multidrug resistance (MDR) has emerged as a significant impediment to the efficacy of cancer chemotherapy in clinical therapy, which could promote the development of effective agents for MDR reversal. In this work, we reported the exploration of novel pyrazolo [1,5-a]pyrimidine derivatives as potent reversal agents capable of enhancing the sensitivity of ABCB1-mediated MDR MCF-7/ADR cells to paclitaxel (PTX). Among them, compound 16q remarkably increased the sensitivity of MCF-7/ADR cells to PTX at 5 µM (IC50 = 27.00 nM, RF = 247.40) and 10 µM (IC50 = 10.07 nM, RF = 663.44). Compound 16q could effectively bind and stabilize ABCB1, and does not affect the expression and subcellular localization of ABCB1 in MCF-7/ADR cells. Compound 16q inhibited the function of ABCB1, thereby increasing PTX accumulation, and interrupting the accumulation and efflux of the ABCB1-mediated Rh123, thus resulting in exhibiting good reversal effects. In addition, due to the potent reversal effects of compound 16q, the abilities of PTX to inhibit tubulin depolymerization, and induce cell cycle arrest and apoptosis in MCF-7/ADR cells under low-dose conditions were restored. These results indicate that compound 16q might be a promising potent reversal agent capable of revising ABCB1-mediated MDR, and pyrazolo [1,5-a]pyrimidine might represent a novel scaffold for the discovery of new ABCB1-mediated MDR reversal agents.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Antineoplastic Agents , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Pyrazoles , Pyrimidines , Humans , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Drug Resistance, Multiple/drug effects , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Pyrazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/chemical synthesis , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Molecular Structure , Paclitaxel/pharmacology , Paclitaxel/chemistry , MCF-7 Cells , Drug Discovery , Dose-Response Relationship, Drug , Cell Proliferation/drug effects , Apoptosis/drug effects
14.
Sci Rep ; 14(1): 19516, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174603

ABSTRACT

The effect of chemotherapy for anti-glioblastoma is limited due to insufficient drug delivery across the blood-brain-barrier. Poloxamer 188-coated nanoparticles can enhance the delivery of nanoparticles across the blood-brain-barrier. This study presents the design, preparation, and evaluation of a combination of PLGA nanoparticles (PLGA NPs) loaded with methotrexate (P-MTX NPs) and PLGA nanoparticles loaded with paclitaxel (P-PTX NPs), both of which were surface-modified with poloxamer188. Cranial tumors were induced by implanting C6 cells in a rat model and MRI demonstrated that the tumors were indistinguishable in the two rats with P-MTX NPs + P-PTX NPs treated groups. Brain PET scans exhibited a decreased brain-to-background ratio which could be attributed to the diminished metabolic tumor volume. The expression of Ki-67 as a poor prognosis factor, was significantly lower in P-MTX NPs + P-PTX NPs compared to the control. Furthermore, the biodistribution of PLGA NPs was determined by carbon quantum dots loaded into PLGA NPs (P-CQD NPs), and quantitative analysis of ex-vivo imaging of the dissected organs demonstrated that 17.2 ± 0.6% of the NPs were concentrated in the brain after 48 h. The findings highlight the efficacy of combination nanochemotherapy in glioblastoma treatment, indicating the need for further preclinical studies.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms , Glioblastoma , Methotrexate , Nanoparticles , Poloxamer , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/diagnostic imaging , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Nanoparticles/chemistry , Rats , Poloxamer/chemistry , Methotrexate/chemistry , Methotrexate/administration & dosage , Methotrexate/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Cell Line, Tumor , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Tissue Distribution , Drug Carriers/chemistry , Male , Drug Delivery Systems , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans
15.
Int J Nanomedicine ; 19: 7775-7797, 2024.
Article in English | MEDLINE | ID: mdl-39099795

ABSTRACT

Purpose: The present study aimed to develop a lipid nanoplatform, denoted as "BAL-PTX-LN", co-loaded with chiral baicalin derivatives (BAL) and paclitaxel (PTX) to promote the anti-lung cancer efficacy of paclitaxel and reduce the toxicity of chemotherapeutic drugs. Methods: BAL-PTX-LN was optimized through central composite design based on a single-factor experiments. BAL-PTX-LN was evaluated by TEM, particle size, encapsulation efficiency, hemolysis rate, release kinetics and stability. And was evaluated by pharmacokinetics and the antitumor efficacy studied both in vitro and in vivo. The in vivo safety profile of the formulation was assessed using hematoxylin and eosin (HE) staining. Results: BAL-PTX-LN exhibited spherical morphology with a particle size of 134.36 ± 3.18 nm, PDI of 0.24 ± 0.02, and with an encapsulation efficiency exceeding 90%, BAL-PTX-LN remained stable after 180 days storage. In vitro release studies revealed a zero-order kinetic model of PTX from the liposomal formulation. No hemolysis was observed in the preparation group. Pharmacokinetic analysis of PTX in the BAL-PTX-LN group revealed an approximately three-fold higher bioavailability and twice longer t1/2 compared to the bulk drug group. Furthermore, the IC50 of BAL-PTX-LN decreased by 2.35 times (13.48 µg/mL vs 31.722 µg/mL) and the apoptosis rate increased by 1.82 times (29.38% vs 16.13%) at 24 h compared to the PTX group. In tumor-bearing nude mice, the BAL-PTX-LN formulation exhibited a two-fold higher tumor inhibition rate compared to the PTX group (62.83% vs 29.95%), accompanied by a ten-fold decrease in Ki67 expression (4.26% vs 45.88%). Interestingly, HE staining revealed no pathological changes in tissues from the BAL-PTX-LN group, whereas tissues from the PTX group exhibited pathological changes and tumor cell infiltration. Conclusion: BAL-PTX-LN improves the therapeutic effect of poorly soluble chemotherapeutic drugs on lung cancer, which is anticipated to emerge as a viable therapeutic agent for lung cancer in clinical applications.


Subject(s)
Lung Neoplasms , Paclitaxel , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Humans , Flavonoids/chemistry , Flavonoids/pharmacology , Flavonoids/pharmacokinetics , Flavonoids/administration & dosage , Particle Size , Nanoparticles/chemistry , Mice , Liposomes/chemistry , Liposomes/pharmacokinetics , A549 Cells , Lipids/chemistry , Male , Mice, Inbred BALB C , Cell Line, Tumor , Drug Liberation , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Mice, Nude , Hemolysis/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage
16.
Bioconjug Chem ; 35(9): 1429-1440, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39159059

ABSTRACT

Drug delivery to the esophagus through systemic administration remains challenging, as minimal drug reaches the desired target. Local delivery offers the potential for improved efficacy while minimizing off-target toxicities but necessitates bioadhesive properties for mucosal delivery. Herein, we describe the synthesis of two new mucoadhesive amphiphilic copolymers prepared by sequential ring-opening copolymerization or postpolymerization click conjugation. Both strategies yield block copolymers containing a hydrophilic amine-functionalized poly-amido-saccharide and either a hydrophobic alkyl derivatized poly-amido-saccharide or poly(lactic acid), respectively. The latter resulting copolymers readily self-assemble into spherical, ≈200 nm diameter, positively charged mucoadhesive nanoparticles. The NPs entrap ultrahigh levels of paclitaxel via encapsulation of free paclitaxel and paclitaxel conjugated to a biodegradable, biocompatible poly(1,2-glycerol carbonate). Paclitaxel-loaded NPs rapidly enter cells, release paclitaxel, are cytotoxic to esophageal OE33 and OE19 tumor cells in vitro, and, importantly, demonstrate improved mucoadhesion compared to conventional poly(ethylene glycol)-poly(lactic acid) nanoparticles to ex vivo esophageal tissue.


Subject(s)
Nanoparticles , Paclitaxel , Polyesters , Paclitaxel/administration & dosage , Paclitaxel/chemistry , Paclitaxel/pharmacology , Nanoparticles/chemistry , Polyesters/chemistry , Humans , Cell Line, Tumor , Polymers/chemistry , Drug Carriers/chemistry , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Animals
17.
Int J Biol Macromol ; 277(Pt 3): 134509, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111508

ABSTRACT

Aiming to improve the retrieval rate of retrievable vena cava filters (RVCF) and extend its dwelling time in vivo, a novel hydrogel coating loaded with 10 mg/mL heparin and 30 mg/mL cyclodextrin/paclitaxel (PTX) inclusion complex (IC) was prepared. The drug-release behavior in the phosphate buffer solution demonstrated both heparin and PTX could be sustainably released over approximately two weeks. Furthermore, it was shown that the hydrogel-coated RVCF (HRVCF) with 10 mg/mL heparin and 30 mg/mL PTX IC effectively extended the blood clotting time to above the detection limit and inhibited EA.hy926 and CCC-SMC-1 cells' proliferation in vitro compared to the commercially available bare RVCF. Both the HRVCF and the bare RVCF were implanted into the vena cava of sheep and retrieved at at 2nd and 4th week after implantation, revealing that the HRVCF had a significantly higher retrieval rate of 67 % than the bare RVCF (0 %) at 4th week. Comprehensive analyses, including histological, immunohistological, and immunofluorescent assessments of the explanted veins demonstrated the HRVCF exhibited anti-hyperplasia and anticoagulation properties in vivo, attributable to the hydrogel coating, thereby improving the retrieval rate in sheep. Consequently, the as-prepared HRVCF shows promising potential for clinical application to enhance the retrieval rates of RVCFs.


Subject(s)
Cyclodextrins , Heparin , Hydrogels , Paclitaxel , Vena Cava Filters , Cyclodextrins/chemistry , Cyclodextrins/pharmacology , Paclitaxel/pharmacology , Paclitaxel/chemistry , Heparin/chemistry , Heparin/pharmacology , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Humans , Sheep , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Blood Coagulation/drug effects , Cell Proliferation/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Drug Liberation
18.
Biomed Pharmacother ; 177: 117102, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38991303

ABSTRACT

Paclitaxel (PTX) is a first-line drug for the treatment of lung cancer, but its targeting and therapeutic effect are unsatisfactory. Herein, lung cancer cell (A549) membrane biomimetic PTX-loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles (AM@PTX-NPs) were constructed to eliminate the shortcomings of PTX. The AM@PTX-NPs were successfully prepared with a high drug loading efficiency (10.90±0.06 %). Moreover, transmission electron microscopy, SDS-PAGE, and western blotting proved that AM@PTX-NPs were spherical nanoparticles camouflaged by the A549 cell membrane. Both in vitro and in vivo assays revealed that the AM@PTX-NPs displayed outstanding targeting capacity due to A549 membrane modification. The cytotoxicity experiment showed that the developed biomimetic formulation was able to effectively reduce the proliferation of A549 cells. Moreover, AM@PTX-NPs exhibited a significant tumor growth inhibition rate (73.00 %) with good safety in the tumor-bearing mice, which was higher than that of the PTX-NPs without A549 membrane coating (37.39 %). Overall, the constructed bioinspired vector could provide a novel platform for the PTX delivery and demonstrated a promising strategy for the targeted cancer treatment.


Subject(s)
Cell Membrane , Lung Neoplasms , Nanoparticles , Paclitaxel , Polylactic Acid-Polyglycolic Acid Copolymer , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Paclitaxel/chemistry , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , A549 Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Nanoparticles/chemistry , Mice , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Mice, Nude , Mice, Inbred BALB C , Xenograft Model Antitumor Assays , Drug Carriers/chemistry
19.
Chem Biol Drug Des ; 104(2): e14600, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39075030

ABSTRACT

Transmembrane protease/serine (TMPRSS2), a type II transmembrane serine protease, plays a crucial role in different stages of cancer. Recent studies have reported that the triggering epidermal growth factor receptor (EGFR) activation through protease action promotes metastasis. However, there are no reports on the interaction of TMPRSS2 with EGFR, especially in triple-negative triple negative (TNBC). The current study investigates the unexplored interaction between TMPRSS2 and EGFR, which are key partners mediating metastasis. This interaction is explored for potential targeting using quercetin (QUE) and taxifolin (TAX). TMPRSS2 expression patterns in breast cancer (BC) tissues and subtypes have been predicted, with the prognostic significance assessed using the GENT2.0 database. Validation of TMPRSS2 expression was performed in normal and TNBC tissues, including drug-resistant cell lines, utilizing GEO datasets. TMPRSS2 was further validated as a predictive biomarker for FDA-approved chemotherapeutics through transcriptomic data from BC patients. The study demonstrated the association of TMPRSS2 with EGFR through in silico analysis and validates the findings in TNBC cohorts using the TIMER2.0 web server and the TCGA dataset through C-Bioportal. Molecular docking and molecular dynamic simulation studies identified QUE and TAX as best leads targeting TMPRSS2. They inhibited cell-free TMPRSS2 activity like clinical inhibitor of TMPRSS2, Camostat mesylate. In cell-based assays focused on paclitaxel-resistant TNBC (TNBC/PR), QUE and TAX demonstrated potent inhibitory activity against extracellular and membrane-bound TMPRSS2, with low IC50 values. Furthermore, ELISA and cell-based AlphaLISA assays demonstrated that QUE and TAX inhibit the interaction of TMPRSS2 with EGFR. Additionally, QUE and TAX exhibited significant inhibition of proliferation and cell cycle accompanied by notable alterations in the morphology of TNBC/PR cells. This study provides valuable insights into potential of QUE and TAX targeting TMPRSS2 overexpressing TNBC.


Subject(s)
Drug Resistance, Neoplasm , ErbB Receptors , Molecular Docking Simulation , Paclitaxel , Quercetin , Serine Endopeptidases , Quercetin/pharmacology , Quercetin/chemistry , Quercetin/metabolism , Quercetin/analogs & derivatives , Humans , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Paclitaxel/pharmacology , Paclitaxel/chemistry , Female , Serine Endopeptidases/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Cell Proliferation/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology
20.
J Med Chem ; 67(15): 12711-12734, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39038808

ABSTRACT

Haspin is an emerging, but rather unexplored, divergent kinase involved in tumor growth by regulating the mitotic phase. In this paper, the in-silico design, synthesis, and biological characterization of a new series of substituted indoles acting as potent Haspin inhibitors are reported. The synthesized derivatives have been evaluated by FRET analysis, showing very potent Haspin inhibition. Then, a comprehensive in-cell investigation highlighted compounds 47 and 60 as the most promising inhibitors. These compounds were challenged for their synergic activity with paclitaxel in 2D and 3D cellular models, demonstrating a twofold improvement of the paclitaxel antitumor activity. Compound 60 also showed remarkable selectivity when tested in a panel of 70 diverse kinases. Finally, in-silico studies provided new insight about the chemical requirements useful to develop new Haspin inhibitors. Biological results, together with the drug-likeness profile of 47 and 60, make these derivatives deserving further studies.


Subject(s)
Indoles , Indoles/pharmacology , Indoles/chemistry , Indoles/chemical synthesis , Humans , Structure-Activity Relationship , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Paclitaxel/pharmacology , Paclitaxel/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor
SELECTION OF CITATIONS
SEARCH DETAIL