Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.131
Filtrer
1.
J Nanobiotechnology ; 22(1): 461, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090622

RÉSUMÉ

BACKGROUND: The combination of programmed cell death ligand-1 (PD-L1) immune checkpoint blockade (ICB) and immunogenic cell death (ICD)-inducing chemotherapy has shown promise in cancer immunotherapy. However, triple-negative breast cancer (TNBC) patients undergoing this treatment often face obstacles such as systemic toxicity and low response rates, primarily attributed to the immunosuppressive tumor microenvironment (TME). METHODS AND RESULTS: In this study, PD-L1-targeted theranostic systems were developed utilizing anti-PD-L1 peptide (APP) conjugated with a bio-orthogonal click chemistry group. Initially, TNBC was treated with azide-modified sugar to introduce azide groups onto tumor cell surfaces through metabolic glycoengineering. A PD-L1-targeted probe was developed to evaluate the PD-L1 status of TNBC using magnetic resonance/near-infrared fluorescence imaging. Subsequently, an acidic pH-responsive prodrug was employed to enhance tumor accumulation via bio-orthogonal click chemistry, which enhances PD-L1-targeted ICB, the pH-responsive DOX release and induction of pyroptosis-mediated ICD of TNBC. Combined PD-L1-targeted chemo-immunotherapy effectively reversed the immune-tolerant TME and elicited robust tumor-specific immune responses, resulting in significant inhibition of tumor progression. CONCLUSIONS: Our study has successfully engineered a bio-orthogonal multifunctional theranostic system, which employs bio-orthogonal click chemistry in conjunction with a PD-L1 targeting strategy. This innovative approach has been demonstrated to exhibit significant promise for both the targeted imaging and therapeutic intervention of TNBC.


Sujet(s)
Antigène CD274 , Chimie click , Immunothérapie , Pyroptose , Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/traitement médicamenteux , Antigène CD274/métabolisme , Animaux , Femelle , Immunothérapie/méthodes , Souris , Pyroptose/effets des médicaments et des substances chimiques , Humains , Lignée cellulaire tumorale , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/usage thérapeutique , Imagerie optique/méthodes , Promédicaments/composition chimique , Promédicaments/pharmacologie
2.
Int J Nanomedicine ; 19: 8071-8090, 2024.
Article de Anglais | MEDLINE | ID: mdl-39130685

RÉSUMÉ

Purpose: Chemotherapy mediated by Reactive oxygen species (ROS)-responsive drug delivery systems can potentially mitigate the toxic side effects of chemotherapeutic drugs and significantly enhance their therapeutic efficacy. However, achieving precise targeted drug delivery and real-time control of ROS-responsive drug release at tumor sites remains a formidable challenge. Therefore, this study aimed to describe a ROS-responsive drug delivery system with specific tumor targeting capabilities for mitigating chemotherapy-induced toxicity while enhancing therapeutic efficacy under guidance of Fluorescence (FL) and Magnetic resonance (MR) bimodal imaging. Methods: Indocyanine green (ICG), Doxorubicin (DOX) prodrug pB-DOX and Superparamagnetic iron oxide (SPIO, Fe3O4) were encapsulated in poly(lactic-co-glycolic acid) (PLGA) by double emulsification method to prepare ICG/ pB-DOX/ Fe3O4/ PLGA nanoparticles (IBFP NPs). The surface of IBFP NPs was functionalized with mammaglobin antibodies (mAbs) by carbodiimide method to construct the breast cancer-targeting mAbs/ IBFP NPs (MIBFP NPs). Thereafter, FL and MR bimodal imaging ability of MIBFP NPs was evaluated in vitro and in vivo. Finally, the combined photodynamic therapy (PDT) and chemotherapy efficacy evaluation based on MIBFP NPs was studied. Results: The multifunctional MIBFP NPs exhibited significant targeting efficacy for breast cancer. FL and MR bimodal imaging clearly displayed the distribution of the targeting MIBFP NPs in vivo. Upon near-infrared laser irradiation, the MIBFP NPs loaded with ICG effectively generated ROS for PDT, enabling precise tumor ablation. Simultaneously, it triggered activation of the pB-DOX by cleaving its sensitive moiety, thereby restoring DOX activity and achieving ROS-responsive targeted chemotherapy. Furthermore, the MIBFP NPs combined PDT and chemotherapy to enhance the efficiency of tumor ablation under guidance of bimodal imaging. Conclusion: MIBFP NPs constitute a novel dual-modality imaging-guided drug delivery system for targeted breast cancer therapy and offer precise and controlled combined treatment options.


Sujet(s)
Tumeurs du sein , Doxorubicine , Vert indocyanine , Imagerie par résonance magnétique , Photothérapie dynamique , Copolymère d'acide poly(lactique-co-glycolique) , Espèces réactives de l'oxygène , Vert indocyanine/composition chimique , Vert indocyanine/pharmacocinétique , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/administration et posologie , Doxorubicine/pharmacocinétique , Espèces réactives de l'oxygène/métabolisme , Animaux , Femelle , Humains , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique , Photothérapie dynamique/méthodes , Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Imagerie par résonance magnétique/méthodes , Souris , Lignée cellulaire tumorale , Systèmes de délivrance de médicaments/méthodes , Promédicaments/composition chimique , Promédicaments/pharmacocinétique , Promédicaments/pharmacologie , Souris de lignée BALB C , Nanoparticules magnétiques d'oxyde de fer/composition chimique , Souris nude , Nanoparticules de magnétite/composition chimique , Libération de médicament , Nanoparticules/composition chimique , Imagerie optique/méthodes
3.
Int J Nanomedicine ; 19: 7799-7816, 2024.
Article de Anglais | MEDLINE | ID: mdl-39099794

RÉSUMÉ

Background: At present, the few photothermal/chemotherapy studies about retinoblastoma that have been reported are mainly restricted to ectopic models involving subcutaneous implantation. However, eyeball is unique physiological structure, the blood-retina barrier (BRB) hinders the absorption of drug molecules through the systemic route. Moreover, the abundant blood circulation in the fundus accelerates drug metabolism. To uphold the required drug concentration, patients must undergo frequent chemotherapy sessions. Purpose: To address these challenges above, we need to develop a secure and effective drug delivery system (FA-PEG-PDA-DOX) for the fundus. Methods: We offered superior therapeutic efficacy with minimal or no side effects and successfully established orthotopic mouse models. We evaluated cellular uptake performance and targeting efficiency of FA-PEG-PDA-DOX nanosystem and assessed its synergistic antitumor effects in vitro and vivo. Biodistribution assessments were performed to determine the retention time and targeting efficiency of the NPs in vivo. Additionally, safety assessments were conducted. Results: Cell endocytosis rates of the FA-PEG-PDA-DOX+Laser group became 5.23 times that of the DOX group and 2.28 times that of FA-PEG-PDA-DOX group without irradiation. The fluorescence signal of FA-PEG-PDA-DOX persisted for more than 120 hours at the tumor site. The number of tumor cells (17.2%) in the proliferative cycle decreased by 61.6% in the photothermal-chemotherapy group, in contrast to that of the saline control group (78.8%). FA-PEG-PDA-DOX nanoparticles(NPs) exhibited favorable biosafety and high biocompatibility. Conclusion: The dual functional targeted nanosystem, with the effects of DOX and mild-temperature elevation by irradiation, resulted in precise chemo/photothermal therapy in nude mice model.


Sujet(s)
Doxorubicine , Indoles , Thérapie photothermique , Polymères , Rétinoblastome , Animaux , Rétinoblastome/thérapie , Doxorubicine/composition chimique , Doxorubicine/pharmacocinétique , Doxorubicine/pharmacologie , Doxorubicine/administration et posologie , Souris , Thérapie photothermique/méthodes , Humains , Indoles/composition chimique , Indoles/pharmacocinétique , Indoles/pharmacologie , Lignée cellulaire tumorale , Polymères/composition chimique , Distribution tissulaire , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacocinétique , Souris nude , Nanoparticules/composition chimique , Systèmes de délivrance de médicaments/méthodes , Tumeurs de la rétine/thérapie , Tumeurs de la rétine/traitement médicamenteux , Souris de lignée BALB C , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/composition chimique , Antibiotiques antinéoplasiques/pharmacocinétique , Antibiotiques antinéoplasiques/administration et posologie , Modèles animaux de maladie humaine , Tests d'activité antitumorale sur modèle de xénogreffe , Système d'administration de médicaments à base de nanoparticules/composition chimique , Système d'administration de médicaments à base de nanoparticules/pharmacocinétique
4.
Biomacromolecules ; 25(8): 4991-5007, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39087761

RÉSUMÉ

The GPS-Nanoconveyor (MA-NV@DOX-Cas13a) is a targeted nanoplatform designed for the imaging and gene/chemotherapy synergistic treatment of melanoma. It utilizes rolling circle amplification (RCA) products as a scaffold to construct a DNA "Nanoconveyor" (NV), which incorporates a multivalent aptamer (MA) as a "GPS", encapsulates doxorubicin (DOX) in the transporter, and equips it with CRISPR/Cas13a ribonucleoproteins (Cas13a RNP). Carrying MA enhances the ability to recognize the overexpressed receptor nucleolin on B16 cells, enabling targeted imaging and precise delivery of MA-NV@DOX-Cas13a through receptor-mediated endocytosis. The activation of signal transducer and activator of transcription 3 (STAT3) in cancer cells triggers cis-cleavage of CRISPR/Cas13a, initiating its trans-cleavage function. Additionally, deoxyribonuclease I (DNase I) degrades MA-NV, releasing DOX for intracellular imaging and as a chemotherapeutic agent. Experiments demonstrate the superior capabilities of this versatile nanoplatform for cellular imaging and co-treatment while highlighting the advantages of these nanodrug delivery systems in mitigating DOX side effects.


Sujet(s)
Systèmes CRISPR-Cas , Doxorubicine , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/administration et posologie , Animaux , Souris , Humains , Aptamères nucléotidiques/composition chimique , Techniques d'amplification d'acides nucléiques/méthodes , Lignée cellulaire tumorale , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/administration et posologie , Antibiotiques antinéoplasiques/composition chimique
5.
Sci Rep ; 14(1): 18311, 2024 08 07.
Article de Anglais | MEDLINE | ID: mdl-39112669

RÉSUMÉ

Finding a novel drug delivery system (DDS) represents one of the most challenging endeavors in cancer therapy. Hence, in this study, we developed a new biocompatible and biodegradable zinc-based nanoscale metal-organic framework (Zn-NMOF) coated with folic acid (FA) functionalized chitosan (CS) to facilitate targeted delivery of doxorubicin (D), a standard chemotherapeutic agent, into breast cancer cells. The synthesis of the NMOF-CS-FA-D nanocomposite preceded its comprehensive characterization via FT-IR, DLS, XRD, SEM, and TEM analyses. Subsequent in vitro studies were conducted on MCF-7 breast cancer cells and HFF-1 normal cells, encompassing assessments of cell viability, expression levels of apoptotic and autophagy genes, cell cycle arrest, and apoptotic analyses. The size of the NMOF-CS-FA-D particles was determined to be less than 80 nm, with a drug loading efficiency of 72 ± 5%. The release kinetics of DOX from the nanocomposite were investigated, revealing controlled release behavior at pH 7.4 and accelerated release at pH 5.0, which is conducive to drug delivery into cancer cells. In vitro results indicated a 17.39% ± 6.34 cell viability after 24 h of treatment with a 40 nM concentration of the NMOF-CS-FA-D nanocomposite. Furthermore, the expression levels of Caspase-9 and BAX, key apoptotic genes, along with BECLIN1, an autophagy gene, were found to increase by two-fold, four-fold, and two-fold, respectively, following 5 h of treatment with the nanocomposite. Additionally, analysis of cell cycle distribution revealed 15.4 ± 2% of cells in the sub-G1 phase, indicative of apoptotic cells, and 31.9% of cells undergoing early and late apoptosis in MCF-7 cells. Collectively, these findings underscore the potential of the NMOF-CS-FA-D nanocomposite in inhibiting cancer cell proliferation with low side effects.


Sujet(s)
Apoptose , Tumeurs du sein , Chitosane , Doxorubicine , Réseaux organométalliques , Nanocomposites , Zinc , Humains , Nanocomposites/composition chimique , Réseaux organométalliques/composition chimique , Réseaux organométalliques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Cellules MCF-7 , Zinc/composition chimique , Zinc/pharmacologie , Chitosane/composition chimique , Femelle , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Acide folique/composition chimique , Acide folique/pharmacologie , Systèmes de délivrance de médicaments , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Libération de médicament , Vecteurs de médicaments/composition chimique , Caspase-9/métabolisme , Caspase-9/génétique , Autophagie/effets des médicaments et des substances chimiques
6.
Sci Rep ; 14(1): 18544, 2024 08 09.
Article de Anglais | MEDLINE | ID: mdl-39122867

RÉSUMÉ

Breast cancer is one of cancer's most deadly varieties. Its variability makes the development of personalized therapies very difficult. Therefore, improvement of classic chemotherapy is still one of the important challenges of cancer research. We addressed this issue applying nanotechnology to verify the influence of silver nanoparticles (AgNPs) on doxorubicin (DOX) anticancer activity and assess if the size of AgNPs affects their interactions with DOX. We employed a broad spectrum of biophysical methods, characterizing 5 and 50 nm AgNPs interactions with DOX using UV-Vis spectroscopy, dynamic light scattering, fluorescence spectroscopy, and atomic force microscopy imaging. Biological effects of observed AgNPs-DOX interactions were assessed utilizing MTT and 3D Matrigel assays on SKBR3 and MDA-MB-231 breast cancer cell lines. Obtained results indicate direct interactions between AgNPs and DOX. Furthermore, AgNPs size influences their interactions with DOX, as evidenced by differences in the heteroaggregates formation observed in biophysical experiments and further supported by in vitro biological assays. We detected reduction of tumor cell viability and/or colony sizes of the analyzed cancer cell lines, registering differences linked to the observed phenomenon. However, the effects may be limited to the outer borders of the tumor microenvironment as evidenced by the 3D model. Summing up, we observed diverse patterns of interactions and biological effects for different sizes of AgNPs with DOX providing insight how the nanoparticles' size affects their interactions with other biologically active compounds. Moreover, obtained data can be further used in experiments on the reduction of tumor size i.e. before the surgical intervention.


Sujet(s)
Doxorubicine , Nanoparticules métalliques , Taille de particule , Argent , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Humains , Argent/composition chimique , Argent/pharmacologie , Nanoparticules métalliques/composition chimique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie
7.
J Am Chem Soc ; 146(32): 22675-22688, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39088029

RÉSUMÉ

Redox-responsive homodimer prodrug nanoassemblies (RHPNs) have emerged as a significant technology for overcoming chemotherapeutical limitations due to their high drug-loading capacity, low excipient-associated toxicity, and straightforward preparation method. Previous studies indicated that α-position disulfide bond bridged RHPNs exhibited rapid drug release rates but unsatisfactory assembly stability. In contrast, γ-disulfide bond bridged RHPNs showed better assembly stability but low drug release rates. Therefore, designing chemical linkages that ensure both stable assembly and rapid drug release remains challenging. To address this paradox of stable assembly and rapid drug release in RHPNs, we developed carbon-spaced double-disulfide bond (CSDD)-bridged RHPNs (CSDD-RHPNs) with two carbon-spaces. Pilot studies showed that CSDD-RHPNs with two carbon-spaces exhibited enhanced assembly stability, reduction-responsive drug release, and improved selective toxicity compared to α-/γ-position single disulfide bond bridged RHPNs. Based on these findings, CSDD-RHPNs with four and six carbon-spaces were designed to further investigate the properties of CSDD-RHPNs. These CSDD-RHPNs exhibited excellent assembly ability, safety, and prolonged circulation. Particularly, CSDD-RHPNs with two carbon-spaces displayed the best antitumor efficacy on 4T1 and B16-F10 tumor-bearing mice. CSDD chemical linkages offer novel perspectives on the rational design of RHPNs, potentially overcoming the design limitations regarding contradictory assembly ability and drug release rate.


Sujet(s)
Carbone , Disulfures , Promédicaments , Disulfures/composition chimique , Promédicaments/composition chimique , Animaux , Souris , Carbone/composition chimique , Humains , Libération de médicament , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Conception de médicament , Lignée cellulaire tumorale , Nanostructures/composition chimique , Dimérisation , Doxorubicine/composition chimique , Doxorubicine/pharmacologie
8.
Sensors (Basel) ; 24(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39123900

RÉSUMÉ

Nanotechnology has ushered in significant advancements in drug design, revolutionizing the prevention, diagnosis, and treatment of various diseases. The strategic utilization of nanotechnology to enhance drug loading, delivery, and release has garnered increasing attention, leveraging the enhanced physical and chemical properties offered by these systems. Polyamidoamine (PAMAM) dendrimers have been pivotal in drug delivery, yet there is room for further enhancement. In this study, we conjugated PAMAM dendrimers with chitosan (CS) to augment cellular internalization in tumor cells. Specifically, doxorubicin (DOX) was initially loaded into PAMAM dendrimers to form DOX-loaded PAMAM (DOX@PAMAM) complexes via intermolecular forces. Subsequently, CS was linked onto the DOX-loaded PAMAM dendrimers to yield CS-conjugated PAMAM loaded with DOX (DOX@CS@PAMAM) through glutaraldehyde crosslinking via the Schiff base reaction. The resultant DOX@CS@PAMAM complexes were comprehensively characterized using Fourier-transform infrared (FTIR) spectroscopy, transmission electron microscopy (TEM), and dynamic light scattering (DLS). Notably, while the drug release profile of DOX@CS@PAMAM in acidic environments was inferior to that of DOX@PAMAM, DOX@CS@PAMAM demonstrated effective acid-responsive drug release, with a cumulative release of 70% within 25 h attributed to the imine linkage. Most importantly, DOX@CS@PAMAM exhibited significant selective cellular internalization rates and antitumor efficacy compared to DOX@PAMAM, as validated through cell viability assays, fluorescence imaging, and flow cytometry analysis. In summary, DOX@CS@PAMAM demonstrated superior antitumor effects compared to unconjugated PAMAM dendrimers, thereby broadening the scope of dendrimer-based nanomedicines with enhanced therapeutic efficacy and promising applications in cancer therapy.


Sujet(s)
Chitosane , Dendrimères , Doxorubicine , Dendrimères/composition chimique , Chitosane/composition chimique , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Humains , Polyamines/composition chimique , Vecteurs de médicaments/composition chimique , Libération de médicament , Survie cellulaire/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments/méthodes , Lignée cellulaire tumorale
9.
Molecules ; 29(15)2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39125024

RÉSUMÉ

Dimeric prodrugs have been investigated intensely as carrier-free drug self-delivery systems (DSDSs) in recent decades, and their stimuli-responsive drug release has usually been controlled by the conjugations between the drug molecules, including the stimuli (pH or redox) and responsive sensitivity. Here, an acid-triggered dimeric prodrug of doxorubicin (DOX) was synthesized by conjugating two DOX molecules with an acid-labile ketal linker. It possessed high drug content near the pure drug, while the premature drug leakage in blood circulation was efficiently suppressed. Furthermore, its aggregation structures were controlled by fabricating nanomedicines via different approaches, such as fast precipitation and slow self-assembly, to regulate the drug release performance. Such findings are expected to enable better anti-tumor efficacy with the desired drug release rate, beyond the molecular structure of the dimeric prodrug.


Sujet(s)
Doxorubicine , Systèmes de délivrance de médicaments , Libération de médicament , Promédicaments , Promédicaments/composition chimique , Promédicaments/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Humains , Concentration en ions d'hydrogène , Vecteurs de médicaments/composition chimique , Structure moléculaire
10.
J Mater Sci Mater Med ; 35(1): 48, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39136805

RÉSUMÉ

The objective of the present study was to develop a novel molybdenum disulfide/iron oxide/gold nanorods (MoS2/Fe3O4/GNR) nanocomposite (MFG) with different concentrations of AgNO3 solution (MFG1, MFG2, and MFG3) for topical doxorubicin (DOX) drug delivery. Then, these nanocomposites were synthesized and characterized by Fourier transform infrared (FTIR), Transmission electron microscopy (TEM), Dynamic light scattering (DLS), and Ultraviolet-visible (UV-Vis) spectroscopies to confirm their structural and optical properties. Cytotoxicity of samples on Hela cell was determined using MTT assay. Results indicated that nanocomposites possess little cytotoxicity without NIR laser irradiation. Also, the relative viabilities of Hela cells decreased when the concentration of AgNO3 solution increased in this nanocomposite. Using NIR irradiation, the relative viabilities of Hela cells decreased when the concentration of samples increased. Acridine orange/propidium iodide (PI) staining, flow cytometry were recruited to evaluate the effect of these nanocomposites on apoptosis of Hela cells. Finally, results revealed when DOX loading increased in nanocomposite, then cell viability was decreased in it. Therefore, these properties make MFG3 nanocomposite a good candidate for photothermal therapy and drug loading.


Sujet(s)
Survie cellulaire , Disulfures , Doxorubicine , Or , Molybdène , Nanocomposites , Humains , Molybdène/composition chimique , Molybdène/pharmacologie , Cellules HeLa , Nanocomposites/composition chimique , Disulfures/composition chimique , Or/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Nanotubes/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Thérapie photothermique/méthodes , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Spectroscopie infrarouge à transformée de Fourier , Photothérapie/méthodes , Composés du fer III/composition chimique
11.
Cancer Med ; 13(15): e70079, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39118454

RÉSUMÉ

BACKGROUND: Cancer remains a formidable global health challenge, currently affecting nearly 20 million individuals worldwide. Due to the absence of universally effective treatments, ongoing research explores diverse strategies to combat this disease. Recent efforts have concentrated on developing combined drug regimens and targeted therapeutic approaches. OBJECTIVE: This study aimed to investigate the anticancer efficacy of a conjugated drug system, consisting of doxorubicin and cisplatin (Dox-Cis), encapsulated within niosomes and modified with MUC-1 aptamers to enhance biocompatibility and target specific cancer cells. METHODS: The chemical structure of the Dox-Cis conjugate was characterized using Fourier Transform Infrared Spectroscopy (FTIR) and Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometry (LC-Q-TOF/MS). The zeta potential and morphological parameters of the niosomal vesicles were determined through Dynamic Light Scattering (DLS) and Transmission Electron Microscopy (TEM). In vitro assessments of cell viability and apoptosis were conducted on MUC-1 positive HeLa cells and MUC-1 negative U87 cells. RESULTS: The findings confirmed the successful conjugation of Dox and Cis within the niosomes. The Nio/Dox-Cis/MUC-1 formulation demonstrated enhanced efficacy compared to the individual drugs and their unencapsulated combination in both cell lines. Notably, the Nio/Dox-Cis/MUC-1 formulation exhibited greater effectiveness on HeLa cells (38.503 ± 1.407) than on U87 cells (46.653 ± 1.297). CONCLUSION: The study underscores the potential of the Dox-Cis conjugate as a promising strategy for cancer treatment, particularly through platforms that facilitate targeted drug delivery to cancer cells. This targeted approach could lead to more effective and personalized cancer therapies.


Sujet(s)
Aptamères nucléotidiques , Survie cellulaire , Cisplatine , Doxorubicine , Liposomes , Mucine-1 , Humains , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Mucine-1/métabolisme , Mucine-1/composition chimique , Liposomes/composition chimique , Cisplatine/pharmacologie , Cisplatine/composition chimique , Aptamères nucléotidiques/composition chimique , Aptamères nucléotidiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Cellules HeLa , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Préparation de médicament/méthodes
12.
Carbohydr Polym ; 342: 122356, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39048219

RÉSUMÉ

In this study, we report the synthesis and characterization of pH-responsive nanoconjugates for targeted drug delivery. Galactomannan extracted from D. regia seeds was oxidized to form aldehyde groups, achieving a percentage of oxidation of 25.6 %. The resulting oxidized galactomannan (GMOX) was then copolymerized with PINIPAm-NH2, yielding a copolymer. The copolymer exhibited signals from both GMOX and PNIPAm-NH2 in its NMR spectrum, confirming successful copolymerization. Critical association concentration (CAC) studies revealed the formation of nanostructures, with lower CAC values observed at higher temperatures. The copolymer and GMOX reacted with doxorubicin (DOX), resulting in nanoconjugates with controlled drug release profiles, especially under acidic conditions similar to tumor microenvironments. Cytotoxicity assays demonstrated significant efficacy of the nanoconjugates against melanoma cells with reduced toxicity towards healthy cells. These findings underscore the potential of the pH-responsive nanoconjugates as promising candidates for targeted cancer therapy, offering improved therapeutic efficacy and reduced systemic side effects.


Sujet(s)
Doxorubicine , Galactose , Mannanes , Nanoconjugués , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Mannanes/composition chimique , Mannanes/pharmacologie , Galactose/composition chimique , Galactose/analogues et dérivés , Humains , Nanoconjugués/composition chimique , Concentration en ions d'hydrogène , Libération de médicament , Lignée cellulaire tumorale , Vecteurs de médicaments/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/composition chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie
13.
Int J Nanomedicine ; 19: 7367-7381, 2024.
Article de Anglais | MEDLINE | ID: mdl-39050872

RÉSUMÉ

Purpose: Chemotherapy remains the primary therapeutic approach for advanced Hepatocellular Carcinoma (HCC). The therapeutic effect of chemotherapy is limited and the toxic side effects are serious. The aim of this study is to develop a nanobubble that is ultrasonically responsive to reduce the toxic side effects of direct chemotherapy. Methods: We developed curcumin/doxorubicin-cis-aconitic anhydride-polyethylene glycol nanobubble (C/DCNB) surface modified with acid-sensitive polyethylene glycol (PEG). And it is loaded with curcumin (CUR) and doxorubicin (DOX), as liposomes at the nanoscale for diagnosis and therapy of tumors. Results: In this study, the acid-sensitive PEG on the surface layer of nanobubbles serves to stabilize them in the blood circulatory system and in normal tissues, while peeling off in the acidic tumor microenvironment (pH 6.8). C/DCNB can identify tumor sites through contrast-enhanced ultrasound (CEUS). And ultrasound-mediated nanobubbles promote permeability of the tumor vascular, thus improving the enhanced permeability and retention (EPR) effects in the tumor, leading to the accumulation of nanobubbles in the tumor. After endocytosis of nanobubbles, drugs are released and curcumin generates reactive oxygen species (ROS) under ultrasound conditions. CUR can enhance the sensitivity of tumor cells to DOX by inhibiting the expression of P-glycoprotein. In vitro and vivo experiments demonstrate that C/DCNB can facilitate contrast-enhanced ultrasound imaging while simultaneously delivering drugs, enabling both imaging and treatment. Conclusion: The combination of C/DCNB and ultrasound provides an effective strategy for improving the efficiency of HCC therapy and imaging.


Sujet(s)
Carcinome hépatocellulaire , Curcumine , Doxorubicine , Tumeurs du foie , Polyéthylène glycols , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/imagerie diagnostique , Carcinome hépatocellulaire/thérapie , Carcinome hépatocellulaire/anatomopathologie , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/pharmacocinétique , Doxorubicine/administration et posologie , Curcumine/composition chimique , Curcumine/pharmacologie , Curcumine/pharmacocinétique , Curcumine/administration et posologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/imagerie diagnostique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/thérapie , Animaux , Humains , Polyéthylène glycols/composition chimique , Lignée cellulaire tumorale , Liposomes/composition chimique , Souris , Espèces réactives de l'oxygène/métabolisme , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/pharmacocinétique , Souris de lignée BALB C , Cellules HepG2 , Souris nude , Nanoparticules/composition chimique , Ultrasonothérapie/méthodes , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Zhongguo Zhong Yao Za Zhi ; 49(13): 3515-3525, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39041123

RÉSUMÉ

Regulating the process of epithelial-mesenchymal transition(EMT) is an essential strategy to inhibit tumor growth and metastasis. This study is based on the EMT process of retinoblastoma and constructs quercetin(QUE) and doxorubicin(DOX) co-loaded liposome(QD Lipo) to investigate the therapeutic effect and mechanisms of combined QUE and DOX treatment on retinoblastoma. Single-factor experiments were conducted to optimize the prescription process of QD Lipo. Eventually, spherical particles with a diameter of(108.87±1.93) nm, a PDI of 0.13±0.02, and a Zeta potential of(-34.83±1.92) mV were obtained. The encapsulation rates of QUE and DOX were 96.20%±4.40% and 91.17%±4.41%, respectively. Y79 human retinoblastoma cells were used as an in vitro cellular model, and confocal microscopy demonstrated that QD Lipo could enhance Y79 uptake efficiency. The CCK-8 assay confirmed that the optimal combination therapy effect of QUE and DOX occurred at a mass ratio of 1∶1 to 1∶2. Flow cytometry showed that QD Lipo enhanced the induction of apoptosis in Y79 cells. Western blot analysis revealed that QD Lipo significantly reduced the expression of EMT pathway-related proteins vimentin and α-SMA. Fluorescence assays detected a significant decrease in ROS levels in Y79 cells after treatment with QD. These results indicated that liposomal co-delivery of QUE and DOX can enhance drug delivery efficiency to retinoblastoma cells, inhibit the EMT process in retinoblastoma by downregulating ROS levels, and enhance the cytotoxicity of DOX against retinoblastoma.


Sujet(s)
Doxorubicine , Transition épithélio-mésenchymateuse , Liposomes , Quercétine , Rétinoblastome , Quercétine/administration et posologie , Quercétine/pharmacologie , Quercétine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/administration et posologie , Rétinoblastome/traitement médicamenteux , Humains , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Liposomes/composition chimique , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments , Espèces réactives de l'oxygène/métabolisme
15.
ACS Nano ; 18(28): 18211-18229, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38946122

RÉSUMÉ

Transarterial chemoembolization (TACE), the mainstay treatment of unresectable primary liver cancer that primarily employs nondegradable drug-loaded embolic agents to achieve synergistic vascular embolization and locoregional chemotherapy effects, suffers from an inferior drug burst behavior lacking long-term drug release controllability that severely limits the TACE efficacy. Here we developed gelatin-based drug-eluting microembolics grafted with nanosized poly(acrylic acid) serving as a biodegradable ion-exchange platform that leverages a counterion condensation effect to achieve high-efficiency electrostatic drug loading with electropositive drugs such as doxorubicin (i.e., drug loading capacity >34 mg/mL, encapsulation efficiency >98%, and loading time <10 min) and an enzymatic surface-erosion degradation pattern (∼2 months) to offer sustained locoregional pharmacokinetics with long-lasting deep-tumor retention capability for TACE treatment. The microembolics demonstrated facile microcatheter deliverability in a healthy porcine liver embolization model, superior tumor-killing capacity in a rabbit VX2 liver cancer embolization model, and stabilized extravascular drug penetration depth (>3 mm for 3 months) in a rabbit ear embolization model. Importantly, the microembolics finally exhibited vessel remodeling-induced permanent embolization with minimal inflammation responses after complete degradation. Such a biodegradable ion-exchange drug carrier provides an effective and versatile strategy for enhancing long-term therapeutic responses of various local chemotherapy treatments.


Sujet(s)
Chimioembolisation thérapeutique , Doxorubicine , Animaux , Chimioembolisation thérapeutique/méthodes , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/administration et posologie , Doxorubicine/pharmacocinétique , Lapins , Tumeurs du foie/thérapie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Suidae , Résines acryliques/composition chimique , Polyélectrolytes/composition chimique , Vecteurs de médicaments/composition chimique , Antibiotiques antinéoplasiques/administration et posologie , Antibiotiques antinéoplasiques/composition chimique , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/pharmacocinétique , Gélatine/composition chimique , Nanoparticules/composition chimique , Humains , Libération de médicament , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/administration et posologie
16.
ACS Nano ; 18(28): 18769-18784, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38950189

RÉSUMÉ

Liposomes are versatile drug delivery systems in clinical use for cancer and many other diseases. Unfortunately, PEGylated liposomal doxorubicin (sLip/DOX) exhibits serious dose-limiting cutaneous toxicities, which are closely related to the extravascular accumulation of sLip/DOX in the dermis. No clinical interventions have been proposed for cutaneous toxicities due to the elusive transport pathways. Herein, we showed that the reciprocal interaction between liposomes and neutrophils played pivotal roles in liposome extravasation into the dermis. Neutrophils captured liposomes via the complement receptor 3 (CD11b/CD18) recognizing the fragment of complement component C3 (iC3b) deposited on the liposomal surface. Uptake of liposomes also activated neutrophils to induce CD11b upregulation and enhanced the ability of neutrophils to migrate outside the capillaries. Furthermore, inhibition of complement activation either by CRIg-L-FH (a C3b/iC3b targeted complement inhibitor) or blocking the phosphate negative charge in mPEG-DSPE could significantly reduce liposome uptake by neutrophils and alleviate the cutaneous accumulation of liposomes. These results validated the liposome extravasation pathway mediated by neutrophils and provided potential solutions to the devastating cutaneous toxicities occurring during sLip/DOX treatment.


Sujet(s)
Doxorubicine , Liposomes , Granulocytes neutrophiles , Polyéthylène glycols , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/analogues et dérivés , Liposomes/composition chimique , Animaux , Polyéthylène glycols/composition chimique , Souris , Peau/métabolisme , Peau/effets des médicaments et des substances chimiques , Activation du complément/effets des médicaments et des substances chimiques , Humains
17.
ACS Nano ; 18(28): 18604-18621, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38952130

RÉSUMÉ

Chemo-immunotherapy has become a promising strategy for cancer treatment. However, the inability of the drugs to penetrate deeply into the tumor and form potent tumor vaccines in vivo severely restricts the antitumor effect of chemo-immunotherapy. In this work, an injectable sodium alginate platform is reported to promote penetration of the chemotherapeutic doxorubicin (DOX) and delivery of personalized tumor vaccines. The injectable multifunctional sodium alginate platform cross-links rapidly in the presence of physiological concentrations of Ca2+, forming a hydrogel that acts as a drug depot and releases loaded hyaluronidase (HAase), DOX, and micelles (IP-NPs) slowly and sustainedly. By degrading hyaluronic acid (HA) overexpressed in tumor tissue, HAase can make tumor tissue "loose" and favor other components to penetrate deeply. DOX induces potent immunogenic cell death (ICD) and produces tumor-associated antigens (TAAs), which could be effectively captured by polyethylenimine (PEI) coated IP-NPs micelles and form personalized tumor vaccines. The vaccines efficaciously facilitate the maturation of dendritic cells (DCs) and activation of T lymphocytes, thus producing long-term immune memory. Imiquimod (IMQ) loaded in the core could further activate the immune system and trigger a more robust antitumor immune effect. Hence, the research proposes a multifunctional drug delivery platform for the effective treatment of colorectal cancer.


Sujet(s)
Alginates , Doxorubicine , Hydrogels , Immunothérapie , Nanoparticules , Alginates/composition chimique , Hydrogels/composition chimique , Animaux , Nanoparticules/composition chimique , Souris , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Humains , Vaccins anticancéreux/composition chimique , Vaccins anticancéreux/administration et posologie , Hyaluronoglucosaminidase/métabolisme , Micelles , Lignée cellulaire tumorale
18.
Int J Nanomedicine ; 19: 6829-6843, 2024.
Article de Anglais | MEDLINE | ID: mdl-39005958

RÉSUMÉ

Background: With the rapid development of nanotechnology, constructing a multifunctional nanoplatform that can deliver various therapeutic agents in different departments and respond to endogenous/exogenous stimuli for multimodal synergistic cancer therapy remains a major challenge to address the inherent limitations of chemotherapy. Methods: Herein, we synthesized hollow mesoporous Prussian Blue@zinc phosphate nanoparticles to load glucose oxidase (GOx) and DOX (designed as HMPB-GOx@ZnP-DOX NPs) in the non-identical pore structures of their HMPB core and ZnP shell, respectively, for photothermally augmented chemo-starvation therapy. Results: The ZnP shell coated on the HMPB core, in addition to providing space to load DOX for chemotherapy, could also serve as a gatekeeper to protect GOx from premature leakage and inactivation before reaching the tumor site because of its degradation characteristics under mild acidic conditions. Moreover, the loaded GOx can initiate starvation therapy by catalyzing glucose oxidation while causing an upgradation of acidity and H2O2 levels, which can also be used as forceful endogenous stimuli to trigger smart delivery systems for therapeutic applications. The decrease in pH can improve the pH-sensitivity of drug release, and O2 can be supplied by decomposing H2O2 through the catalase-like activity of HMPBs, which is beneficial for relieving the adverse conditions of anti-tumor activity. In addition, the inner HMPB also acts as a photothermal agent for photothermal therapy and the generated hyperthermia upon laser irradiation can serve as an external stimulus to further promote drug release and enzymatic activities of GOx, thereby enabling a synergetic photothermally enhanced chemo-starvation therapy effect. Importantly, these results indicate that HMPB-GOx@ZnP-DOX NPs can effectively inhibit tumor growth by 80.31% and exhibit no obvious systemic toxicity in mice. Conclusion: HMPB-GOx@ZnP-DOX NPs can be employed as potential theranostic agents that incorporate multiple therapeutic modes to efficiently inhibit tumors.


Sujet(s)
Doxorubicine , Hexacyanoferrates II , Glucose oxidase , Phosphates , Thérapie photothermique , Composés du zinc , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Doxorubicine/administration et posologie , Doxorubicine/pharmacocinétique , Animaux , Glucose oxidase/composition chimique , Glucose oxidase/pharmacologie , Souris , Hexacyanoferrates II/composition chimique , Hexacyanoferrates II/pharmacologie , Humains , Composés du zinc/composition chimique , Phosphates/composition chimique , Phosphates/pharmacologie , Thérapie photothermique/méthodes , Porosité , Nanoparticules/composition chimique , Lignée cellulaire tumorale , Libération de médicament , Souris de lignée BALB C , Systèmes de délivrance de médicaments/méthodes , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Vecteurs de médicaments/composition chimique
19.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39000306

RÉSUMÉ

The unique structure of G4.0 PAMAM dendrimers allows a drug to be enclosed in internal spaces or immobilized on the surface. In the conducted research, the conditions for the formation of the active G4.0 PAMAM complex with doxorubicin hydrochloride (DOX) were optimized. The physicochemical properties of the system were monitored using dynamic light scattering (DLS), circular dichroism (CD), and fluorescence spectroscopy. The Quartz Crystal Microbalance with Dissipation Monitoring (QCM-D) method was chosen to determine the preferential conditions for the complex formation. The highest binding efficiency of the drug to the cationic dendrimer was observed under basic conditions when the DOX molecule was deprotonated. The decrease in the zeta potential of the complex confirms that DOX immobilizes through electrostatic interaction with the carrier's surface amine groups. The binding constants were determined from the fluorescence quenching of the DOX molecule in the presence of G4.0 PAMAM. The two-fold way of binding doxorubicin in the structure of dendrimers was visible in the Isothermal calorimetry (ITC) isotherm. Fluorescence spectra and release curves identified the reversible binding of DOX to the nanocarrier. Among the selected cancer cells, the most promising anticancer activity of the G4.0-DOX complex was observed in A375 malignant melanoma cells. Moreover, the preferred intracellular location of the complexes concerning the free drug was found, which is essential from a therapeutic point of view.


Sujet(s)
Dendrimères , Doxorubicine , Dendrimères/composition chimique , Doxorubicine/composition chimique , Doxorubicine/pharmacologie , Humains , Lignée cellulaire tumorale , Vecteurs de médicaments/composition chimique , Systèmes de délivrance de médicaments/méthodes , Libération de médicament , Survie cellulaire/effets des médicaments et des substances chimiques
20.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39000317

RÉSUMÉ

Chemotherapy is among the main classical approaches to the treatment of oncologic diseases. Its efficiency has been comprehensively proven by clinical examinations; however, the low selectivity of chemotherapeutic agents limits the possibilities of this method, making it necessary to search for new approaches to the therapy of oncologic diseases. Photodynamic therapy is the least invasive method and a very efficient alternative for the treatment of malignant tumors; however, its efficiency depends on the depth of light penetration into the tissue and on the degree of oxygenation of the treatment zone. In this work, a hitherto unknown conjugate of a natural bacteriochlorin derivative and doxorubicin was obtained. In vitro and in vivo studies showed a more pronounced activity of the conjugate against MCF-7 and 4T1 cells and its higher tumorotropicity in animal tumor-bearing animals compared to free anthracycline antibiotic. The suggested conjugate implements the advantages of photodynamic therapy and chemotherapy and has great potential in cancer treatment.


Sujet(s)
Doxorubicine , Photothérapie dynamique , Porphyrines , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Doxorubicine/usage thérapeutique , Photothérapie dynamique/méthodes , Animaux , Humains , Souris , Porphyrines/composition chimique , Porphyrines/pharmacologie , Porphyrines/usage thérapeutique , Femelle , Cellules MCF-7 , Photosensibilisants/composition chimique , Photosensibilisants/usage thérapeutique , Photosensibilisants/pharmacologie , Lignée cellulaire tumorale , Souris de lignée BALB C , Antibiotiques antinéoplasiques/composition chimique , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE