Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 18.370
1.
Int J Nanomedicine ; 19: 4893-4906, 2024.
Article En | MEDLINE | ID: mdl-38828202

Introduction: The tumor microenvironment (TME) has attracted considerable attention as a potential therapeutic target for cancer. High levels of reactive oxygen species (ROS) in the TME may act as a stimulus for drug release. In this study, we have developed ROS-responsive hyaluronic acid-bilirubin nanoparticles (HABN) loaded with doxorubicin (DOX@HABN) for the specific delivery and release of DOX in tumor tissue. The hyaluronic acid shell of the nanoparticles acts as an active targeting ligand that can specifically bind to CD44-overexpressing tumors. The bilirubin core has intrinsic anti-cancer activity and ROS-responsive solubility change properties. Methods & Results: DOX@HABN showed the HA shell-mediated targeting ability, ROS-responsive disruption leading to ROS-mediated drug release, and synergistic anti-cancer activity against ROS-overproducing CD44-overexpressing HeLa cells. Additionally, intravenously administered HABN-Cy5.5 showed remarkable tumor-targeting ability in HeLa tumor-bearing mice with limited distribution in major organs. Finally, intravenous injection of DOX@HABN into HeLa tumor-bearing mice showed synergistic anti-tumor efficacy without noticeable side effects. Conclusion: These findings suggest that DOX@HABN has significant potential as a cancer-targeting and TME ROS-responsive nanomedicine for targeted cancer treatment.


Bilirubin , Doxorubicin , Hyaluronan Receptors , Hyaluronic Acid , Nanomedicine , Nanoparticles , Reactive Oxygen Species , Tumor Microenvironment , Hyaluronic Acid/chemistry , Tumor Microenvironment/drug effects , Animals , Reactive Oxygen Species/metabolism , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/administration & dosage , Nanoparticles/chemistry , Mice , HeLa Cells , Hyaluronan Receptors/metabolism , Bilirubin/chemistry , Bilirubin/pharmacology , Bilirubin/pharmacokinetics , Drug Liberation , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Neoplasms/metabolism
2.
Oncoimmunology ; 13(1): 2362454, 2024.
Article En | MEDLINE | ID: mdl-38846084

Rituximab (RTX) plus chemotherapy (R-CHOP) applied as a first-line therapy for lymphoma leads to a relapse in approximately 40% of the patients. Therefore, novel approaches to treat aggressive lymphomas are being intensively investigated. Several RTX-resistant (RR) cell lines have been established as surrogate models to study resistance to R-CHOP. Our study reveals that RR cells are characterized by a major downregulation of CD37, a molecule currently explored as a target for immunotherapy. Using CD20 knockout (KO) cell lines, we demonstrate that CD20 and CD37 form a complex, and hypothesize that the presence of CD20 stabilizes CD37 in the cell membrane. Consequently, we observe a diminished cytotoxicity of anti-CD37 monoclonal antibody (mAb) in complement-dependent cytotoxicity in both RR and CD20 KO cells that can be partially restored upon lysosome inhibition. On the other hand, the internalization rate of anti-CD37 mAb in CD20 KO cells is increased when compared to controls, suggesting unhampered efficacy of antibody drug conjugates (ADCs). Importantly, even a major downregulation in CD37 levels does not hamper the efficacy of CD37-directed chimeric antigen receptor (CAR) T cells. In summary, we present here a novel mechanism of CD37 regulation with further implications for the use of anti-CD37 immunotherapies.


Antigens, CD20 , Immunotherapy , Lymphoma, B-Cell , Rituximab , Tetraspanins , Humans , Antigens, CD20/immunology , Antigens, CD20/metabolism , Antigens, CD20/genetics , Rituximab/pharmacology , Rituximab/therapeutic use , Tetraspanins/genetics , Tetraspanins/metabolism , Cell Line, Tumor , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/drug therapy , Immunotherapy/methods , Antigens, Neoplasm/immunology , Antigens, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Vincristine/pharmacology , Vincristine/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Gene Expression Regulation, Neoplastic
3.
Cell Death Dis ; 15(6): 386, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38824143

Doxorubicin's antitumor effectiveness may be constrained with ineffective tumor penetration, systemic adverse effects, as well as drug resistance. The co-loading of immune checkpoint inhibitors and doxorubicin into liposomes can produce synergistic benefits and address problems, including quick drug clearance, toxicity, and low drug penetration efficiency. In our previous study, we modified a nanobody targeting CTLA-4 onto liposomes (LPS-Nb36) to be an extremely potent CTLA-4 signal blocker which improve the CD8+ T-cell activity against tumors under physiological conditions. In this study, we designed a drug delivery system (LPS-RGD-Nb36-DOX) based on LPS-Nb36 that realized the doxorubicin and anti-CTLA-4 Nb co-loaded and RGD modification, and was applied to antitumor therapy. We tested whether LPS-RGD-Nb36-DOX could targets the tumor by in vivo animal photography, and more importantly, promote cytotoxic T cells proliferation, pro-inflammatory cytokine production, and cytotoxicity. Our findings demonstrated that the combination of activated CD8+ T cells with doxorubicin/anti-CTLA-4 Nb co-loaded liposomes can effectively eradicate tumor cells both in vivo and in vitro. This combination therapy is anticipated to have synergistic antitumor effects. More importantly, it has the potential to reduce the dose of chemotherapeutic drugs and improve safety.


CTLA-4 Antigen , Doxorubicin , Drug Delivery Systems , Liposomes , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Animals , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/metabolism , Mice , Drug Delivery Systems/methods , Humans , Cell Line, Tumor , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Female , Mice, Inbred BALB C , Mice, Inbred C57BL
4.
Mol Biol Rep ; 51(1): 773, 2024 Jun 21.
Article En | MEDLINE | ID: mdl-38904710

Protein nanocages resemble natural biomimetic carriers and can be engineered to act as targeted delivery systems, making them an attractive option for various drug delivery and biomedical applications. Our research investigated the genetic link of a specific anti-HER2 peptide (LTVSPWY) to the exposed N-terminal region of the maize (Zea mays) ferritin 1 (ZmFer1) protein nanocage, employing either a 7-amino acid (for LTVS-ZmFer1) or 16-amino acid (for LTVS-L-ZmFer1) linker. We utilized a heat treatment method to load the chemotherapeutic drug doxorubicin into the protein nanocage. The construct with the longer linker (LTVS-L) produced a greater amount of soluble protein nanocage and was selected for further experiments. The average size, polydispersity index, and zeta potential of the engineered protein nanocage were 19.01 nm, 0.168, and - 2.13 mV, respectively. The LTVS-L-ZmFer1 protein nanocage exhibited excellent thermal stability, withstanding temperatures up to 100 °C with only partial denaturation. Furthermore, we observed that cellular uptake of the LTVS-L-ZmFer1 protein nanocages in HER2-positive breast cancer cells was significantly higher compared to ZmFer1 after labeling with FITC (fluorescein isothiocyanate) (P-value = 0.0001). In addition, we observed a significant decrease in the viability of SKBR3 cells when treated with DOX-loaded LTVS-L-ZmFer1 protein nanocages compared to cells treated with DOX-loaded ZmFer1 protein nanocages. Therefore, this new treatment strategy may prove to be an effective way to reduce both the side effects and toxicity associated with conventional cancer treatments in patients with HER2-positive breast cancer.


Doxorubicin , Drug Delivery Systems , Receptor, ErbB-2 , Humans , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Cell Line, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ferritins/chemistry , Ferritins/metabolism , Ferritins/genetics , Zea mays/genetics , Protein Engineering/methods , Female , Drug Carriers/chemistry , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism
5.
Sci Rep ; 14(1): 13352, 2024 06 10.
Article En | MEDLINE | ID: mdl-38858467

Liver cancer ranks as the fifth leading cause of cancer-related death globally. Direct intratumoral injections of anti-cancer therapeutics may improve therapeutic efficacy and mitigate adverse effects compared to intravenous injections. Some challenges of intratumoral injections are that the liquid drug formulation may not remain localized and have unpredictable volumetric distribution. Thus, drug delivery varies widely, highly-dependent upon technique. An X-ray imageable poloxamer 407 (POL)-based drug delivery gel was developed and characterized, enabling real-time feedback. Utilizing three needle devices, POL or a control iodinated contrast solution were injected into an ex vivo bovine liver. The 3D distribution was assessed with cone beam computed tomography (CBCT). The 3D distribution of POL gels demonstrated localized spherical morphologies regardless of the injection rate. In addition, the gel 3D conformal distribution could be intentionally altered, depending on the injection technique. When doxorubicin (DOX) was loaded into the POL and injected, DOX distribution on optical imaging matched iodine distribution on CBCT suggesting spatial alignment of DOX and iodine localization in tissue. The controllability and localized deposition of this formulation may ultimately reduce the dependence on operator technique, reduce systemic side effects, and facilitate reproducibility across treatments, through more predictable standardized delivery.


Cone-Beam Computed Tomography , Doxorubicin , Drug Delivery Systems , Hydrogels , Needles , Poloxamer , Hydrogels/chemistry , Animals , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Poloxamer/chemistry , Cattle , Cone-Beam Computed Tomography/methods , Liver/diagnostic imaging , Liver/metabolism
6.
Nanotechnology ; 35(36)2024 Jun 21.
Article En | MEDLINE | ID: mdl-38861968

Extracellular vesicles (EVs) have great potential in oncology drug delivery because of their unique biological origin. Apoptotic bodies (ABs), as a member of the EV family, offer distinct advantages in terms of size, availability and membrane properties, but have been neglected for a long time. Here, using ABs and Ti2N nanosheets, we propose a novel drug delivery system (Ti2N-DOX@ABs), which exhibit a homologous targeting ability for dual-strategy tumor therapy with intrinsic biological property. The experimental results demonstrate that such a drug delivery system possesses a drug loading capacity of 496.5% and a near-infrared photothermal conversion efficiency of 38.4%. In addition, the investigation of drug internalization process proved that Ti2N-DOX@ABs featured a supreme biocompatibility. Finally, the dual-strategy response based on photothermal and chemotherapeutic effects was studied under near-infrared laser radiation. This work explores the opportunity of apoptosome membranes in nanomedicine systems, which provides a technical reference for cancer-oriented precision medicine research.


Doxorubicin , Photothermal Therapy , Titanium , Humans , Photothermal Therapy/methods , Titanium/chemistry , Titanium/pharmacology , Doxorubicin/pharmacology , Doxorubicin/chemistry , Drug Delivery Systems/methods , Nanostructures/chemistry , Cell Line, Tumor , Extracellular Vesicles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Infrared Rays , Animals , Cell Survival/drug effects
7.
Mikrochim Acta ; 191(7): 404, 2024 06 18.
Article En | MEDLINE | ID: mdl-38888740

The unprecedented navigation ability in micro/nanoscale and tailored functionality tunes micro/nanomotors as new target drug delivery systems, open up new horizons for biomedical applications. Herein, we designed a light-driven rGO/Cu2 + 1O tubular nanomotor for active targeting of cancer cells as a drug delivery system. The propulsion performance is greatly enhanced in real cell media (5% glucose cells isotonic solution), attributing to the introduction of oxygen vacancy and reduced graphene oxide (rGO) layer for separating photo-induced electron-hole pairs. The motion speed and direction can be readily modulated. Meanwhile, doxorubicin (DOX) can be loaded quickly on the rGO layer because of π-π bonding effect. The Cu2 + 1O matrix in the tiny robots not only serves as a photocatalyst to generate a chemical concentration gradient as the driving force but also acts as a nanomedicine to kill cancer cells as well. The strong propulsion of light-driven rGO/Cu2 + 1O nanomotors coupled with tiny size endow them with active transmembrane transport, assisting DOX and Cu2 + 1O breaking through the barrier of the cell membrane. Compared with non-powered nanocarrier and free DOX, light-propelled rGO/Cu2 + 1O nanomotors exhibit greater transmembrane transport efficiency and significant therapeutic efficacy. This proof-of-concept nanomotor design presents an innovative approach against tumor, enlarging the list of biomedical applications of light-driven micro/nanomotors to the superficial tissue treatment.


Copper , Doxorubicin , Graphite , Light , Copper/chemistry , Humans , Doxorubicin/pharmacology , Doxorubicin/chemistry , Graphite/chemistry , Drug Delivery Systems , Drug Carriers/chemistry , Drug Carriers/radiation effects , Cell Survival/drug effects , Drug Liberation , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor
8.
Drug Des Devel Ther ; 18: 2021-2032, 2024.
Article En | MEDLINE | ID: mdl-38863768

In the era of single and combination maintenance therapies as well as platinum and Poly (ADP-ribose) polymerase inhibitors (PARPi) resistance, the choice of subsequent treatments following first-line platinum-based chemotherapy in recurrent ovarian cancer (ROC) patients has become increasingly complex. Within the ovarian cancer treatment algorithm, particularly in the emerging context of PARPi resistance, the role of trabectedin, in combination with pegylated liposomal doxorubicin (PLD) still preserves its significance. This paper offers valuable insights into the multifaceted role and mechanism of action of trabectedin in ROC. The main results of clinical trials and studies involving trabectedin/PLD, along with hints of Breast Cancer genes (BRCA)-mutated and BRCAness phenotype cases, are critically discussed. Moreover, this review provides and contextualizes potential scenarios of administering trabectedin in combination with PLD in ROC, according to established guidelines and beyond.


Ovarian Neoplasms , Trabectedin , Trabectedin/therapeutic use , Trabectedin/pharmacology , Trabectedin/administration & dosage , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Female , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Alkylating/administration & dosage , Tetrahydroisoquinolines/pharmacology , Tetrahydroisoquinolines/therapeutic use , Tetrahydroisoquinolines/administration & dosage , Dioxoles/pharmacology , Dioxoles/therapeutic use , Dioxoles/administration & dosage , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
9.
Sci Adv ; 10(24): eadn6157, 2024 Jun 14.
Article En | MEDLINE | ID: mdl-38865468

Lung metastasis poses a formidable challenge in the realm of cancer treatment, with conventional chemotherapy often falling short due to limited targeting and low accumulation in the lungs. Here, we show a microrobot approach using motile algae for localized delivery of drug-loaded nanoparticles to address lung metastasis challenges. The biohybrid microrobot [denoted "algae-NP(DOX)-robot"] combines green microalgae with red blood cell membrane-coated nanoparticles containing doxorubicin, a representative chemotherapeutic drug. Microalgae provide autonomous propulsion in the lungs, leveraging controlled drug release and enhanced drug dispersion to exert antimetastatic effects. Upon intratracheal administration, algae-NP(DOX)-robots efficiently transport their drug payload deep into the lungs while maintaining continuous motility. This strategy leads to rapid drug distribution, improved tissue accumulation, and prolonged retention compared to passive drug-loaded nanoparticles and free drug controls. In a melanoma lung metastasis model, algae-NP(DOX)-robots exhibit substantial improvement in therapeutic efficacy, reducing metastatic burden and extending survival compared to control groups.


Doxorubicin , Lung Neoplasms , Nanoparticles , Lung Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Animals , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Nanoparticles/chemistry , Mice , Cell Line, Tumor , Humans , Drug Delivery Systems , Microalgae , Robotics , Disease Progression , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry
10.
Int J Nanomedicine ; 19: 5781-5792, 2024.
Article En | MEDLINE | ID: mdl-38882546

Background: While nanoplatform-based cancer theranostics have been researched and investigated for many years, enhancing antitumor efficacy and reducing toxic side effects is still an essential problem. Methods: We exploited nanoparticle coordination between ferric (Fe2+) ions and telomerase-targeting hairpin DNA structures to encapsulate doxorubicin (DOX) and fabricated Fe2+-DNA@DOX nanoparticles (BDDF NPs). This work studied the NIR fluorescence imaging and pharmacokinetic studies targeting the ability and biodistribution of BDDF NPs. In vitro and vivo studies investigated the nano formula's toxicity, imaging, and synergistic therapeutic effects. Results: The enhanced permeability and retention (EPR) effect and tumor targeting resulted in prolonged blood circulation times and high tumor accumulation. Significantly, BDDF NPs could reduce DOX-mediated cardiac toxicity by improving the antioxidation ability of cardiomyocytes based on the different telomerase activities and iron dependency in normal and tumor cells. The synergistic treatment efficacy is enhanced through Fe2+-mediated ferroptosis and the ß-catenin/p53 pathway and improved the tumor inhibition rate. Conclusion: Harpin DNA-based nanoplatforms demonstrated prolonged blood circulation, tumor drug accumulation via telomerase-targeting, and synergistic therapy to improve antitumor drug efficacy. Our work sheds new light on nanomaterials for future synergistic chemotherapy.


Doxorubicin , Telomerase , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Animals , Humans , Telomerase/metabolism , Cell Line, Tumor , Mice , DNA/chemistry , DNA/pharmacokinetics , DNA/administration & dosage , Tissue Distribution , Nanoparticles/chemistry , Neoplasms/drug therapy , Ferroptosis/drug effects , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/administration & dosage , Mice, Inbred BALB C , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics
11.
Ren Fail ; 46(2): 2365408, 2024 Dec.
Article En | MEDLINE | ID: mdl-38874119

Podocyte loss in glomeruli is a fundamental event in the pathogenesis of chronic kidney diseases. Currently, mitotic catastrophe (MC) has emerged as the main cause of podocyte loss. However, the regulation of MC in podocytes has yet to be elucidated. The current work aimed to study the role and mechanism of p53 in regulating the MC of podocytes using adriamycin (ADR)-induced nephropathy. In vitro podocyte stimulation with ADR triggered the occurrence of MC, which was accompanied by hyperactivation of p53 and cyclin-dependent kinase (CDK1)/cyclin B1. The inhibition of p53 reversed ADR-evoked MC in podocytes and protected against podocyte injury and loss. Further investigation showed that p53 mediated the activation of CDK1/cyclin B1 by regulating the expression of Wee1. Restraining Wee1 abolished the regulatory effect of p53 inhibition on CDK1/cyclin B1 and rebooted MC in ADR-stimulated podocytes via p53 inhibition. In a mouse model of ADR nephropathy, the inhibition of p53 ameliorated proteinuria and podocyte injury. Moreover, the inhibition of p53 blocked the progression of MC in podocytes in ADR nephropathy mice through the regulation of the Wee1/CDK1/cyclin B1 axis. Our findings confirm that p53 contributes to MC in podocytes through regulation of the Wee1/CDK1/Cyclin B1 axis, which may represent a novel mechanism underlying podocyte injury and loss during the progression of chronic kidney disorder.


CDC2 Protein Kinase , Cell Cycle Proteins , Cyclin B1 , Doxorubicin , Mitosis , Podocytes , Protein-Tyrosine Kinases , Tumor Suppressor Protein p53 , Podocytes/metabolism , Podocytes/pathology , Animals , CDC2 Protein Kinase/metabolism , Tumor Suppressor Protein p53/metabolism , Mice , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Doxorubicin/pharmacology , Cyclin B1/metabolism , Cell Cycle Proteins/metabolism , Disease Models, Animal , Humans , Male
12.
Biomed Pharmacother ; 176: 116895, 2024 Jul.
Article En | MEDLINE | ID: mdl-38876055

BACKGROUND: Soft tissue sarcomas (STS) are rare diseases typically arising from connective tissues in children and adults. However, chemotherapies involved in the treatment of STS may cause toxic side effects and multi-drug chemoresistance, making the treatment even more challenging. Histone deacetylase inhibitors (HDACi) are epigenetic agents which have shown anti-tumor effects as single agent as well as combination use with other drugs. Our project intends to prove the same effects in STS. METHODS: Panobinostat (LBH589) plus doxorubicin was selected for investigations based on our previous research. Tumor xenografts were tried in an epithelioid sarcoma model to validate good synergy effects in vivo and a leiomyosarcoma model was used as a negative comparison group. Gene profile changes were studied afterwards. The possible pathway changes caused by HDACi were explored and validated by several assays. RESULTS: Synergy effect of LBH589 plus doxorubicin was successfully validated in STS cell lines and an epithelioid sarcoma mice model. We tried to reduce the dose of doxorubicin to a lower level and found the drug combination can still inhibit tumor size in mice. Furthermore, gene profile changes caused by LBH589 was studied by RNA-Sequencing analysis. Results showed LBH589 can exert effects on a group of target genes which can regulate potential biological functions especially in the cell cycle pathway.


Doxorubicin , Drug Synergism , Histone Deacetylase Inhibitors , Panobinostat , Sarcoma , Xenograft Model Antitumor Assays , Panobinostat/pharmacology , Doxorubicin/pharmacology , Animals , Sarcoma/drug therapy , Sarcoma/pathology , Humans , Cell Line, Tumor , Histone Deacetylase Inhibitors/pharmacology , Mice , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Mice, Nude , Gene Expression Regulation, Neoplastic/drug effects
13.
Cell Commun Signal ; 22(1): 325, 2024 Jun 13.
Article En | MEDLINE | ID: mdl-38872211

BACKGROUND: Multidrug resistance (MDR) limits successful cancer chemotherapy. P-glycoprotein (P-gp), BCRP and MRP1 are the key triggers of MDR. Unfortunately, no MDR modulator was approved by FDA to date. Here, we will investigate the effect of BI-2865, a pan-KRAS inhibitor, on reversing MDR induced by P-gp, BCRP and MRP1 in vitro and in vivo, and its reversal mechanisms will be explored. METHODS: The cytotoxicity of BI-2865 and its MDR removal effect in vitro were tested by MTT assays, and the corresponding reversal function in vivo was assessed through the P-gp mediated KBv200 xenografts in mice. BI-2865 induced alterations of drug discharge and reservation in cells were estimated by experiments of Flow cytometry with fluorescent doxorubicin, and the chemo-drug accumulation in xenografts' tumor were analyzed through LC-MS. Mechanisms of BI-2865 inhibiting P-gp substrate's efflux were analyzed through the vanadate-sensitive ATPase assay, [125I]-IAAP-photolabeling assay and computer molecular docking. The effects of BI-2865 on P-gp expression and KRAS-downstream signaling were detected via Western blotting, Flow cytometry and/or qRT-PCR. Subcellular localization of P-gp was visualized by Immunofluorescence. RESULTS: We found BI-2865 notably fortified response of P-gp-driven MDR cancer cells to the administration of chemo-drugs including paclitaxel, vincristine and doxorubicin, while such an effect was not observed in their parental sensitive cells and BCRP or MRP1-driven MDR cells. Importantly, the mice vivo combination study has verified that BI-2865 effectively improved the anti-tumor action of paclitaxel without toxic injury. In mechanism, BI-2865 prompted doxorubicin accumulating in carcinoma cells by directly blocking the efflux function of P-gp, which more specifically, was achieved by BI-2865 competitively binding to the drug-binding sites of P-gp. What's more, at the effective MDR reversal concentrations, BI-2865 neither varied the expression and location of P-gp nor reduced its downstream AKT or ERK1/2 signaling activity. CONCLUSIONS: This study uncovered a new application of BI-2865 as a MDR modulator, which might be used to effectively, safely and specifically improve chemotherapeutic efficacy in the clinical P-gp mediated MDR refractory cancers.


Drug Resistance, Multiple , Drug Resistance, Neoplasm , Humans , Animals , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Multiple/drug effects , Mice , Cell Line, Tumor , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Xenograft Model Antitumor Assays , Mice, Nude , Doxorubicin/pharmacology , Mice, Inbred BALB C , Female
14.
ACS Appl Bio Mater ; 7(6): 3731-3745, 2024 Jun 17.
Article En | MEDLINE | ID: mdl-38842103

Photosensitizing agents have received increased attention from the medical community, owing to their higher photothermal efficiency, induction of hyperthermia, and sustained delivery of bioactive molecules to their targets. Micro/nanorobots can be used as ideal photosensitizing agents by utilizing various physical stimuli for the targeted killing of pathogens (e.g., bacteria) and cancer cells. Herein, we report sunflower-pollen-inspired spiky zinc oxide (s-ZnO)-based nanorobots that effectively kill bacteria and cancer cells under near-infrared (NIR) light irradiation. The as-fabricated s-ZnO was modified with a catechol-containing photothermal agent, polydopamine (PDA), to improve its NIR-responsive properties, followed by the addition of antimicrobial (e.g., tetracycline/TCN) and anticancer (e.g., doxorubicin/DOX) drugs. The fabricated s-ZnO/PDA@Drug nanobots exhibited unique locomotory behavior with an average speed ranging from 13 to 14 µm/s under 2.0 W/cm2 NIR light irradiation. Moreover, the s-ZnO/PDA@TCN nanobots exhibited superior antibacterial activity against E. coli and S. epidermidis under NIR irradiation. The s-ZnO/PDA@DOX nanobots also displayed sufficient reactive oxygen species (ROS) amplification in B16F10 melanoma cells and induced apoptosis under NIR light, indicating their therapeutic efficacy. We hope the sunflower pollen-inspired s-ZnO nanorobots have tremendous potential in biomedical engineering from the phototherapy perspective, with the hope to reduce pathogen infections.


Anti-Bacterial Agents , Antineoplastic Agents , Biocompatible Materials , Drug Screening Assays, Antitumor , Helianthus , Particle Size , Photosensitizing Agents , Zinc Oxide , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Helianthus/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Zinc Oxide/chemistry , Zinc Oxide/pharmacology , Materials Testing , Microbial Sensitivity Tests , Pollen/chemistry , Escherichia coli/drug effects , Staphylococcus epidermidis/drug effects , Cell Survival/drug effects , Cell Line, Tumor , Indoles/chemistry , Indoles/pharmacology , Animals , Mice , Doxorubicin/pharmacology , Doxorubicin/chemistry , Infrared Rays
15.
Mol Biol Rep ; 51(1): 721, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38829450

BACKGROUND: Cancer and multidrug resistance are regarded as concerns related to poor health outcomes. It was found that the monolayer of 2D cancer cell cultures lacks many important features compared to Multicellular Tumor Spheroids (MCTS) or 3D cell cultures which instead have the ability to mimic more closely the in vivo tumor microenvironment. This study aimed to produce 3D cell cultures from different cancer cell lines and to examine the cytotoxic activity of anticancer medications on both 2D and 3D systems, as well as to detect alterations in the expression of certain genes levels. METHOD: 3D cell culture was produced using 3D microtissue molds. The cytotoxic activities of colchicine, cisplatin, doxorubicin, and paclitaxel were tested on 2D and 3D cell culture systems obtained from different cell lines (A549, H1299, MCF-7, and DU-145). IC50 values were determined by MTT assay. In addition, gene expression levels of PIK3CA, AKT1, and PTEN were evaluated by qPCR. RESULTS: Similar cytotoxic activities were observed on both 3D and 2D cell cultures, however, higher concentrations of anticancer medications were needed for the 3D system. For instance, paclitaxel showed an IC50 of 6.234 µM and of 13.87 µM on 2D and 3D H1299 cell cultures, respectively. Gene expression of PIK3CA in H1299 cells also showed a higher fold change in 3D cell culture compared to 2D system upon treatment with doxorubicin. CONCLUSION: When compared to 2D cell cultures, the behavior of cells in the 3D system showed to be more resistant to anticancer treatments. Due to their shape, growth pattern, hypoxic core features, interaction between cells, biomarkers synthesis, and resistance to treatment penetration, the MCTS have the advantage of better simulating the in vivo tumor conditions. As a result, it is reasonable to conclude that 3D cell cultures may be a more promising model than the traditional 2D system, offering a better understanding of the in vivo molecular changes in response to different potential treatments and multidrug resistance development.


Antineoplastic Agents , Cell Culture Techniques , Spheroids, Cellular , Humans , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Spheroids, Cellular/drug effects , Cell Culture Techniques/methods , Doxorubicin/pharmacology , Paclitaxel/pharmacology , Cisplatin/pharmacology , Tumor Microenvironment/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Cell Culture Techniques, Three Dimensional/methods , MCF-7 Cells , Gene Expression Regulation, Neoplastic/drug effects , Cell Survival/drug effects
16.
Carbohydr Polym ; 340: 122270, 2024 Sep 15.
Article En | MEDLINE | ID: mdl-38858000

Targeted and stimuli-responsive drug delivery enhances therapeutic efficacy and minimizes undesirable side effects of cancer treatment. Although cellulose nanocrystals (CNCs) are used as drug carriers because of their robustness, spindle shape, biocompatibility, renewability, and nontoxicity, the lack of programmability and functionality of CNCs-based platforms hampers their application. Thus, high adaptability and the capacity to form dynamic 3D nanostructures of DNA may be advantageous, as they can provide functionalities such as target-specific and stimuli-responsive drug release. Using DNA nanotechnology, the functional polymeric form of DNA nanostructures can be replicated using rolling circle amplification (RCA), and the biologically and physiologically stable DNA nanostructures may overcome the challenges of CNCs. In this study, multifunctional polymeric DNAs produced with RCA were strongly complexed with surface-modified CNCs via electrostatic interactions to form polymeric DNA-decorated CNCs (pDCs). Particle size, polydispersity, zeta potential, and biostability of the nanocomplexes were analyzed. As a proof of concept, the dynamic structural functionalities of DNA nanostructures were verified by observing cancer-targeted intracellular delivery and pH-responsive drug release. pDCs showed anticancer properties without side effects in vitro, owing to their aptamer and i-motif functionalities. In conclusion, pDCs exhibited multifunctional anticancer activities, demonstrating their potential as a promising hybrid nanocomplex platform for targeted cancer therapy.


Cellulose , DNA , Drug Carriers , Drug Liberation , Nanoparticles , Nanostructures , Cellulose/chemistry , Humans , Nanoparticles/chemistry , DNA/chemistry , Nanostructures/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Polymers/chemistry , Hydrogen-Ion Concentration , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Cell Survival/drug effects
17.
Opt Express ; 32(10): 17239-17254, 2024 May 06.
Article En | MEDLINE | ID: mdl-38858913

Doxorubicin (DOX) is an important drug for cancer treatment, but its clinical application is limited due to its toxicity and side effects. Therefore, detecting the concentration of DOX during treatment is crucial for enhancing efficacy and reducing side effects. In this study, the authors developed a biophotonic fiber sensor based on localized surface plasmon resonance (LSPR) with the multimode fiber (MMF)-four core fiber (FCF)-seven core fiber (SCF)-MMF-based direct-taper and anti-taper structures for the specific detection of DOX. Compared to other detection methods, it has the advantages of high sensitivity, low cost, and strong anti-interference ability. In this experiment, multi-walled carbon nanotubes (MWCNTs), cerium-oxide nanorods (CeO2-NRs), and gold nanoparticles (AuNPs) were immobilized on the probe surface to enhance the sensor's biocompatibility. MWCNTs and CeO2-NRs provided more binding sites for the fixation of AuNPs. By immobilizing AuNPs on the surface, the LSPR was stimulated by the evanescent field to detect DOX. The sensor surface was functionalized with DOX aptamers for specific detection, enhancing its specificity. The experiments demonstrated that within a linear detection range of 0-10 µM, the sensitivity of the sensor is 0.77 nm/µM, and the limit of detection (LoD) is 0.42 µM. Additionally, the probe's repeatability, reproducibility, stability, and selectivity were evaluated, indicating that the probe has high potential for detecting DOX during cancer treatment.


Doxorubicin , Gold , Metal Nanoparticles , Surface Plasmon Resonance , Doxorubicin/pharmacology , Humans , Surface Plasmon Resonance/instrumentation , Gold/chemistry , Metal Nanoparticles/chemistry , Neoplasms/drug therapy , Nanotubes, Carbon/chemistry , Biosensing Techniques/instrumentation , Optical Fibers , Equipment Design , Antibiotics, Antineoplastic/analysis , Cerium/chemistry , Fiber Optic Technology/instrumentation
18.
Molecules ; 29(11)2024 May 24.
Article En | MEDLINE | ID: mdl-38893345

Among brain tumors, glioblastoma (GBM) is very challenging to treat as chemotherapeutic drugs can only penetrate the brain to a limited extent due to the blood-brain barrier (BBB). Nanoparticles can be an attractive solution for the treatment of GBM as they can transport drugs across the BBB into the tumor. In this study, normal and GBM organoids comprising six brain cell types were developed and applied to study the uptake, BBB penetration, distribution, and efficacy of fluorescent, ultrasmall gold nanoparticles (AuTio-Dox-AF647s) conjugated with doxorubicin (Dox) and AlexaFluor-647-cadaverine (AF647) by confocal laser scanning microscopy (CLSM), using a mixture of dissolved doxorubicin and fluorescent AF647 molecules as a control. It was shown that the nanoparticles could easily penetrate the BBB and were found in normal and GBM organoids, while the dissolved Dox and AF647 molecules alone were unable to penetrate the BBB. Flow cytometry showed a reduction in glioblastoma cells after treatment with AuTio-Dox nanoparticles, as well as a higher uptake of these nanoparticles by GBM cells in the GBM model compared to astrocytes in the normal cell organoids. In summary, our results show that ultrasmall gold nanoparticles can serve as suitable carriers for the delivery of drugs into organoids to study BBB function.


Blood-Brain Barrier , Doxorubicin , Glioblastoma , Gold , Metal Nanoparticles , Organoids , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Metal Nanoparticles/chemistry , Gold/chemistry , Humans , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Organoids/drug effects , Organoids/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor
19.
Molecules ; 29(11)2024 May 25.
Article En | MEDLINE | ID: mdl-38893380

Breast cancer is a major health concern and the leading cause of death among women worldwide. Standard treatment often involves surgery, radiotherapy, and chemotherapy, but these come with side effects and limitations. Researchers are exploring natural compounds like baicalin and baicalein, derived from the Scutellaria baicalensis plant, as potential complementary therapies. This study investigated the effects of baicalin and baicalein on the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel, commonly used chemotherapeutic drugs for breast cancer. The analysis included breast cancer cells (MCF-7) and human endothelial cells (HUVEC-ST), to assess potential effects on healthy tissues. We have found that baicalin and baicalein demonstrated cytotoxicity towards both cell lines, with more potent effects observed in baicalein. Both flavonoids, baicalin (167 µmol/L) and baicalein (95 µmol/L), synergistically enhanced the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel in breast cancer cells. In comparison, their effects on endothelial cells were mixed and depended on concentration and time. The results suggest that baicalin and baicalein might be promising complementary agents to improve the efficacy of doxorubicin and docetaxel anticancer activity. However, further research is needed to validate their safety and efficacy in clinical trials.


Apoptosis , Breast Neoplasms , Docetaxel , Doxorubicin , Flavanones , Flavonoids , Humans , Flavonoids/pharmacology , Flavanones/pharmacology , Docetaxel/pharmacology , Doxorubicin/pharmacology , MCF-7 Cells , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , DNA Damage/drug effects , Drug Synergism , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Human Umbilical Vein Endothelial Cells/drug effects
20.
Molecules ; 29(11)2024 May 30.
Article En | MEDLINE | ID: mdl-38893446

Doxorubicin (DOX) is a commonly used chemotherapeutic drug, from the anthracycline class, which is genotoxic to neoplastic cells via a DNA intercalation mechanism. It is effective and universal; however, it also causes numerous side effects. The most serious of them are cardiotoxicity and a decrease in the number of myeloid cells. For this reason, targeted DOX delivery systems are desirable, since they would allow lowering the drug dose and therefore limiting systemic side effects. Recently, synthetic dyes, in particular Congo red (CR), have been proposed as possible DOX carriers. CR is a planar molecule, built of a central biphenyl moiety and two substituted naphthalene rings, connected with diazo bonds. In water, it forms elongated ribbon-shaped supramolecular structures, which are able to selectively interact with immune complexes. In our previous studies, we have shown that CR aggregates can intercalate DOX molecules. In this way, they preclude DOX precipitation in water solutions and increase its uptake by MCF7 breast cancer cells. In the present work, we further explore the interactions between DOX, CR, and their aggregates (CR/DOX) with phospholipid membranes. In addition to neutral molecules, the protonated doxorubicin form, DXP, is also studied. Molecular dynamics simulations are employed to study the transfer of CR, DOX, DXP, and their aggregates through POPC bilayers. Interactions of CR, DOX, and CR/DOX with model monolayers are studied with Langmuir trough measurements. This study shows that CR may support the transfer of doxorubicin molecules into the bilayer. Both electrostatic and van der Waals interactions with lipids are important in this respect. The former promote the initial stages of the insertion process, the latter keep guest molecules inside the bilayer.


Congo Red , Doxorubicin , Molecular Dynamics Simulation , Phospholipids , Doxorubicin/chemistry , Doxorubicin/pharmacology , Phospholipids/chemistry , Congo Red/chemistry , Humans , Lipid Bilayers/chemistry , Drug Carriers/chemistry , MCF-7 Cells
...