Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38542073

ABSTRACT

Modulated electro-hyperthermia (mEHT) is an adjuvant cancer therapy that enables tumor-selective heating (+2.5 °C). In this study, we investigated whether mEHT accelerates the tumor-specific delivery of doxorubicin (DOX) from lyso-thermosensitive liposomal doxorubicin (LTLD) and improves its anticancer efficacy in mice bearing a triple-negative breast cancer cell line (4T1). The 4T1 cells were orthotopically injected into Balb/C mice, and mEHT was performed on days 9, 12, and 15 after the implantation. DOX, LTLD, or PEGylated liposomal DOX (PLD) were administered for comparison. The tumor size and DOX accumulation in the tumor were measured. The cleaved caspase-3 (cC3) and cell proliferation were evaluated by cC3 or Ki67 immunohistochemistry and Western blot. The LTLD+mEHT combination was more effective at inhibiting tumor growth than the free DOX and PLD, demonstrated by reductions in both the tumor volume and tumor weight. LTLD+mEHT resulted in the highest DOX accumulation in the tumor one hour after treatment. Tumor cell damage was associated with cC3 in the damaged area, and with a reduction in Ki67 in the living area. These changes were significantly the strongest in the LTLD+mEHT-treated tumors. The body weight loss was similar in all mice treated with any DOX formulation, suggesting no difference in toxicity. In conclusion, LTLD combined with mEHT represents a novel approach for DOX delivery into cancer tissue.


Subject(s)
Doxorubicin/analogs & derivatives , Hyperthermia, Induced , Neoplasms , Mice , Animals , Liposomes , Ki-67 Antigen , Hyperthermia, Induced/methods , Doxorubicin/pharmacology , Hyperthermia , Cell Line, Tumor , Polyethylene Glycols
2.
Pharm Dev Technol ; 28(6): 501-508, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37191345

ABSTRACT

Immunotherapy is a promising cancer treatment strategy. In contrast, programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitors are associated with low response rates and are only useful in a small group of cancer patients. A combination of treatments may be effective for overcoming this clinical issue. Preladenant is an adenosine (ADO) receptor inhibitor that can block the ADO pathway and improve the tumor microenvironment (TME), thereby enhancing the immunotherapeutic effect of PD-1 inhibitors. However, its poor water solubility and low targeting limit its clinical applications. We designed a PEG-modified thermosensitive-liposome (pTSL) loaded with ADO small molecule inhibitor preladenant (P-pTSL) to overcome these problems and enhance the effect of PD-1 inhibitor on breast cancer immunotherapy. The prepared P-pTSL was round and uniformly distributed with a particle size of (138.9 ± 1.22) nm, PDI: 0.134 ± 0.031, and zeta potential (-10.1 ± 1.63) mV; preladenant was released slowly at 37 °C but released fast at 42 °C from P-pTSL, which was 76.52 ± 0.44%. P-pTSL has good long-term and serum stability and excellent tumor-targeting ability in mice. Moreover, the combination with PD-1 inhibitor significantly enhanced the anti-tumor effect, and the improvement of related factors in serum and lymph was more obvious under the condition of 42 °C thermotherapy in vitro.


Subject(s)
Immune Checkpoint Inhibitors , Liposomes , Mice , Animals , Immunotherapy , Cell Line, Tumor , Immunity
3.
Colloids Surf B Biointerfaces ; 226: 113317, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37105064

ABSTRACT

Phototherapies, in the form of photodynamic therapy (PDT) and photothermal therapy (PTT), have great application prospects in the field of biomedical science due to high precision and non-invasiveness. Because of the limited therapeutic efficacy of single phototherapy, researchers start to focus on combined PTT-PDT. Here, we designed a composite nanomaterial for PTT-PDT. H-TiO2 mesoporous spheres were prepared by sol-gel method and hydrogenation treatment. After modification with polydopamine (PDA), they were combined with indocyanine green (ICG) and NPe6 photosensitizers and coated by thermosensitive liposomes to prepare H-TiO2 @PDA@ICG@NPe6 @Lipo nanocomposite component. The results indicated a substantial improvement of the component in the aspects of spectral response range, photothermal conversion efficiency and light absorption performance by modification and photosensitizers, in the absence of any toxicities on cells. Thermal induction and sequential irradiation with 808 nm and 664 nm lasers induced the aggregation of H-TiO2 @PDA@ICG@NPe6 @Lipo at the tumor site to generate hyperthermia and massive reactive oxygen species (ROS), resulting in decreased cell activity or even cell apoptosis and restrained growth of allograft tumors. These findings underscore the favorable effects of H-TiO2 @PDA@ICG@NPe6 @Lipo on the combined phototherapies and provide approaches for the development of nano-drugs in the context of liver cancer.


Subject(s)
Hyperthermia, Induced , Liver Neoplasms , Nanocomposites , Nanoparticles , Photochemotherapy , Humans , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Hyperthermia, Induced/methods , Phototherapy/methods , Indocyanine Green/pharmacology , Liver Neoplasms/drug therapy , Nanoparticles/therapeutic use , Cell Line, Tumor
4.
Pharmaceutics ; 15(3)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36986754

ABSTRACT

Doxorubicin (DOX) is a potent anti-cancer agent that has garnered great interest in research due to its high efficacy despite dose-limiting toxicities. Several strategies have been exploited to enhance the efficacy and safety profile of DOX. Liposomes are the most established approach. Despite the improvement in safety properties of liposomal encapsulated DOX (in Doxil and Myocet), the efficacy is not superior to conventional DOX. Functionalized (targeted) liposomes present a more effective system to deliver DOX to the tumor. Moreover, encapsulation of DOX in pH-sensitive liposomes (PSLs) or thermo-sensitive liposomes (TSLs) combined with local heating has improved DOX accumulation in the tumor. Lyso-thermosensitive liposomal DOX (LTLD), MM-302, and C225-immunoliposomal(IL)-DOX have reached clinical trials. Further functionalized PEGylated liposomal DOX (PLD), TSLs, and PSLs have been developed and evaluated in preclinical models. Most of these formulations improved the anti-tumor activity compared to the currently available liposomal DOX. However, the fast clearance, the optimization of ligand density, stability, and release rate need more investigations. Therefore, we reviewed the latest approaches applied to deliver DOX more efficiently to the tumor, preserving the benefits obtained from FDA-approved liposomes.

5.
J Control Release ; 354: 19-33, 2023 02.
Article in English | MEDLINE | ID: mdl-36503069

ABSTRACT

Triggered drug delivery strategies have been shown to enhance drug accumulation at target diseased sites in comparison to administration of free drug. In particular, many studies have demonstrated improved targetability of chemotherapeutics when delivered via thermosensitive liposomes. However, most studies continue to focus on encapsulating doxorubicin while many other drugs would benefit from this targeted and localized delivery approach. The proposed study explores the therapeutic potential of a thermosensitive liposome formulation of the commonly used chemotherapy drug vinorelbine in combination with mild hyperthermia (39-43 °C) in a murine model of rhabdomyosarcoma. Rhabdomyosarcoma, the most common soft tissue sarcoma in children, is largely treated using conventional chemotherapy which is associated with significant adverse long-term sequelae. In this study, mild hyperthermia was pursued as a non-invasive, non-toxic means to improve the efficacy and safety profiles of vinorelbine. Thorough assessment of the pharmacokinetics, biodistribution, efficacy and toxicity of vinorelbine administered in the thermosensitive liposome formulation was compared to administration in a traditional, non-thermosensitive liposome formulation. This study shows the potential of an advanced formulation technology in combination with mild hyperthermia as a means to target an untargeted therapeutic agent and result in a significant improvement in its therapeutic index.


Subject(s)
Hyperthermia, Induced , Rhabdomyosarcoma , Child , Mice , Humans , Animals , Liposomes , Vinorelbine , Tissue Distribution , Drug Delivery Systems , Doxorubicin , Cell Line, Tumor
6.
Angew Chem Int Ed Engl ; 61(42): e202205608, 2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36037071

ABSTRACT

Phase transitions of lipid bilayer membranes should affect passive transport of molecules. While this hypothesis has been used to design drug-releasing thermosensitive liposomes, the effect has yet to be quantified. Herein, we use time-resolved second harmonic light scattering to measure transport of a molecular cation across membranes of unilamellar liposomes composed of the total lipid extract of E. coli from 9 °C to 36 °C, in which two distinct phase transitions (gel to liquid-disordered phase) have been identified. While the transport rate slowly increases with temperature as a diffusion process, dramatic jumps are observed at 14.7 °C and 27.6 °C, the known phase transitions. The transport rate constant measured as (7.3±0.8)×10-3  s-1 in the liquid-disordered phase at 36 °C is 35-times faster than (2.1±0.2)×10-4  s-1 of the gel phase at 9 °C. For the mixed-phase between these two phases, the measured rates are consistent with a structure of gel domains among a liquid-disordered bulk.


Subject(s)
Lipid Bilayers , Liposomes , Cell Membrane , Diffusion , Escherichia coli , Lipid Bilayers/chemistry , Liposomes/chemistry , Temperature , Unilamellar Liposomes
7.
ACS Biomater Sci Eng ; 8(5): 1892-1906, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35404565

ABSTRACT

Organic near-infrared fluorescent dye mediated photothermal therapy (PTT) and photodynamic therapy (PDT) suffer from heat shock response, since, heat shock proteins (HSPs) are overexpressed and can repair the proteins damaged by PTT and PDT. Starvation therapy by glucose oxide (GOx) can inhibit the heat shock response by limiting the energy supply. However, the delivery of sufficient and active GOx remains a challenge. To solve this problem, we utilize liposomes as drug carriers and prepare GOx loaded liposome (GOx@Lipo) with a high drug loading content (12.0%) and high enzymatic activity. The successful delivery of GOx shows excellent inhibition of HSPs and enhances PTT and PDT. Additionally, we apply the same liposome formulation to load near-infrared dye 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbo cyanine iodide (DiR) and prepare DiR contained liposomes (DiR@Lipo) for PTT and PDT. The liposomal formulation substantially enhances the PTT and PDT properties of DiR as well as the cellular uptake and tumor accumulation. Finally, the combination therapy shows excellent tumor inhibition on 4T1 tumor-bearing mice. Interestingly, we also find that the starvation therapy can efficiently inhibit tumor metastasis, which is probably due to the immunogenic effect. Our work presents a biocompatible and effective carrier for the combination of starvation therapy and phototherapy, emphasizing the importance of auxiliary starvation therapy against tumor metastasis and offering important guidance for clinical PTT and PDT.


Subject(s)
Neoplasms , Photochemotherapy , Animals , Glucose Oxidase/therapeutic use , Liposomes/therapeutic use , Mice , Neoplasms/drug therapy , Photothermal Therapy
8.
J Liposome Res ; 32(3): 265-275, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34904521

ABSTRACT

In order to enhance the targeting efficiency and reduce the side effects and drug resistance, crizotinib (Cri) and F7 were co-loaded in a thermosensitive liposome (TSL) (F7-Cri-TSL), which showed enhanced permeability and retention in breast cancer model, as well as local controlled release by external hyperthermia. Cri is an inhibitor for cell proliferation and a promoter of apoptosis, by inhibiting the phosphorylation of intracellular ALK and c-Met, but its drug resistance limits its application. F7 is a novel drug candidate with significant resistance to cyclin-dependent kinase, but its use was restricted by its high toxicity. The F7-Cri-TSL was found with excellent particle size (about 108 nm), high entrapment efficiency (>95%), significant thermosensitive property, and good stability. Furthermore, F7-Cri-TSL/H had strongest cell lethality compared with other formulations. On the MCF-7 xenograft mice model, the F7-Cri-TSL also exhibited therapeutic synergism of Cri, F7 and hyperthermia. Meanwhile, it was shown that the TSL reduced the systemic toxicity of the chemotherapy drug. Therefore, the F7-Cri-TSL may serve as a promising system for temperature triggered breast cancer treatment.


Subject(s)
Breast Neoplasms , Liposomes , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Crizotinib/pharmacology , Crizotinib/therapeutic use , Doxorubicin , Female , Humans , Liposomes/therapeutic use , Mice , Temperature
9.
Biomed Mater ; 16(6)2021 10 25.
Article in English | MEDLINE | ID: mdl-34492645

ABSTRACT

Hydrogel crosslinking by external stimuli is a versatile strategy to control and modulate hydrogel properties. Besides photonic energy, thermal energy is one of the most accessible external stimuli and widely applicable for many biomedical applications. However, conventional thermal crosslinking systems require a relatively high temperature (over 100 °C) to initiate covalent bond formation. To our knowledge, there has not been a thermally tunable hydrogel crosslinking system suitable for biological applications. This work demonstrates a unique approach to utilize temperature sensitive liposomes to control and modulate hydrogel crosslinking over mild temperature range (below 50 °C). Temperature sensitive liposomes were used to control the release of chemical crosslinkers by moderate temperature changes. The thermally controlled crosslinker release resulted in tunable mechanical and transport properties of the hydrogel. No significant inflammable response observed in the histology results ensured the biocompatibility of the liposome-mediated crosslinkable hydrogel. This work opens new opportunities to implement thermal energy system for control and modulate hydrogel properties.


Subject(s)
Hydrogels , Liposomes , Hydrogels/chemistry , Temperature
10.
J Control Release ; 333: 1-15, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33741385

ABSTRACT

Various thermosensitive liposome (TSL) formulations have been described to date and it is currently unclear which are optimal for solid tumor treatment. Sufficient circulation half-life is important and most liposomes obtain this by polyethylene glycol (PEG) surface modification. 1,2-dipalmitoyl-sn-glycero-3-phosphodiglycerol (DPPG2) has been described as a promising alternative which increases TSL circulation half-life and facilitates rapid drug release under mild hyperthermia at 20-30 mol%. The present work describes an investigation of the DPPG2-TSL protein corona, blood cell interactions, complement activation in human plasma/blood and hypersensitivity reactions in rats. Furthermore, accelerated blood clearance (ABC) was investigated to obtain a complete assessment of DPPG2-TSL interactions with components of the blood and identify drivers for circulation half-life. A higher mol% DPPG2 increased Apolipoprotein E (ApoE) adsorption and decreased complement activation and granulocyte interaction in vitro. In contrast to PEG-TSL, DPPG2-TSL showed no ABC effect. In vivo hypersensitivity assessment by eicosanoid measurements, platelet and lymphocyte counting resembled the results of in vitro complement activation assays although here all DPPG2-TSL formulations induced hypersensitive responses upon i.v. administration. Prolonged circulation half-life of DPPG2-TSL may be ApoE-induced and the absent ABC effect demonstrates an advantage over PEG-TSL. Low complement activation in human plasma and blood for 20-30 mol% DPPG2-TSL presents a unique formulation attribute with the potential to strengthen clinical evaluation.


Subject(s)
Hyperthermia, Induced , Liposomes , Animals , Doxorubicin , Half-Life , Polyethylene Glycols , Rats
11.
J Control Release ; 328: 551-561, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32896612

ABSTRACT

Studies have demonstrated the advantages associated with heat-triggered drug delivery via thermosensitive liposomes for the treatment of localized cancer. Challenges that traditional liposomal systems face such as limited drug release and homogeneous distribution throughout the region of interest can potentially be overcome when triggering intravascular drug release. The most prominent example is a thermosensitive liposome formulation of doxorubicin known as ThermoDox®. Many other drugs may benefit from the same targeted and localized delivery approach using thermosensitive liposomes as it can result in a significant improvement in the therapeutic index. Vinorelbine is a semi-synthetic vinca alkaloid which has shown to be active in a broad range of cancers. Several liposome formulations encapsulating vinorelbine have been developed as a means to reduce systemic drug exposure. The present study takes a systematic approach in exploring formulation and drug loading parameters and their influence on performance characteristics of a rapidly releasing thermosensitive liposome formulation of vinorelbine. More broadly, this study shows that trends observed for non-thermosensitive liposome formulations of specific drugs (i.e. vinorelbine) can not be easily translated to their thermosensitive counterparts. The profound impact of the presence of albumin on stability and in vitro release is also highlighted. This is of significance given that a number of recent reports examine drug release in the absence of biologically relevant components. As a result, a strong recommendation emanating from this is a thorough challenge of the liposome formulation in vitro in order to gain a better understanding of its likely behaviour in vivo as well as potential for future clinical translation.


Subject(s)
Antineoplastic Agents , Liposomes , Antibiotics, Antineoplastic , Antineoplastic Agents/therapeutic use , Doxorubicin , Drug Delivery Systems , Vinorelbine
12.
Adv Drug Deliv Rev ; 163-164: 98-124, 2020.
Article in English | MEDLINE | ID: mdl-32681862

ABSTRACT

Hyperthermia has demonstrated clinical success in improving the efficacy of both chemo- and radio-therapy in solid tumors. Pre-clinical and clinical research studies have demonstrated that targeted hyperthermia can increase tumor blood flow and increase the perfused fraction of the tumor in a temperature and time dependent manner. Changes in tumor blood circulation can produce significant physiological changes including enhanced vascular permeability, increased oxygenation, decreased interstitial fluid pressure, and reestablishment of normal physiological pH conditions. These alterations in tumor physiology can positively impact both small molecule and nanomedicine chemotherapy accumulation and distribution within the tumor, as well as the fraction of the tumor susceptible to radiation therapy. Hyperthermia can trigger drug release from thermosensitive formulations and further improve the accumulation, distribution, and efficacy of chemotherapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Hyperthermia, Induced/methods , Hyperthermia/physiopathology , Neoplasms/therapy , Radiotherapy/methods , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Capillary Permeability/physiology , Combined Modality Therapy , Drug Delivery Systems/methods , Drug Liberation , Humans , Hydrogen-Ion Concentration , Neoplasms/blood supply , Neoplasms/physiopathology , Oxygen/blood , Time Factors , Tumor Microenvironment/physiology
13.
Drug Deliv ; 27(1): 836-847, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32508162

ABSTRACT

In order to enhance the targeting efficiency and reduce anti-tumor drug's side effects, topotecan (TPT) and F7 were co-loaded in thermosensitive liposomes (F7-TPT-TSL), which show enhanced permeability and retention in tumors, as well as local controlled release by heating in vitro. TPT is a water-soluble inhibitor of topoisomerase I that is converted to an inactive carboxylate structure under physiological conditions (pH 7.4). F7 is a novel drug significantly resistant to cyclin-dependent kinase but its use was restricted by its high toxicity. F7-TPT-TSL had excellent particle distribution (about 103 nm), high entrapment efficiency (>95%), obvious thermosensitive property, and good stability. Confocal microscopy demonstrated specific higher accumulation of TSL in tumor cells. MTT proved F7-TPT-TSL/H had strongest cell lethality compared with other formulations. Then therapeutic efficacy revealed synergism of TPT and F7 co-loaded in TSL, together with hyperthermia. Therefore, the F7-TPT-TSL may serve as a promising system for temperature triggered cancer treatment.


Subject(s)
Drug Delivery Systems/methods , Liposomes , Topotecan , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Biological Availability , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Hyperthermia , Liposomes/chemistry , Liposomes/pharmacokinetics , Mice , Nanostructures , Tissue Distribution/drug effects , Topoisomerase I Inhibitors/chemistry , Topoisomerase I Inhibitors/pharmacokinetics , Topotecan/chemistry , Topotecan/pharmacokinetics , Transition Temperature , Xenograft Model Antitumor Assays
14.
Biomaterials ; 245: 119840, 2020 07.
Article in English | MEDLINE | ID: mdl-32037007

ABSTRACT

Poor penetration and resultant low accumulation of nanomedicines in deep tumor tissues greatly reduce the chemotherapeutic efficiency. How to maximize the tumor accumulation is still a great challenge in the development of nanocarriers. Here, we developed a cyclic Arg-Gly-Asp-Phe-Lys peptide (cRGD) modified and near-infrared (NIR) light triggered disintegratable liposomal nanoplatform (PAM/Pt@IcLipo), where photosensitizer indocyanine green (ICG) was loaded in the out layer and polyamindoamine (PAMAM) dendrimers grafting cisplatin prodrug (PAM/Pt) were encapsulated inside. The cRGD ligands render the liposomes to target αvß3 integrin receptors overexpressed by vascular endothelial cells in tumor tissues. Long blood circulation can be achieved owing to the relative large size (~162 nm) of the liposomes. When irradiated by NIR light locally at tumor site, ICG heating detonated the thermosensitive liposomes to release the small sized PAM/Pt nanoparticles (~8.6 nm), which were capable of penetrating into the deep tumor tissue. The in vivo results also showed that the PAM/Pt@IcLipo could significantly improve the penetration of cisplatin drug in deep tumor tissues under NIR light irradiation, resulting in an excellent antitumor activity. This nanoplatform solved the dilemma of long blood circulation of large sized nanoparticles and deep penetration of small sized nanoparticles, opening up a new strategy in the development of nanomedicines for cancer therapy.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Cell Line, Tumor , Endothelial Cells , Humans , Indocyanine Green , Neoplasms/drug therapy
15.
Pharmaceutics ; 11(12)2019 Nov 29.
Article in English | MEDLINE | ID: mdl-31795486

ABSTRACT

Thermosensitive liposome-mediated drug delivery has shown promising results in terms of improved therapeutic efficacy and reduced side effects compared to conventional chemotherapeutics. In order to facilitate our understanding of the transport mechanisms and their complex interplays in the drug delivery process, computational models have been developed to simulate the multiple steps involved in liposomal drug delivery to solid tumours. In this study we employ a multicompartmental model for drug-loaded thermosensitive liposomes, with an aim to identify the key transport parameters in determining therapeutic dosing and outcomes. The computational model allows us to not only examine the temporal and spatial variations of drug concentrations in the different compartments by utilising the tumour cord concept, but also assess the therapeutic efficacy and toxicity. In addition, the influences of key factors on systemic plasma concentration and intracellular concentration of the active drug are investigated; these include different chemotherapy drugs, release rate constants and heating duration. Our results show complex relationships between these factors and the predicted therapeutic outcome, making it difficult to identify the "best" parameter set. To overcome this challenge, a model-based optimisation method is proposed in an attempt to find a set of release rate constants and heating duration that can maximise intracellular drug concentration while minimising systemic drug concentration. Optimisation results reveal that under the operating conditions and ranges examined, the best outcome would be achieved with a low drug release rate at physiological temperature, combined with a moderate to high release rate at mild hyperthermia and 1 h heating after injection.

16.
J Control Release ; 308: 197-208, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31195059

ABSTRACT

Doxorubicin is a clinically important anthracycline chemotherapeutic agent that is used to treat many cancers. Nanomedicine formulations including Doxil® and ThermoDox® have been developed to mitigate doxorubicin cardiotoxicity. Doxil is used clinically to treat ovarian cancer, AIDS-related Kaposi's sarcoma, and multiple myeloma, but there is evidence that therapeutic efficacy is hampered by lack of drug release. ThermoDox is a lipid-based heat-activated formulation of doxorubicin that relies on externally applied energy to increase tissue temperatures and efficiently trigger drug release, thereby affording therapeutic advantages compared to Doxil. However, elevating tissue temperatures is a complex treatment process requiring significant time, cost, and expertise compared to standard intravenous chemotherapy. This work endeavors to develop a companion therapeutic to ThermoDox that also relies on heat-triggered release in order to increase the therapeutic index of doxorubicin. To this end, a thermosensitive liposome formulation of the heat shock protein 90 inhibitor alvespimycin has been developed and characterized. This research demonstrates that both doxorubicin and alvespimycin are potent anti-cancer agents and that heat amplifies their cytotoxic effects. Furthermore, the two drugs are proven to act synergistically when cancer cells are treated with the drugs in combination. The formulation of alvespimycin was rationally designed to exhibit similar pharmacokinetics and drug release kinetics compared to ThermoDox, enabling the two drugs to be delivered to heated tumors at similar efficiencies resulting in control of a particular synergistic ratio of drugs. In vivo measurements demonstrated effective heat-mediated triggering of doxorubicin and alvespimycin release from thermosensitive liposomes within tumor vasculature. This treatment strategy resulted in a ~10-fold increase in drug concentration within tumors compared to free drug administered without tumor heating.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzoquinones/administration & dosage , Drug Delivery Systems , Lactams, Macrocyclic/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Doxorubicin/administration & dosage , Drug Liberation , Drug Synergism , Female , Hot Temperature , Humans , Liposomes , Mice , Mice, SCID
17.
Artif Cells Nanomed Biotechnol ; 47(1): 406-419, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30724609

ABSTRACT

A novel nanoparticle (Au-LTSL-GA.A) uses the thermosensitive liposome (LTSL) to encapsulate ganoderic acid A (GA.A), which successfully transforms the polarity of GA.A and has excellent water solubility. The multifunctional Au-LTSL-GA.A, a self-assembled thermal nanomaterial, was used in antibacterial and anticancer applications in combination with near-infrared (NIR) irradiation. The designed Au-LTSL-GA.A nanoparticle was used as a nano-photosensitizer to achieve synergistic photochemotherapy based on the phototherapy sensitization property of Au nanorods (NRs) and antitumour activity of GA.A. In the antibacterial experiments, the Au-LTSL-GA.A + NIR irradiation had a broad-spectrum antibacterial effect, exhibiting a strong antibacterial activity against drug-resistant Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) compared with the raw GA.A and LTSL-GA.A. In the anticancer experiments, Au-LTSL-GA.A + NIR irradiation, which combined phototherapy sensitization property of Au NRs with antitumour activity of GA.A, exhibited high anticancer activity against MCF-7 cells. The IC50 value of Au-LTSL-GA.A + NIR irradiation (12.1 ± 1.3 µg/mL) was almost similar to cisplatin in MCF-7 cells. The evaluation of the potential in vivo toxicity of Au-LTSL-GA.A revealed no toxicity in mice. The results of this study suggest that Au-LTSL-GA.A has a wide range of potential industrial and clinical applications, such as in antibacterial treatment and cancer photochemotherapy.


Subject(s)
Anti-Bacterial Agents , Escherichia coli/growth & development , Gold , Metal Nanoparticles , Nanotubes/chemistry , Neoplasms/drug therapy , Photochemotherapy , Staphylococcus aureus/growth & development , Triterpenes , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Gold/chemistry , Gold/pharmacology , Humans , Liposomes , MCF-7 Cells , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Neoplasms/metabolism , Neoplasms/pathology , Triterpenes/chemistry , Triterpenes/pharmacology
18.
J Liposome Res ; 29(1): 86-93, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29671386

ABSTRACT

This study aimed to develop novel temperature-sensitive liposomes loading paclitaxel (PTX-TSL) and evaluate them in vitro to improve the delivery efficiency and targeting of PTX. K237 peptide was conjugated to the terminal NHS of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[hydroxyl succinimidyl (polyethylene glycol)-(DSPE-PEG-NHS), and K237-modified PTX-TSL (K237-PTX-TSL) was prepared using a film dispersion method. K237-TSL encapsulation with calcein was synthesized and used to determine the cellular uptake of TSL. The morphology of K237-PTX-TSL was observed using a transmission electron microscope. The particle size and potential were measured using a laser particle size analyzer. The phase transition temperature was detected using the differential scanning calorimetry. The Cell Counting Kit-8 assay and flow cytometry were used to evaluate the effects of K237-PTX-TSL on the proliferation and cell cycle of cell lines SKOV-3 and human umbilical vein endothelial cell (HUVEC). The encapsulation efficiency of K237-PTX-TSL was 94.23% ± 0.76%. The particle diameter was 88.3 ± 4.7 nm. K237-PTX-TSL showed a fast release profile at 42 °C, while it was stable at 37 °C. PTX-TSL combined with hyperthermia significantly inhibited the cell proliferation of SKOV-3 cells and HUVECs due to increased cell arrest in the G2/M phase. The half-minimal inhibitory concentration value of K237-PTX-TSL on SKOV-3 cells and HUVECs was 13.61 ± 1.81 and 5.54 ± 0.95 nmol/L, respectively, which were significantly lower than those with PTX-TSL (p < 0.01). K237 modification could increase the targeting efficiency of TSL to cancer cells and vascular endothelial cells, thus resulting in higher cytotoxicities compared with PTX-TSL, which might be a potential formulation for targeting cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Liposomes , Oligopeptides , Paclitaxel/administration & dosage , Animals , Cell Line , Humans , Liposomes/chemistry , Phosphatidylethanolamines , Polyethylene Glycols , Transition Temperature
19.
ACS Appl Mater Interfaces ; 10(17): 14426-14437, 2018 May 02.
Article in English | MEDLINE | ID: mdl-29651836

ABSTRACT

Biofilm is closely related to chronic infections and is difficult to eradicate. Development of effective therapy strategies to control biofilm infection is still challenging. Aiming at biofilm architecture, we designed and prepared near-infrared-activated thermosensitive liposomes with photothermal and antibiotic synergistic therapy capacity to eliminate Pseudomonas aeruginosa biofilm. The liposomes with positive charge and small size aided to enter the biofilm microchannels and locally released antibiotics in infection site. The liposomes could remain stable at 37 °C and release about 80% antibiotics over 45 °C. The biofilm dispersion rate was up to 80%, which was a 7- to 8-fold rise compared to excess antibiotic alone, indicating that the localized antibiotic release and photothermal co-therapy improved the antimicrobial efficiency. In vivo drug-loaded liposomes in treating P. aeruginosa-induced abscess exhibited an outstanding therapeutic effect. Furthermore, photothermal treatment could stimulate the expression of bcl2-associated athanogene 3 to prevent normal tissue from thermal damage. The near-infrared-activated nanoparticle carriers had the tremendous therapeutic potential to dramatically enhance the efficacy of antibiotics through thermos-triggered drug release and photothermal therapy.


Subject(s)
Biofilms , Anti-Bacterial Agents , Infrared Rays , Liposomes , Pseudomonas aeruginosa
20.
J Control Release ; 282: 35-45, 2018 07 28.
Article in English | MEDLINE | ID: mdl-29673642

ABSTRACT

Triple negative breast cancer is an aggressive disease that accounts for at least 15% of breast cancer diagnoses, and a disproportionately high percentage of breast cancer related morbidity. Intensive research efforts are focused on the development of more efficacious treatments for this disease, for which therapeutic options remain limited. The high incidence of mutations in key DNA repair pathways in triple negative breast cancer results in increased sensitivity to DNA damaging agents, such as platinum-based chemotherapies. Hyperthermia has been successfully used in breast cancer treatment to sensitize tumors to radiation therapy and chemotherapy. It has also been used as a mechanism to trigger drug release from thermosensitive liposomes. In this study, mild hyperthermia is used to trigger release of cisplatin from thermosensitive liposomes in the vasculature of human triple negative breast cancer tumors implanted orthotopically in mice. This heat-triggered liposomal formulation of cisplatin resulted in significantly delayed tumor growth and improved overall survival compared to treatment with either non-thermosensitive liposomes containing cisplatin or free cisplatin, as was observed in two independent tumor models (i.e. MDA-MB-231 and MDA-MB-436). The in vitro sensitivity of the cell lines to cisplatin and hyperthermia alone and in combination was characterized extensively using enzymatic assays, clonogenic assays, and spheroid growth assays. Evaluation of correlations between the in vitro and in vivo results served to identify the in vitro approach that is most predictive of the effects of hyperthermia in vivo. Relative expression of several heat shock proteins and the DNA damage repair protein BRCA1 were assayed at baseline and in response to hyperthermia both in vitro and in vivo. Interestingly, delivery of cisplatin in thermosensitive liposomes in combination with hyperthermia resulted in the most significant tumor growth delay, relative to free cisplatin, in the less cisplatin-sensitive cell line (i.e. MDA-MB-231). This work demonstrates that thermosensitive cisplatin liposomes used in combination with hyperthermia offer a novel method for effective treatment of triple negative breast cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Delayed-Action Preparations/chemistry , Drug Delivery Systems/methods , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Breast/blood supply , Breast/drug effects , Breast/pathology , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Humans , Hyperthermia, Induced/methods , Liposomes/chemistry , Mice, SCID , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL