Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Mater Chem B ; 10(39): 7955-7966, 2022 10 12.
Article in English | MEDLINE | ID: mdl-35792081

ABSTRACT

Here, a novel joint chemo/photothermal/chemodynamic therapy was developed using a pH/GSH/photo triple-responsive 2D-covalent organic framework (COF) drug carriers for passive target treatment of tumors with extraordinarily high efficiency. The well-designed COF (DiSe-Por) with simultaneous dynamic diselenium and imine bonds, synthesized by the copolymerization of 4,4'-diselanediyldibenzaldehyde (DiSe) with 5,10,15,20-(tetra-4-aminophenyl)-porphyrin (Por) via Schiff base chemistry, which was applied as the host for effective encapsulation and highly controlled release of anticancer drug (DOX), was stable under normal physiological settings and can effectively accumulate in tumor sites. After being internalized into the tumor cells, the unique microenvironment i.e., acidic pH and overexpressed GSH, triggered substantial degradation of DiSe-Por-DOX, promoting DOX release to kill the cancer cells. Meanwhile, the breaking of Se-Se bonds boosted the generation of intracellular ROS, disturbing the redox balance of tumor cells. The highly extended 2D structure endowed the drug delivery system with significant photothermal performance. The rise of temperature with external laser irradiation (808 nm) further promoted drug release. Additionally, the phototherapy effect was further augmented after the loading of DOX, guaranteeing an almost complete drug release to tumor tissue. As a result, the triple-responsive drug delivery system achieved a synergistic amplified therapeutic efficacy with a growth inhibitory rate of approximately 93.5% for the tumor xenografted in nude mice. Moreover, the body metabolizable and clearable DiSe-Por-DOX presented negligible toxicities toward major organs in vivo. All these characteristics verified the great potential of DiSe-Por-DOX nanosheets for multi-modality tumor treatment, accelerating the application range of COFs in biomedical fields.


Subject(s)
Antineoplastic Agents , Hyperthermia, Induced , Metal-Organic Frameworks , Neoplasms , Porphyrins , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Doxorubicin/chemistry , Drug Carriers/chemistry , Drug Liberation , Hydrogen-Ion Concentration , Metal-Organic Frameworks/metabolism , Metal-Organic Frameworks/pharmacology , Mice , Mice, Nude , Neoplasms/drug therapy , Phototherapy , Porphyrins/metabolism , Reactive Oxygen Species/metabolism , Schiff Bases
2.
Chem Biol Interact ; 351: 109718, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34717915

ABSTRACT

The ABCG2 transporter plays a pivotal role in multidrug resistance, however, no clinical trial using specific ABCG2 inhibitors have been successful. Although ABC transporters actively extrude a wide variety of substrates, photodynamic therapeutic agents with porphyrinic scaffolds are exclusively transported by ABCG2. In this work, we describe for the first time a porphyrin derivative (4B) inhibitor of ABCG2 and capable to overcome multidrug resistance in vitro. The inhibition was time-dependent and 4B was not itself transported by ABCG2. Independently of the substrate, the porphyrin 4B showed an IC50 value of 1.6 µM and a mixed type of inhibition. This compound inhibited the ATPase activity and increased the binding of the conformational-sensitive antibody 5D3. A thermostability assay confirmed allosteric protein changes triggered by the porphyrin. Long-timescale molecular dynamics simulations revealed a different behavior between the ABCG2 porphyrinic substrate pheophorbide a and the porphyrin 4B. Pheophorbide a was able to bind in three different protein sites but 4B showed one binding conformation with a strong ionic interaction with GLU446. The inhibition was selective toward ABCG2, since no inhibition was observed for P-glycoprotein and MRP1. Finally, this compound successfully chemosensitized cells that overexpress ABCG2. These findings reinforce that substrates may be a privileged source of chemical scaffolds for identification of new inhibitors of multidrug resistance-linked ABC transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Adenosine Triphosphatases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Porphyrins/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2/chemistry , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Resistance, Multiple/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Irinotecan/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Porphyrins/chemistry , Porphyrins/metabolism , Protein Binding , Protein Conformation/drug effects
3.
Chem Commun (Camb) ; 57(59): 7296-7299, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34223569

ABSTRACT

A chlorine e6 (Ce6) and curcumin (Cur) based self-delivery nanomedicine (CeCu) is prepared for chemotherapy sensitized photodynamic therapy (PDT). The chemotherapeutic agent of Cur could inhibit the TrxR activity to destroy the cellular ROS-defence system for enhanced PDT, which provides synergistic effects for tumor precision therapy in consideration of the unfavorable tumor microenvironments.


Subject(s)
Nanomedicine , Photochemotherapy/methods , Animals , Cell Line , Cell Survival/drug effects , Chlorophyllides , Curcumin/chemistry , Curcumin/metabolism , Curcumin/pharmacology , Humans , Mice , Microscopy, Confocal , Neoplasms/drug therapy , Neoplasms/pathology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Porphyrins/chemistry , Porphyrins/metabolism , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Transplantation, Heterologous , Tumor Microenvironment
4.
J Inorg Biochem ; 219: 111431, 2021 06.
Article in English | MEDLINE | ID: mdl-33798828

ABSTRACT

Oxidative stress that results from an imbalance between the concentrations of reactive species (RS) and antioxidant defenses is associated with many pathologies. Superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase are among the key enzymes that maintain the low nanomolar physiological concentrations of superoxide and hydrogen peroxide. The increase in the levels of these species and their progeny could have deleterious effects. In this context, chemists have developed SOD and CAT mimics to supplement them when cells are overwhelmed with oxidative stress. However, the beneficial activity of such molecules in cells depends not only on their intrinsic catalytic activities but also on their stability in biological context, their cell penetration and their cellular localization. We have employed cellular assays to characterize several compounds that possess SOD and CAT activities and have been frequently used in cellular and animal models. We used cellular assays that address SOD and CAT activities of the compounds. Finally, we determined the effect of compounds on the suppression of the inflammation in HT29-MD2 cells challenged by lipopolysaccharide. When the assay requires penetration inside cells, the SOD mimics Mn(III) meso-tetrakis(N-(2'-n-butoxyethyl)pyridinium-2-yl)porphyrin (MnTnBuOE-2-PyP5+) and Mn(II) dichloro[(4aR,13aR,17aR,21aR)-1,2,3,4,4a,5,6,12,13,13a,14,15,16,17,17a,18,19,20,21,21a-eicosahydro-11,7-nitrilo-7Hdibenzo[b,h] [1,4, 7,10] tetraazacycloheptadecine-κN5,κN13,κN18,κN21,κN22] (Imisopasem manganese, M40403, CG4419) were found efficacious at 10 µM, while Mn(II) chloro N-(phenolato)-N,N'-bis[2-(N-methyl-imidazolyl)methyl]-ethane-1,2-diamine (Mn1) requires an incubation at 100 µM. This study thus demonstrates that MnTnBuOE-2-PyP5+, M40403 and Mn1 were efficacious in suppressing inflammatory response in HT29-MD2 cells and such action appears to be related to their ability to enter the cells and modulate reactive oxygen species (ROS) levels.


Subject(s)
Catalase/metabolism , Manganese/metabolism , Organometallic Compounds/metabolism , Superoxide Dismutase/metabolism , Animals , Antioxidants/metabolism , Cell Line , Glutathione Peroxidase/metabolism , Humans , Hydrogen Peroxide/metabolism , Metalloporphyrins/metabolism , Molecular Mimicry , Oxidation-Reduction , Oxidative Stress , Porphyrins/metabolism , Reactive Oxygen Species/metabolism , Superoxides/metabolism
5.
ACS Appl Mater Interfaces ; 11(36): 32633-32646, 2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31429272

ABSTRACT

The concept of integrating immunogenic cell death (ICD) with tailoring the immunosuppressive tumor microenvironment (TME) is promising for immunotherapy. Photothermal therapy (PTT) could efficiently induce ICD, while an indoleamine 2,3-dioxygenase (IDO) inhibitor could convert the "cold" TME. Therefore, combination of PTT and the IDO inhibitor is an attractive approach for immunotherapy. Unfortunately, combination of PTT and the IDO inhibitor for tumor therapy is rarely reported. Herein, organic photothermal agent IR820 and IDO inhibitor 1-methyl-tryptophan (1MT) were, for the first time, designed to be an all-rolled-into-one molecule nanoplatform via a molecular engineering strategy. The designed IR820-1MT molecule could self-assemble into nanoparticles with remarkably high dual-therapeutic agent loading (88.8 wt %). Importantly, poor water solubility of 1MT and inadequate targeting and short lifetime of IR820 were all well solved within as-prepared IR820-1MT nanoparticles. The laser-triggered IR820-1MT nanoparticles remarkably enhanced accumulation of cytotoxic T cells, helper T cells, and memory T cells and simultaneously suppressed a proportion of regulatory T cells, resulting in excellent immunotherapy against tumor metastasis and recurrence. Our molecular engineering strategy provides a promising alternative option for design of a robust immunotherapy weapon against tumor metastasis and recurrence.


Subject(s)
Immunotherapy , Nanoparticles/chemistry , Phototherapy , Animals , Chlorophyllides , Dendritic Cells/metabolism , Female , Immunoglobulin G/metabolism , Indocyanine Green/analogs & derivatives , Indocyanine Green/chemistry , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Neoplasm Metastasis , Porphyrins/metabolism
6.
Nanoscale ; 11(19): 9457-9467, 2019 May 16.
Article in English | MEDLINE | ID: mdl-31042245

ABSTRACT

Multifunctional nanodrugs with the integration of precise diagnostic and effective therapeutic functions have shown great promise in improving the efficacy of cancer therapy. We report herein a simple and effective approach to directly assemble an anticancer drug (curcumin), a photodynamic agent (Ce6) and tumor environment-sensitive molecules into cross-linked polyphosphazene and coat on superparamagnetic Fe3O4 nanoclusters to form discrete nanoparticles (termed as FHCPCe NPs). FHCPCe NPs have high physiological stability and good biocompatibility, and can enhance accumulation in tumor tissue via the enhanced permeability and retention effect. Meanwhile, the FHCPCe NPs exhibit an effective performance of dual-modality magnetic resonance imaging (MRI) due to the Fe3O4 cores and fluorescence imaging (FL) in the xenografted HeLa tumor because of the fluorescence of Ce6. Importantly, under the conditions of supernormal glutathione levels and acidic microenvironment in tumor tissue, curcumin and Ce6 can be effectively released by the degradation of FHCPCe NPs. Therefore, excellent anti-tumor effects both in vitro and in vivo have been achieved by synergistic chemotherapy/photodynamic therapy (CT/PDT) using multifunctional NPs. Our study highlights the promise of developing multifunctional nanomaterials for accurate multimodal imaging-guided highly sensitive therapy of cancer.


Subject(s)
Nanoparticles/chemistry , Organophosphorus Compounds/chemistry , Polymers/chemistry , Animals , Chlorophyllides , Curcumin/chemistry , Curcumin/metabolism , Curcumin/therapeutic use , Drug Carriers/chemistry , Ferrosoferric Oxide/chemistry , HeLa Cells , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Oxidation-Reduction , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Photosensitizing Agents/therapeutic use , Porphyrins/chemistry , Porphyrins/metabolism , Porphyrins/therapeutic use , Reactive Oxygen Species/metabolism , Transplantation, Heterologous , Whole Body Imaging
7.
Theranostics ; 8(19): 5501-5518, 2018.
Article in English | MEDLINE | ID: mdl-30555560

ABSTRACT

The limited clinical efficacy of monotherapies in the clinic has urged the development of novel combination platforms. Taking advantage of light-triggered photodynamic treatment combined together with the controlled release of nanomedicine, it has been possible to treat cancer without eliciting any adverse effects. However, the challenges imposed by limited drug loading capacity and complex synthesis process of organic nanoparticles (NPs) have seriously impeded advances in chemo-photodynamic combination therapy. In this experiment, we utilize our previously synthesized porphyrin-grafted lipid (PGL) NPs to load highly effective chemotherapeutic drug, doxorubicin (DOX) for synergistic chemo-photodynamic therapy. Methods: A relatively simple and inexpensive rapid injection method was used to prepare porphyrin-grafted lipid (PGL) NPs. The self-assembled PGL NPs were used further to encapsulate DOX via a pH-gradient loading protocol. The self-assembled liposome-like PGL NPs having a hydrophilic core were optimized to load DOX at an encapsulation efficiency (EE) of ~99%. The resultant PGL-DOX NPs were intact, highly stable and importantly these NPs successfully escaped from the endo-lysosomal compartment after laser irradiation to release DOX in the cytosol. The therapeutic efficacy of the aforementioned formulation was validated both in vitro and in vivo. Results: PGL-DOX NPs demonstrated excellent cellular uptake, chemo-photodynamic response, and fluorescence imaging ability in different cell lines. Under laser irradiation, cells treated with a low molar concentration of PGL-DOX NPs reduced cell viability significantly. Moreover, in vivo experiments conducted in a xenograft mouse model further demonstrated the excellent tumor accumulation capability of PGL-DOX NPs driven by the enhanced permeability and retention (EPR) effect. Through fluorescence imaging, the biodistribution of PGL-DOX NPs in tumor and major organs was also easily monitored in real time in vivo. The inherent ability of porphyrin to generate ROS under laser irradiation combined with the cytotoxic effect of the anticancer drug DOX significantly suppressed tumor growth in vivo. Conclusion: In summary, the PGL-DOX NPs combined chemo-photodynamic nanoplatform may serve as a potential candidate for cancer theranostics.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Doxorubicin/metabolism , Drug Carriers/chemistry , Fluorescent Dyes/metabolism , Liposomes/chemistry , Porphyrins/metabolism , Theranostic Nanomedicine/methods , Animals , Carcinoma/therapy , Cell Survival/drug effects , Disease Models, Animal , Drug Therapy/methods , HeLa Cells , Heterografts , Humans , Low-Level Light Therapy , Mice , Nanoparticles/chemistry , Neoplasm Transplantation , Optical Imaging/methods , PC-3 Cells , Photochemotherapy/methods , Treatment Outcome
8.
J Photochem Photobiol B ; 178: 182-191, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29156346

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) of malignant brain tumors is a promising adjunct to standard treatment, especially if tumor stem cells thought to be responsible for tumor progression and therapy resistance were also susceptible to this kind of treatment. However, some photosensitizers have been reported to be substrates of ABCG2, one of the membrane transporters mediating resistance to chemotherapy. Here we investigate, whether inhibition of ABCG2 can restore sensitivity to photosensitizer chlorin e6-mediated PDT. METHODS: Accumulation of chlorin e6 in wild type U87 and doxycycline-inducible U251 glioblastoma cells with or without induction of ABCG2 expression or ABCG2 inhibition by KO143 was analyzed using flow cytometry. In U251 cells, ABCG2 was inducible by doxycycline after stable transfection with a tet-on expression plasmid. Tumor sphere cultivation under low attachment conditions was used to enrich for cells with stem cell-like properties. PDT was done on monolayer cell cultures by irradiation with laser light at 665nm. RESULTS: Elevated levels of ABCG2 in U87 cells grown as tumor spheres or in U251 cells after ABCG2 induction led to a 6-fold lower accumulation of chlorin e6 and the light dose needed to reduce cell viability by 50% (LD50) was 2.5 to 4-fold higher. Both accumulation and PDT response can be restored by KO143, an efficient non-toxic inhibitor of ABCG2. CONCLUSION: Glioblastoma stem cells might escape phototoxic destruction by ABCG2-mediated reduction of photosensitizer accumulation. Inhibition of ABCG2 during photosensitizer accumulation and irradiation promises to restore full susceptibility of this crucial tumor cell population to photodynamic treatment.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Diketopiperazines/toxicity , Gene Expression/drug effects , Heterocyclic Compounds, 4 or More Rings/toxicity , Photosensitizing Agents/metabolism , Porphyrins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chlorophyllides , Doxycycline/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Light , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/toxicity , Porphyrins/chemistry , Porphyrins/toxicity , Sorafenib
9.
J Eur Acad Dermatol Venereol ; 32(1): 113-116, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28662298

ABSTRACT

BACKGROUND: Aesthetically pleasing results and fast, uneventful recovery are highly desirable after rejuvenating ablative laser procedures. Wound dressings following ablative laser procedures should ideally improve and optimize the wound healing environment. OBJECTIVE: The purpose of this comparative split-face, single-blinded, prospective observational study was to assess the efficacy and acceptability of two primary wound dressings immediately after a full-face fractional CO2 laser resurfacing procedure. METHODS: The assessments of an innovative film-forming dressing called Stratacel (SC) vs spring thermal water + Vaseline (V+) were conducted after a standardized, single-pass, full-face ablative fractional CO2 laser skin resurfacing procedure. Clinical parameters, such as haemoglobin - HB; surface temperature - ST; micro-textural modifications - MT; superficial melanin - M; intrafollicular porphyrins - P, were assessed at different phases of the healing process using standardized, non-invasive technologies. RESULTS: Five female volunteers were enrolled in this inpatient, controlled pilot study. Most of the clinical parameters considered, including 3D surface texture analysis, revealed a better performance of SC vs. V+ during the early, more delicate phases of the healing process. CONCLUSIONS: This preliminary study, even if performed on a small number of volunteers, confirmed a definite advantage of the tested semipermeable film-forming formula (SC) over a more conventional postoperative skin care regime (V+). Clinical results could be explained by a better uniformity of distribution of SC over the micro-irregularities induced by ablative fractional CO2 laser resurfacing. Its thin, semipermeable film might, in fact, act as an efficient, perfectly biocompatible, full contact, temporary skin barrier, able to protect extremely delicate healing surfaces from potential environmental irritations.


Subject(s)
Cosmetic Techniques , Lasers, Gas/therapeutic use , Occlusive Dressings , Petrolatum/therapeutic use , Water , Ablation Techniques/adverse effects , Adult , Carbon Dioxide , Cosmetic Techniques/adverse effects , Face , Female , Gels , Hemoglobins/metabolism , Humans , Pilot Projects , Porphyrins/metabolism , Prospective Studies , Skin/metabolism , Skin Physiological Phenomena , Water/metabolism , Wound Healing
10.
Eur J Cancer ; 83: 19-27, 2017 09.
Article in English | MEDLINE | ID: mdl-28709135

ABSTRACT

Photodynamic therapy (PDT) has been shown to destroy tumour-associated lymphatic vessels. Therefore, we sought to investigate the functional outcomes of PDT-mediated damage to the lymphatic vessels. We observed that PDT with verteporfin, completely but transiently, blocks the functional lymphatic drainage in the orthotopic mammary tumour models. Sustained inhibition of lymphatic vessels regeneration induced by lenalidomide or the soluble form of vascular endothelial growth factor receptor 3 (sVEGFR3) that neutralises lymphangiogenic vascular endothelial growth factor C (VEGF-C), significantly impaired antitumour efficacy of PDT. Antilymphangiogenic compounds also significantly inhibited the ability of intratumourally inoculated dendritic cells (DCs) to translocate to local lymph nodes and diminished the number of tumour-infiltrating interferon-γ-secreting or tumour antigen-specific CD8+ T cells. Lenalidomide also abrogated antitumour effects of the combination immunotherapy with PDT and anti-programmed death-ligand 1 (PD-L1) antibodies. Altogether, these findings indicate that PDT-mediated damage to the lymphatic vessels negatively affects development of antitumour immunity, and that drugs that impair lymphatic vessel regeneration might not be suitable for the use in combination with PDT.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Lymphangiogenesis/drug effects , Photochemotherapy , Porphyrins/metabolism , Porphyrins/pharmacology , Thalidomide/analogs & derivatives , Animals , Disease Models, Animal , Female , Lenalidomide , Lymphangiogenesis/radiation effects , Lymphatic Vessels/drug effects , Lymphatic Vessels/pathology , Mice , Mice, Inbred C57BL , Photosensitizing Agents/pharmacology , Thalidomide/pharmacology , Vascular Endothelial Growth Factor Receptor-3/pharmacology , Verteporfin
SELECTION OF CITATIONS
SEARCH DETAIL