Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.095
Filter
1.
Photodiagnosis Photodyn Ther ; 46: 104047, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38503388

ABSTRACT

BACKGROUND: Hypoxia is a characteristic feature of many tumors. It promotes tumor proliferation, metastasis, and invasion and can reduce the effectiveness of many types of cancer treatment. OBJECTIVE: The aim of this study was to investigate the pharmacokinetics of methylene blue (MB) and its impact on the tumor oxygenation level at mouse Lewis lung carcinoma (LLC) model using spectroscopic methods. APPROACH: The pharmacokinetics of MB were studied qualitatively and quantitatively using video fluorescence imaging and fluorescence spectroscopy. The degree of hemoglobin oxygenation in vivo was examined by calculating hemoglobin optical absorption from the measured diffuse reflectance spectra. The distribution of MB fluorescence and the lifetime of NADH were analyzed using laser scanning microscopy and fluorescence lifetime imaging microscopy (FLIM) to assess cellular metabolism. RESULTS: After intravenous administration of MB at 10-20 mg/kg, it quickly transitioned in the tumor to a colorless leucomethylene blue, with maximum accumulation in the tumor occurring after 5-10 min. A concentration of 10 mg/kg resulted in a relative increase of the tumor oxygenation level for small tumors (volume 50-75 mm3) and normal tissue 120 min after the introduction of MB. A shift in tumor metabolism towards oxidative phosphorylation (according to the lifetime of the NADH coenzyme) was measured using FLIM method after intravenous administration of 10 mg/kg of MB. Intravenous administration of MB at 20 mg/kg results in a long-term decrease in oxygenation, which persisted for at least 120 min after the administration and did not return to its initial level. CONCLUSIONS: Administration of MB at 10 mg/kg shown to increase tumor oxygenation level, potentially leading to more effective antitumor therapy. However, at higher doses (20 mg/kg), MB may cause long-term decrease in oxygenation.


Subject(s)
Carcinoma, Lewis Lung , Methylene Blue , Methylene Blue/pharmacology , Methylene Blue/pharmacokinetics , Animals , Mice , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Oxygen/metabolism , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Mice, Inbred C57BL , Dose-Response Relationship, Drug , Photochemotherapy/methods , Cell Line, Tumor , Spectrometry, Fluorescence
2.
Adv Drug Deliv Rev ; 182: 114134, 2022 03.
Article in English | MEDLINE | ID: mdl-35122881

ABSTRACT

Photoimaging and phototherapy have become major platforms for the diagnosis and treatment of various health complications. These applications require a photosensitizer (PS) that is capable of absorbing light from a source and converting it into other energy forms for detection and therapy. While synthetic inorganic materials such as quantum dots and gold nanorods have been widely explored for their medical diagnosis and photodynamic (PDT) and photothermal (PTT) therapy capabilities, translation of these technologies has lagged, primarily owing to potential cytotoxicity and immunogenicity issues. Of the various photoreactive molecules, the naturally occurring endogenous compound heme, a constituent of red blood cells, and its derivatives, porphyrin, biliverdin and bilirubin, have shown immense potential as noteworthy candidates for clinically translatable photoreactive agents, as evidenced by previous reports. While porphyrin-based photomedicines have attracted significant attention and are well documented, research on photomedicines based on two other heme-derived compounds, biliverdin and bilirubin, has been relatively lacking. In this review, we summarize the unique photoproperties of heme-derived compounds and outline recent efforts to use them in biomedical imaging and phototherapy applications.


Subject(s)
Diagnostic Imaging/methods , Heme/pharmacology , Photosensitizing Agents/pharmacology , Phototherapy/methods , Heme/administration & dosage , Heme/pharmacokinetics , Humans , Nanoparticle Drug Delivery System , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Porphyrins/administration & dosage , Porphyrins/pharmacology
3.
Drug Deliv ; 29(1): 238-253, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35001784

ABSTRACT

Photodynamic therapy (PDT) has been applied in cancer treatment by utilizing reactive oxygen species (ROS) to kill cancer cells. However, the effectiveness of PDT is greatly reduced due to local hypoxia. Hypoxic activated chemotherapy combined with PDT is expected to be a novel strategy to enhance anti-cancer therapy. Herein, a novel liposome (LCT) incorporated with photosensitizer (PS) and bioreductive prodrugs was developed for PDT-activated chemotherapy. In the design, CyI, an iodinated cyanine dye, which could simultaneously generate enhanced ROS and heat than other commonly used cyanine dyes, was loaded into the lipid bilayer; while tirapazamine (TPZ), a hypoxia-activated prodrug was encapsulated in the hydrophilic nucleus. Upon appropriate near-infrared (NIR) irradiation, CyI could simultaneously produce ROS and heat for synergistic PDT and photothermal therapy (PTT), as well as provide fluorescence signals for precise real-time imaging. Meanwhile, the continuous consumption of oxygen would result in a hypoxia microenvironment, further activating TPZ free radicals for chemotherapy, which could induce DNA double-strand breakage and chromosome aberration. Moreover, the prepared LCT could stimulate acute immune response through PDT activation, leading to synergistic PDT/PTT/chemo/immunotherapy to kill cancer cells and reduce tumor metastasis. Both in vitro and in vivo results demonstrated improved anticancer efficacy of LCT compared with traditional PDT or chemotherapy. It is expected that these iodinated cyanine dyes-based liposomes will provide a powerful and versatile theranostic strategy for tumor target phototherapy and PDT-induced chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Hypoxia/pathology , Nanoparticle Drug Delivery System/chemistry , Photosensitizing Agents/pharmacology , Phototherapy/methods , Tirapazamine/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cell Survival/drug effects , Chemistry, Pharmaceutical , Chromosome Aberrations/drug effects , DNA Damage/drug effects , Drug Carriers/chemistry , Drug Liberation , Liposomes/chemistry , Mice , Mice, Inbred BALB C , Particle Size , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Reactive Oxygen Species/metabolism , Surface Properties , Tirapazamine/administration & dosage , Tirapazamine/pharmacokinetics , Xenograft Model Antitumor Assays
4.
Clin Pharmacol Drug Dev ; 11(4): 535-550, 2022 04.
Article in English | MEDLINE | ID: mdl-34633154

ABSTRACT

The nanoemulsion-based 10% aminolevulinic acid (ALA) hydrochloride gel BF-200 ALA optimizes epidermal penetration of its active ingredient and is approved for topical photodynamic therapy (PDT) for the treatment of actinic keratosis in the United States and Europe. To characterize systemic absorption from dermal application during PDT, ALA and its key active metabolite protoporphyrin IX (PpIX) were analyzed in 2 maximal usage pharmacokinetic trials (MUsT) in patients severely affected with actinic keratosis. The primary objective of both MUsTs was to assess baseline-adjusted plasma concentration-time curves for ALA and PpIX after a single PDT treatment applying either 2 g (1 tube) of BF-200 ALA on the face (MUsT-1) or applying 6 g (3 tubes) of BF-200 ALA on the face/scalp or body periphery (MUsT-2), to 20 or 60 cm2 , respectively. All PDTs were performed using red light at around 635 nm wavelength. Safety and tolerability were documented along with pharmacokinetics. In both MUsTs, ALA plasma concentrations were transiently increased to a maximum concentration at about 2.5 to 3.3 times above endogenous baseline with time to maximum concentration at ≈3 hours after dosing. Plasma levels subsequently returned to baseline within 10 hours after dosing. Overall baseline-adjusted mean area under the baseline-adjusted plasma concentration-time curve from time zero to the last sampling time point at which the concentration was at or above the lower limit of quantification ranged from 142.8 to 146.2, indicating that a similar, minor fraction of topical ALA is systemically absorbed under both dosing regimens. Systemic PpIX exposure after administration of either dose of BF-200 ALA was equally minimal. Application site skin reactions were treatment area size-related, albeit transient and consistent with the known safety profile of BF-200 ALA.


Subject(s)
Keratosis, Actinic , Photochemotherapy , Aminolevulinic Acid/adverse effects , Aminolevulinic Acid/analogs & derivatives , Humans , Keratosis, Actinic/drug therapy , Photochemotherapy/adverse effects , Photochemotherapy/methods , Photosensitizing Agents/adverse effects , Photosensitizing Agents/pharmacokinetics
5.
J Nanobiotechnology ; 19(1): 440, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34930284

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) is a promising therapeutic modality that can convert oxygen into cytotoxic reactive oxygen species (ROS) via photosensitizers to halt tumor growth. However, hypoxia and the unsatisfactory accumulation of photosensitizers in tumors severely diminish the therapeutic effect of PDT. In this study, a multistage nanoplatform is demonstrated to overcome these limitations by encapsulating photosensitizer IR780 and oxygen regulator 3-bromopyruvate (3BP) in poly (lactic-co-glycolic acid) (PLGA) nanocarriers. RESULTS: The as-synthesized nanoplatforms penetrated deeply into the interior region of tumors and preferentially remained in mitochondria due to the intrinsic characteristics of IR780. Meanwhile, 3BP could efficiently suppress oxygen consumption of tumor cells by inhibiting mitochondrial respiratory chain to further improve the generation of ROS. Furthermore, 3BP could abolish the excessive glycolytic capacity of tumor cells and lead to the collapse of ATP production, rendering tumor cells more susceptible to PDT. Successful tumor inhibition in animal models confirmed the therapeutic precision and efficiency. In addition, these nanoplatforms could act as fluorescence (FL) and photoacoustic (PA) imaging contrast agents, effectuating imaging-guided cancer treatment. CONCLUSIONS: This study provides an ideal strategy for cancer therapy by concurrent oxygen consumption reduction, oxygen-augmented PDT, energy supply reduction, mitochondria-targeted/deep-penetrated nanoplatforms and PA/FL dual-modal imaging guidance/monitoring. It is expected that such strategy will provide a promising alternative to maximize the performance of PDT in preclinical/clinical cancer treatment.


Subject(s)
Mitochondria/drug effects , Nanoparticles/chemistry , Photosensitizing Agents/pharmacology , Tumor Hypoxia/drug effects , Animals , Cell Line, Tumor , Drug Synergism , Female , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/pharmacology , Indoles/therapeutic use , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Pyruvates/chemistry , Pyruvates/pharmacokinetics , Pyruvates/pharmacology , Pyruvates/therapeutic use , Reactive Oxygen Species/metabolism , Tissue Distribution , Transplantation, Heterologous
6.
J Nanobiotechnology ; 19(1): 379, 2021 Nov 21.
Article in English | MEDLINE | ID: mdl-34802438

ABSTRACT

Matrix metalloproteinase (MMP) 2 and 9 are the family members of proteases normally up-regulated in tumor to enhance the invasion and metastatic of tumor cells, and are associated with poor outcome of head and neck squamous cell carcinomas (HNSCCs). In the present work, MMPs-degradable gelatin nanoparticles (GNPs) are simultaneously loaded with photosensitizer indocyanine green (ICG) along with signal transducer activator of transcription 3 (STAT3) inhibitor NSC74859 (NSC, N) for efficient photothermal therapy (PTT) and immunotherapy of HNSCCs. In the tumor tissue, Gel-N-ICG nanoparticle was degraded and encapsulated ICG and NSC were effectively released. Under near-infrared (NIR) irradiation, the released ICG nanoparticles enabled effective photothermal destruction of tumors, and the STAT3 inhibitor NSC elicited potent antitumor immunity for enhanced cancer therapy. Based on two HNSCC mouse models, we demonstrated that Gel-N-ICG significantly delayed tumor growth without any appreciable body weight loss. Taken together, the strategy reported here may contribute that the stimuli-responsive proteases triggered nanoplatform could reduce tumor size more effectively in complex tumor microenvironment (TME) through combination of PTT and immunotherapy.


Subject(s)
Gelatinases/metabolism , Nanoparticles , Photosensitizing Agents , Protein Inhibitors of Activated STAT , Animals , Cell Line, Tumor , Cell Survival/drug effects , Immunotherapy , Indocyanine Green/chemistry , Indocyanine Green/pharmacokinetics , Mice , Nanoparticles/chemistry , Nanoparticles/metabolism , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Photothermal Therapy , Protein Inhibitors of Activated STAT/chemistry , Protein Inhibitors of Activated STAT/pharmacokinetics , Protein Inhibitors of Activated STAT/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors
7.
Exp Eye Res ; 213: 108842, 2021 12.
Article in English | MEDLINE | ID: mdl-34793829

ABSTRACT

Avoiding damage of the endothelial cells, especially in thin corneas, remains a challenge in corneal collagen crosslinking (CXL). Knowledge of the riboflavin gradients and the UV absorption characteristics after topical application of riboflavin in concentrations ranging from 0.1% to 0.5% could optimize the treatment. In this study, we present a model to calculate the UV-intensity depending on the corneal thickness. Ten groups of de-epithelialized porcine corneas were divided into 2 subgroups. Five groups received an imbibition of 10 min and the other five groups for 30 min. The applied riboflavin concentrations were 0.1%, 0.2%, 0.3%, 0.4% and 0.5% diluted in a 15% dextran solution for each subgroup. After the imbibition process, two-photon fluorescence microscopy was used to determine fluorescence intensity, which was compared to samples after saturation, yielding the absolute riboflavin concentration gradient of the cornea. The extinction coefficient of riboflavin solutions was measured using a spectrophotometer. Combining the obtained riboflavin concentrations and the extinction coefficients, a depth-dependent UV-intensity profile was calculated for each group. With increasing corneal depth, the riboflavin concentration decreased for all imbibition solutions and application times. The diffusion coefficients of 10 min imbibition time were higher than for 30 min. A higher RF concentration and a longer imbibition time resulted in higher UV-absorption and a lower UV-intensity in the depth of the cornea. Calculated UV-transmission was 6 percentage points lower compared to the measured transmission. By increasing the riboflavin concentration of the imbibition solution, a substantially higher UV-absorption inside the cornea is achieved. This offers a simple treatment option to control the depth of crosslinking e.g. in thin corneas, resulting in a lower risk of endothelial damage.


Subject(s)
Absorption, Radiation/drug effects , Corneal Stroma/metabolism , Photosensitizing Agents/pharmacokinetics , Riboflavin/pharmacokinetics , Ultraviolet Rays , Administration, Ophthalmic , Animals , Corneal Pachymetry , Corneal Stroma/radiation effects , Cross-Linking Reagents , Microscopy, Fluorescence, Multiphoton , Ophthalmic Solutions , Photochemotherapy , Photosensitizing Agents/administration & dosage , Riboflavin/administration & dosage , Swine
8.
J Nanobiotechnology ; 19(1): 338, 2021 Oct 24.
Article in English | MEDLINE | ID: mdl-34689768

ABSTRACT

BACKGROUND: Photoactivation targeting macrophages has emerged as a therapeutic strategy for atherosclerosis, but limited targetable ability of photosensitizers to the lesions hinders its applications. Moreover, the molecular mechanistic insight to its phototherapeutic effects on atheroma is still lacking. Herein, we developed a macrophage targetable near-infrared fluorescence (NIRF) emitting phototheranostic agent by conjugating dextran sulfate (DS) to chlorin e6 (Ce6) and estimated its phototherapeutic feasibility in murine atheroma. Also, the phototherapeutic mechanisms of DS-Ce6 on atherosclerosis were investigated. RESULTS: The phototheranostic agent DS-Ce6 efficiently internalized into the activated macrophages and foam cells via scavenger receptor-A (SR-A) mediated endocytosis. Customized serial optical imaging-guided photoactivation of DS-Ce6 by light illumination reduced both atheroma burden and inflammation in murine models. Immuno-fluorescence and -histochemical analyses revealed that the photoactivation of DS-Ce6 produced a prominent increase in macrophage-associated apoptotic bodies 1 week after laser irradiation and induced autophagy with Mer tyrosine-protein kinase expression as early as day 1, indicative of an enhanced efferocytosis in atheroma. CONCLUSION: Imaging-guided DS-Ce6 photoactivation was able to in vivo detect inflammatory activity in atheroma as well as to simultaneously reduce both plaque burden and inflammation by harmonic contribution of apoptosis, autophagy, and lesional efferocytosis. These results suggest that macrophage targetable phototheranostic nanoagents will be a promising theranostic strategy for high-risk atheroma.


Subject(s)
Atherosclerosis/metabolism , Foam Cells/metabolism , Photosensitizing Agents , Theranostic Nanomedicine/methods , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Endocytosis/drug effects , Infrared Rays , Male , Mice , Mice, Knockout , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , RAW 264.7 Cells
9.
Molecules ; 26(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34684822

ABSTRACT

Cyclopeptidic photosensitizer prodrugs (cPPPs) are compounds designed to specifically target overexpressed hydrolases such as serine proteases, resulting in their specific activation in close proximity to tumor cells. In this study, we explored a series of conjugates that can be selectively activated by the urokinase plasminogen activator (uPA). They differ from each other by their pheophorbide a (Pha) loading, their number of PEG chains and the eventual presence of black hole quenchers (BHQ3). The involvement of a peptidic linker between the drugs and the cyclopeptidic carrier allows specific cleavage by uPA. Restoration of the photophysical activity was observed in vitro on A549 lung and MCF7 breast cancer cells that exhibited an increase in red fluorescence emission up to 5.1-fold and 7.8-fold, respectively for uPA-cPPQ2+2/5. While these cPPP conjugates do not show dark toxicity, they revealed their phototoxic potential in both cell lines at 5 µM of Phaeq and a blue light fluence of 12.7 J/cm2 that resulted in complete cell death with almost all conjugates. This suggests, in addition to the promising use for cancer diagnosis, a use as a PDT agent. Intravenous injection of tetrasubstituted conjugates in fertilized hen eggs bearing a lung cancer nodule (A549) showed that a double PEGylation was favorable for the selective accumulation of the unquenched Pha moieties in the tumor nodules. Indeed, the diPEGylated uPA-cPPP4/52 induced a 5.2-fold increase in fluorescence, while the monoPEGylated uPA-cPPP4/5 or uPA-cPPQ2+2/5 led to a 0.4-fold increase only.


Subject(s)
Chorioallantoic Membrane/metabolism , Photosensitizing Agents/metabolism , Prodrugs/metabolism , A549 Cells , Animals , Biological Transport, Active , Chick Embryo , Drug Delivery Systems/methods , Female , Humans , In Vitro Techniques , MCF-7 Cells , Models, Biological , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacokinetics , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Polyethylene Glycols/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Xenograft Model Antitumor Assays
10.
J Photochem Photobiol B ; 224: 112328, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34628206

ABSTRACT

The race against ovarian cancer continue to motivate the research worldwide. It is known that many antitumor drugs have limited penetration into solid tumor tissues due to its microenvironment, thus contributing to their low efficacy. Therapeutic modalities have been exploited to elicit antitumor effects based on microenvironment of tumor, including Photodynamic therapy (PDT). Prospection of natural small molecules and nanotechnology are important tools in the development of new ways of obtaining photoactive compounds that are biocompatible. The Betulinic acid (BA) has shown potential biological effect as bioactive drug, but it has low water solubility. Thus, in the present study, owing to the poor solubility of the BA, its free form (BAF) was compared to a spray dried microparticle betulinic acid/HP-ß-CD formulation (BAC) aiming to assess the BAF and BAC efficacy as a photosensitizer in PDT for application in ovarian cancer. BAF and BAC were submitted to assays in the presence of LED (λ = 420 nm) under different conditions (2.75 J/cm2, 5.5 J/cm2, and 11 J/cm2) and in absence of irradiation, after 5 min or 4 h of contact with ovarian carcinoma cells (A2780) or fibroblast murine cells (3T3). Furthermore, HPLC-MS/MS and MALDI-MSI methods were developed and validated in plasma and tumor of mice proving suitable for in vivo studies. The results found a greater photoinduced cytotoxic effect for the BAC at low concentration for A2780 when irradiated with LED with similar results for fluorescence microscopy. The results motivate us to continue the studies with the BA as a potential antitumor bioactive compound.


Subject(s)
Ovarian Neoplasms/pathology , Pentacyclic Triterpenes/therapeutic use , Photochemotherapy , Photosensitizing Agents/therapeutic use , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Female , Humans , In Vitro Techniques , Limit of Detection , Mice, Nude , Ovarian Neoplasms/drug therapy , Pentacyclic Triterpenes/blood , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/pharmacokinetics , Photosensitizing Agents/blood , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Reproducibility of Results , Spray Drying , Tandem Mass Spectrometry , Betulinic Acid
11.
Bull Exp Biol Med ; 171(4): 468-471, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34542759

ABSTRACT

The efficacy of a new photosensitizer of chlorin E6 conjugated with a prostate-specific membrane antigen (PSMA) in photodynamic therapy of murine melanoma B16 was studied in in vivo experiments. The dynamics of photosensitizer accumulation in the tumor and surrounding tissues was evaluated and antitumor efficacy of photodynamic therapy was assessed by parameters of regression and morphological characteristics of experimental transplanted melanoma B16. The inhibitory effect of photodynamic therapy on melanoma was evaluated by complete regression of the tumor, absolute tumor growth coefficient in animals with continuation of tumor growth, and the increase in life span in comparison with the control; the criterion of cure was the absence of signs of tumor recurrence in mice within 90 days after therapy. The therapeutic potential of photodynamic therapy was determined by devitalization of tumor cells (histological examination of the zones of laser exposure on day 21 after treatment). The photosensitizer with PSMA-ligand exhibited high antitumor activity in photodynamic therapy for melanoma B16. Photodynamic therapy carried out at the optimum time after photosensitizer injection with experimentally determined parameters of laser exposure allows achieving the maximum inhibitory effect on melanoma. Pathomorphological study in the zones of exposure detected no survived tumor cells.


Subject(s)
Chlorophyllides/therapeutic use , Melanoma, Experimental/drug therapy , Photochemotherapy/methods , Skin Neoplasms/drug therapy , Urea/analogs & derivatives , Animals , Cell Line, Tumor , Chlorophyllides/chemistry , Chlorophyllides/pharmacokinetics , Female , Ligands , Melanoma, Experimental/diagnostic imaging , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Urea/chemistry , Urea/pharmacokinetics , Urea/therapeutic use
12.
Drug Deliv ; 28(1): 1923-1931, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34550040

ABSTRACT

Transdermal drug delivery for local or systemic therapy provides a potential anticancer modality with a high patient compliance. However, the drug delivery efficiency across the skin is highly challenging due to the physiological barriers, which limit the desired therapeutic effects. In this study, we prepared liposome-in-hydrogels containing a tumor targeting photosensitizer IR780 (IR780/lipo/gels) for tumor photothermal therapy (PTT). The formulation effectively delivered IR780 to subcutaneous tumor and deep metastatic sites, while the hydrogels were applied on the skin overlying the tumor or on an area of distant normal skin. The photothermal antitumor activity of topically administered IR780/lipo/gels was evaluated following laser irradiation. We observed significant inhibition of the rate of the tumor growth without any toxicity associated with the topical administration of hydrogels. Collectively, the topical administration of IR780/lipo/gels represents a new noninvasive and safe strategy for targeted tumor PTT.


Subject(s)
Hydrogels/chemistry , Indoles/pharmacology , Liposomes/chemistry , Photosensitizing Agents/pharmacology , Photothermal Therapy/methods , Administration, Cutaneous , Animals , Body Weight , Chemistry, Pharmaceutical , Drug Carriers/chemistry , Indoles/administration & dosage , Indoles/adverse effects , Indoles/pharmacokinetics , Low-Level Light Therapy/adverse effects , Low-Level Light Therapy/methods , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/adverse effects , Photosensitizing Agents/pharmacokinetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Mol Pharm ; 18(11): 4046-4057, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34554752

ABSTRACT

Cutaneous melanoma is one of the most aggressive and metastatic forms of skin cancer. However, current therapeutic options present several limitations, and the annual death rate due to melanoma increases every year. Dermal delivery of nanomedicines can effectively eradicate primary melanoma lesions, avoid the metastatic process, and improve survival. Rose Bengal (RB) is a sono-photosensitizer drug with intrinsic cytotoxicity toward melanoma without external stimuli but the biopharmaceutical profile limits its clinical use. Here, we propose deformable lipid nanovesicles, also known as transfersomes (TF), for the targeted dermal delivery of RB to melanoma lesions to eradicate them in the absence of external stimuli. Considering RB's poor ability to cross the stratum corneum and its photosensitizer nature, transfersomal carriers were selected simultaneously to enhance RB penetration to the deepest skin layers and protect RB from undesired photodegradation. RB-loaded TF dispersion (RB-TF), prepared by a modified reverse-phase evaporation method, were nanosized with a ζ-potential value below -30 mV. The spectrophotometric and fluorimetric analysis revealed that RB efficiently interacted with the lipid phase. The morphological investigations (transmission electron microscopy and small-angle X-ray scattering) proved that RB intercalated within the phospholipid bilayer of TF originating unilamellar and deformable vesicles, in contrast to the rigid multilamellar unloaded ones. Such outcomes agree with the results of the in vitro permeation study, where the lack of a burst RB permeation peak for RB-TF, observed instead for the free drug, suggests that a significant amount of RB interacted with lipid nanovesicles. Also, RB-TF proved to protect RB from undesired photodegradation over 24 h of direct light exposure. The ex vivo epidermis permeation study proved that RB-TF significantly increased RB's amount permeating the epidermis compared to the free drug (78.31 vs 38.31%). Finally, the antiproliferative assays on melanoma cells suggested that RB-TF effectively reduced cell growth compared to free RB at the concentrations tested (25 and 50 µM). RB-TF could potentially increase selectivity toward cancer cells. Considering the outcomes of the characterization and cytotoxicity studies performed on RB-TF, we conclude that RB-TF represents a valid potential alternative tool to fight against primary melanoma lesions via dermal delivery in the absence of light.


Subject(s)
Melanoma/drug therapy , Nanoparticle Drug Delivery System/chemistry , Photosensitizing Agents/administration & dosage , Rose Bengal/administration & dosage , Skin Neoplasms/drug therapy , Administration, Cutaneous , Animals , Cell Line, Tumor , Drug Liberation , Epidermis/metabolism , Epidermis/pathology , Humans , Light , Lipids/chemistry , Melanoma/pathology , Photochemotherapy/methods , Photosensitizing Agents/pharmacokinetics , Rose Bengal/pharmacokinetics , Skin Absorption/radiation effects , Skin Neoplasms/pathology , Swine
14.
Drug Deliv ; 28(1): 1769-1784, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34470548

ABSTRACT

Traditional combined photodynamic and photothermal therapy (PDT/PTT) was limited in clinical treatment of cancer due to the exceptionally low drug delivery efficiency to tumor sites and the activation by laser excitation with different wavelengths. We have accidentally discovered that our synthesized chlorin e6-C-15-ethyl ester (HB, a new type of photosensitizer) be activated by a laser with an excitation wavelength of 660 nm. Herein, we utilized Au nanorods (AuNRs) as 660 nm-activated PTT carriers to be successively surface-functionalized with HB and tumor-targeting peptide cyclic RGD (cRGD) to develop HB-AuNRs@cRGD for single NIR laser-induced targeted PDT/PTT. The HB-AuNRs@cRGD could be preferentially accumulated within tumor sites and rapidly internalized by cancer cells. Thereby, the HB-AuNRs@cRGD could exhibit amplified therapeutic effects by producing both significant reactive oxygen species (ROS) and hyperthermia simultaneously under the guidance of fluorescence imaging. The tumor inhibition rate on ECA109 esophageal cancer model was approximately 77.04%, and the negligible systematic toxicity was observed. This study proposed that HB-AuNRs@cRGD might be a promising strategy for single NIR laser-induced and imaging-guided targeted bimodal phototherapy.


Subject(s)
Gold/chemistry , Nanotubes/chemistry , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photothermal Therapy/methods , Animals , Cell Line , Female , Low-Level Light Therapy , Mice , Mice, Inbred BALB C , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Reactive Oxygen Species/metabolism
15.
ACS Appl Mater Interfaces ; 13(33): 39055-39065, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34433248

ABSTRACT

Polymer photosensitizers (PPSs) with the distinctive properties of good light-harvesting capability, high photostability, and excellent tumor retention effects have aroused great research interest in photodynamic therapy (PDT). However, their potential translation into clinic was often constrained by the hypoxic nature of tumor microenvironment, the aggregation-caused reduced production of reactive oxygen species (ROS), and the tedious procedure of manufacture. As a powerful and versatile strategy, vacancy engineering possesses the unique capability to effectively improve the photogenerated electron efficiency of nanomaterials for high-performance O2 and ROS production. Herein, by introducing vacancy engineering into the design of PPSs for PDT for the first time, we synthesized a novel PPS of Au-decorated polythionine (PTh) nanoconstructs (PTh@Au NCs) with the unique integrated features of distinguished O2 self-evolving function and highly efficient ROS generation for achieving the greatly enhanced PDT efficacy toward hypoxic tumor both in vitro and in vivo. The incorporation of Au into PTh leads to the special PTh-Au heterostructure-induced sulfur vacancies in PTh@Au NCs, which results in an efficient electron-hole separation performance and also plays a key role in a long lifetime of free electrons and holes. Accordingly, an ∼2- to 3-fold ROS generation and an ∼1.5-fold increase of O2 self-supply than the pure PTh nanoparticles (NPs) were obtained even under hypoxic conditions upon exposure to 650 nm light. By combining such superior ROS generation and O2 self-supply performances with the outstanding cellular internalization and tumor accumulation capacities, an advanced antitumor effect with the achievement of almost complete hypoxic tumor elimination in vivo or 88% cell destruction in vitro was acquired by the PTh@Au NCs. In addition, the distinctive facile one-step redox strategy for PTh@Au NCs synthesis compared to the reported PPSs for PDT also makes it beneficial for potential practical application. The first introduction of vacancy engineering concept into PPSs in the field of PDT proposed in this work offers a new strategy for the development and design highly efficient PPSs for PDT applications.


Subject(s)
Antineoplastic Agents/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Phenothiazines/chemistry , Photosensitizing Agents/chemistry , Polymers/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Membrane Permeability , Female , Humans , Hypoxia/metabolism , Mice , Neoplasms, Experimental , Oxygen/metabolism , Phenothiazines/pharmacokinetics , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Polymers/pharmacokinetics , Reactive Oxygen Species/metabolism , Theranostic Nanomedicine , Tumor Microenvironment
16.
Macromol Biosci ; 21(12): e2100229, 2021 12.
Article in English | MEDLINE | ID: mdl-34390189

ABSTRACT

The integration of hypoxia-activated chemotherapy with photodynamic therapy (PDT) has newly become a potent strategy for tumor treatment. Herein, a reactive oxygen species (ROS)-responsive drug carriers (PS@AQ4N/mPEG-b-PSe NPs) are fabricated based on the amphiphilic selenium-containing methoxy poly(ethylene glycol)-polycarbonate (mPEG-b-PSe), the hydrophobic photosensitizer (PS), and hypoxia-activated prodrug Banoxantrone (AQ4N). The obtained nanoparticles are spherical with an average diameter of 100 nm as characterized by transmission electron microscope (TEM) and dynamic laser scattering (DLS) respectively. The encapsulation efficiency of the PS and AQ4N reaches 92.83% and 51.04% at different conditions, respectively, by UV-vis spectrophotometer. It is found that the drug release is accelerated due to the good ROS responsiveness of mPEG-b-PSe and the cumulative release of AQ4N is up to 89% within 30 h. The cell test demonstrates that the nanoparticles dissociate when triggered by the ROS stimuli in the cancer cells, thus the PS is exposed to more oxygen and the ROS generation efficiency is enhanced accordingly. The consumption of oxygen during PDT leads to the increased tumor hypoxia, and subsequently activates AQ4N into cytotoxic counterpart to inhibit tumor growth. Therefore, the synergistic therapeutic efficacy demonstrates this drug delivery has great potential for antitumor therapy.


Subject(s)
Drug Carriers , Nanoparticles , Photochemotherapy , Photosensitizing Agents , Prodrugs , Reactive Oxygen Species/metabolism , Selenium , Cell Line, Tumor , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Prodrugs/chemical synthesis , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Selenium/chemistry , Selenium/pharmacokinetics , Selenium/pharmacology
17.
ACS Appl Mater Interfaces ; 13(33): 39934-39948, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34396771

ABSTRACT

There are two severe obstacles in cancer immunotherapy. The first is that the low response rate challenges the immune response owing to the immunosuppressive tumor microenvironment (ITM) and poor immunogenicity of the tumor. The second obstacle is that the dense and intricate pathophysiology barrier seriously restricts deep drug delivery in solid tumors. A laser/glutathione (GSH)-activatable nanosystem with tumor penetration for achieving highly efficient immunotherapy is reported. The core of the nanosystem was synthesized by coordinating zinc ions with GSH-activatable oxaliplatin (OXA) prodrugs and carboxylated phthalocyanine. Such an OXA/phthalocyanine-based coordination polymer nanoparticle (OPCPN) was wrapped by a phospholipid bilayer and NTKPEG. NTKPEG is a PEGylated indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor prodrug containing a thioketal (TK) linker, which was modified on the OPCPN (OPCPN@NTKPEG). Upon the laser irradiation tumor site, ROS production of the OPCPN@NTKPEG triggers cleavage of NTKPEG by degradation of TK for promoted tumor penetration and uptake. OXA, phthalocyanine, and IDO1 inhibitor were released by the intracellular high-level GSH. OXA inhibits cell growth and is combined with photodynamic therapy (PDT) to induce immunogenic cell death (ICD). The IDO1 inhibitor reversed the ITM by suppressing IDO1-mediated Trp degradation and exhaustion of cytotoxic T cells. Laser/GSH-activatable drug delivery was more conducive to enhancing ICD and reversing ITM in deep tumors. Chemo-PDT with OPCPN@NTKPEG significantly regressed tumor growth and reduced metastasis by improved cancer immunotherapy.


Subject(s)
Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Glutathione/chemistry , Indoles/chemistry , Nanoparticles/chemistry , Oxaliplatin/chemistry , Photosensitizing Agents/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation/drug effects , Combined Modality Therapy , Drug Liberation , Glutathione/metabolism , Humans , Immunogenic Cell Death/drug effects , Immunogenic Cell Death/radiation effects , Immunotherapy , Indoles/pharmacokinetics , Isoindoles , Lasers , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/radiotherapy , Oxaliplatin/pharmacokinetics , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Polyethylene Glycols/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Tissue Distribution , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects
18.
Molecules ; 26(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209713

ABSTRACT

Antiviral action of various photosensitizers is already summarized in several comprehensive reviews, and various mechanisms have been proposed for it. However, a critical consideration of the matter of the area is complicated, since the exact mechanisms are very difficult to explore and clarify, and most publications are of an empirical and "phenomenological" nature, reporting a dependence of the antiviral action on illumination, or a correlation of activity with the photophysical properties of the substances. Of particular interest is substance-assisted photogeneration of highly reactive singlet oxygen (1O2). The damaging action of 1O2 on the lipids of the viral envelope can probably lead to a loss of the ability of the lipid bilayer of enveloped viruses to fuse with the lipid membrane of the host cell. Thus, lipid bilayer-affine 1O2 photosensitizers have prospects as broad-spectrum antivirals against enveloped viruses. In this short review, we want to point out the main types of antiviral photosensitizers with potential affinity to the lipid bilayer and summarize the data on new compounds over the past three years. Further understanding of the data in the field will spur a targeted search for substances with antiviral activity against enveloped viruses among photosensitizers able to bind to the lipid membranes.


Subject(s)
Antiviral Agents , Membrane Lipids/metabolism , Photosensitizing Agents , Viral Envelope/metabolism , Virus Diseases , Viruses/metabolism , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Singlet Oxygen , Virus Diseases/drug therapy , Virus Diseases/metabolism
19.
ACS Appl Mater Interfaces ; 13(26): 30359-30372, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34142813

ABSTRACT

One of the recent advances in nanotechnology within the medical field is the development of a nanoformulation of anticancer drugs or photosensitizers. Cancer cell-specific drug delivery and upregulation of the endogenous level of reactive oxygen species (ROS) are important in precision anticancer treatment. Within our article, we report a new therapeutic nanoformulation of cancer cell targeting using endogenous ROS self-generation without an external initiator and a switch-on drug release (ROS-induced cascade nanoparticle degradation and anticancer drug generation). We found a substantial cellular ROS generation by treating an isothiocyanate-containing chemical and functionalizing it onto the surface of porous silicon nanoparticles (pSiNPs) that are biodegradable and ROS-responsive nanocarriers. Simultaneously, we loaded an ROS-responsive prodrug (JS-11) that could be converted to the original anticancer drug, SN-38, and conducted further surface functionalization with a cancer-targeting peptide, CGKRK. We demonstrated the feasibility as a cancer-targeting and self-activating therapeutic nanoparticle in a pancreatic cancer xenograft mouse model, and it showed a superior therapeutic efficacy through ROS-induced therapy and drug-induced cell death. The work presented is a new concept of a nanotherapeutic and provides a more feasible clinical translational pathway.


Subject(s)
Antineoplastic Agents/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Liberation , Female , Humans , Irinotecan/pharmacokinetics , Irinotecan/therapeutic use , Isothiocyanates/chemistry , Isothiocyanates/pharmacokinetics , Male , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Photosensitizing Agents/pharmacokinetics , Precision Medicine , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Silanes/chemistry , Silanes/pharmacokinetics , Silicon/chemistry , Silicon/pharmacokinetics , Xenograft Model Antitumor Assays
20.
BMC Cancer ; 21(1): 547, 2021 May 13.
Article in English | MEDLINE | ID: mdl-33985453

ABSTRACT

BACKGROUND: Photodynamic therapy (PDT) is an anticancer treatment that utilizes the interaction of light and a photosensitiser (PS), promoting tumour cell death mediated by generation of reactive oxygen species. In this study, we evaluated the in vitro photoactivity of four meso-substituted porphyrins and a porphyrin coupled to a fullerene. METHODS: The cell line employed was the LM3 mammary adenocarcinoma, and the PS with the best photokilling activity was administered to mice bearing the LM3 subcutaneously implanted adenocarcinoma. The TEMCP4+ porphyrin and its analogue TEMCC4+ chlorine contain four identical carbazoyl substituents at the meso positions of the tetrapyrrolic macrocycle and have A4 symmetry. The TAPP derivative also has A4 symmetry, and it is substituted at the meso positions by aminopropoxy groups. The DAPP molecule has ABAB symmetry with aminopropoxy and the trifluoromethyl substituents in trans positions. The TCP-C604+ dyad is formed by a porphyrin unit covalently attached to the fullerene C60. RESULTS: The PSs are taken up by the cells with the following efficiency: TAPP> TEMCP4+ = TEMCC4+ > DAPP >TCP-C604+, and the amount of intracellular PS correlates fairly with the photodamage degree, but also the quantum yields of singlet oxygen influence the PDT outcome. TAPP, DAPP, TEMCC4+ and TEMCP4+ exhibit high photoactivity against LM3 mammary carcinoma cells, being TAPP the most active. After topical application of TAPP on the skin of mice bearing LM3 tumours, the molecule is localized mainly in the stratum corneum, and at a lower extent in hair follicles and sebaceous glands. Systemic administration of TAPP produces a tumour: normal skin ratio of 31.4, and high accumulation in intestine and lung. CONCLUSION: The results suggest a potential use of topical TAPP for the treatment of actinic keratosis and skin adnexal neoplasms. In addition, selectivity for tumour tissue after systemic administration highlights the selectivity of and potentiality of TAPP as a new PS.


Subject(s)
Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Skin Neoplasms/drug therapy , Animals , Male , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Photosensitizing Agents/pharmacokinetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...