Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Complementary Medicines
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Arch Dermatol Res ; 313(3): 173-180, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32449013

ABSTRACT

The treatment of acne remains a challenge for dermatologists. A variety of conventional therapies are available for acne treatment such as topical and systemic medications. Although many of these traditional acne treatments are effective, the wide-spread nature of the disease and its sometimes resistant nature delineate the need for alternative therapies. Therefore, over the past decade, phototherapy has been introduced for the treatment of acne, such as pulsed dye lasers (PDLs) and photodynamic therapy (PDT). The aim of this study was to compare the safety and efficacy of PDL and methylene blue-mediated photodynamic therapy (MB-PDT) in the treatment of mild to moderate acne. Split-face clinical trial including fifteen patients presenting with mild to moderate acne were treated with 585 nm PDL on the right side of the face and MB-PDT with 665-nm diode laser on the left side. The photosensitizer MB was prepared in nanoemulgel formulation, and the treatment was carried out for three sessions maximum at 2-weeks intervals. Results revealed that both PDL and MB-PDT were effective therapies in the treatment of acne, as manifested by the reduction of inflammatory and non-inflammatory lesions throughout the treatment period. However, the latter therapy was proven more potent in the reduction of acne severity, and in terms of patients' tolerance. Therefore, it can be concluded that MB in the nanoemulgel form is a promising treatment approach for acne, and can be further experimented in the treatment of other dermatological diseases.


Subject(s)
Acne Vulgaris/therapy , Lasers, Dye/adverse effects , Methylene Blue/administration & dosage , Photochemotherapy/adverse effects , Photosensitizing Agents/administration & dosage , Acne Vulgaris/diagnosis , Adolescent , Adult , Face , Female , Humans , Male , Methylene Blue/adverse effects , Methylene Blue/pharmacokinetics , Nanogels/administration & dosage , Photochemotherapy/methods , Photosensitizing Agents/adverse effects , Photosensitizing Agents/pharmacokinetics , Prospective Studies , Severity of Illness Index , Skin/drug effects , Skin/radiation effects , Treatment Outcome , Young Adult
8.
J Mater Chem B ; 8(32): 7149-7159, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32617545

ABSTRACT

Photodynamic therapy (PDT) has been demonstrated to be a promising strategy for the treatment of cancer, while its therapeutic efficacy is often compromised due to excessive concentrations of glutathione (GSH) as a reactive oxygen species (ROS) scavenger in cancer cells. Herein, we report the development of near-infrared (NIR) photothermal liposomal nanoantagonists (PLNAs) for amplified PDT through through the reduction of intracellular GSH biosynthesis. Such PLNAs were constructed via encapsulating a photosensitizer, indocyanine green (ICG) and a GSH synthesis antagonist, l-buthionine sulfoximine (BSO) into a thermal responsive liposome. Under NIR laser irradiation at 808 nm, PLNAs generate mild heat via a ICG-mediated photothermal conversion effect, which leads to the destruction of thermal responsive liposomes for a controlled release of BSO in a tumor microenvironment, ultimately reducing GSH levels. This amplifies intracellular oxidative stresses and thus synergizes with PDT to afford an enhanced therapeutic efficacy. Both in vitro and in vivo data verify that PLNA-mediated phototherapy has an at least 2-fold higher efficacy in killing cancer cells and inhibiting tumor growth compared to sole PDT. This study thus demonstrates a NIR photothermal drug delivery nanosystem for amplified photomedicine.


Subject(s)
Antineoplastic Agents/chemistry , Buthionine Sulfoximine/chemistry , Enzyme Inhibitors/chemistry , Glutathione/antagonists & inhibitors , Indocyanine Green/chemistry , Liposomes/chemistry , Photosensitizing Agents/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols , Buthionine Sulfoximine/pharmacokinetics , Drug Liberation , Enzyme Inhibitors/pharmacokinetics , Humans , Hyperthermia, Induced , Indocyanine Green/pharmacokinetics , Infrared Rays , Mice , Neoplasms, Experimental , Oxidation-Reduction , Oxidative Stress/drug effects , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Reactive Oxygen Species/metabolism
9.
Photodiagnosis Photodyn Ther ; 30: 101801, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32360854

ABSTRACT

Nasopharyngeal carcinoma (NPC) is of high prevalence in Hong Kong and southern China. The pathogenesis of NPC is closely associated with Epstein-Barr virus (EBV) infection via regulation of viral oncoprotein latent membrane protein 1 (LMP1). The conventional treatment for NPC is chemo-radiotherapy, but the prognosis remains poor for advanced stage, recurrent and metastatic NPC. Photodynamic therapy (PDT) is a therapeutic approach to combat tumors. PDT effectiveness depends on the interaction of photosensitizers, light and molecular oxygen. 5- aminolevulinic acid hexyl derivative (H-ALA) is one of the photosensitizers derived from 5-ALA. H-ALA with improved lipophilic properties by adding a long lipophilic chain (hexyl group) to 5-ALA, resulted in better penetration into cell cytoplasm. In this study, the effect of H-ALA-PDT on NPC cells (EBV positive C666-1 and EBV negative CNE2) was investigated. The H-ALA mediated cellular uptake and cytotoxicity was revealed via flow cytometry analysis and MTT assay respectively. H-ALA PDT mediated protein modulation was analysed by western blot analysis. Our finding reported that the cellular uptake of H-ALA in C666-1 and CNE2 cells was in a time dependent manner. H-ALA PDT was effective to C666-1 and CNE2 cells. EBV LMP1 proteins was expressed in C666-1 cells only and its expression was responsive to H-ALA PDT in a dose dependent manner. This work revealed the potential of H-ALA PDT as a treatment regiment for EBV positive NPC cells. Understanding the mechanism of H-ALA mediated PDT could develop improved strategies for the treatment of NPC.


Subject(s)
Aminolevulinic Acid/analogs & derivatives , Low-Level Light Therapy/methods , Nasopharyngeal Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Viral Matrix Proteins/drug effects , Aminolevulinic Acid/pharmacokinetics , Aminolevulinic Acid/pharmacology , Cell Line, Tumor , Cell Survival , Herpesvirus 4, Human/drug effects , Humans , Lasers, Semiconductor/therapeutic use , Photosensitizing Agents/pharmacokinetics , Protoporphyrins/pharmacokinetics
10.
Mater Sci Eng C Mater Biol Appl ; 110: 110638, 2020 May.
Article in English | MEDLINE | ID: mdl-32204072

ABSTRACT

Glioblastoma multiforme (GBM) is an extremely aggressive malignant brain tumor. Despite advances in treatment modalities, it remains largely incurable. This unfavorable prognosis for GBM is at least partly due to the lack of a successful drug delivery system across the blood-brain barrier (BBB). The delivery of drugs through nanomedicines combined with less invasive alternative therapies represents an important hope for the future of these incurable brain tumors. Whey protein nanocarriers represent promising strategy for targeted drug delivery to tumor cells by enhancing the drug's bioavailability and distribution, and reducing the body's response towards drug resistance. They have been extensively studied to find new alternatives for capacity to encapsulate different drugs and no need for cross-linkers. In this study, we report for the first time the incorporation and administration of Aluminum phthalocyanine chloride (AlClPc)-loaded whey protein drug delivery system (AlClPc-PDDS) for the treatment of glioblastoma brain cancer. This system was designed and optimized (with the use of the spray drying technique) to obtain the required particle size (in the range of 100 to 300 nm), zeta potential and drug loading. Our results suggest that we have developed a drug delivery system from a low-cost raw material and preparation method that is capable of incorporating hydrophobic drugs which, in combination with irradiation, cause photodamage to neoplasic cells, working as an effective adjuvant treatment for malignant glioma.


Subject(s)
Brain Neoplasms/drug therapy , Drug Carriers , Glioblastoma/drug therapy , Nanoparticles , Photosensitizing Agents , Whey Proteins , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/pharmacology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Whey Proteins/chemistry , Whey Proteins/pharmacokinetics , Whey Proteins/pharmacology
11.
Curr Drug Discov Technol ; 17(5): 661-669, 2020.
Article in English | MEDLINE | ID: mdl-30973109

ABSTRACT

BACKGROUND: Detailed photochemical and photocytotoxicity studies of two new porphyrins: 5,10,15,20-meso-tetrakis-(4-acetoxy-3-methoxyphenyl) porphyrin (P2.1) and 5-(4-hydroxy-3- methoxyphenyl)-10,15,20-tris-(4-acetoxy-3-methoxyphenyl)porphyrin (P2.2) are reported, as potential candidates for theranostics. For powdered samples of P2.1 and P2.2 adsorbed onto a powdered biocompatible substrate, polyethylene glycol (PEG), a concentration study was performed, correlating the fluorescence emission intensity with sample absorption to determine the useful concentration range for photodynamic therapy of cancer (PDT) in which aggregation does not occur. Cytotoxicity studies were performed in dark and illuminated conditions. METHODS: The laser induced luminescence set-up is home-made, a N2 laser is used as the excitation source and a time gated charged-coupled device (ICCD) as the detector. Fluorescence lifetime determinations were made using pulsed light sources from the excitation LEDs and measures of the fluorescence intensities at different time delays after the excitation pulse. The singlet oxygen formation quantum yields ΦΔ measurements were obtained by comparing the total area of the emission spectra for the reference compound and also for the samples under study in the same solvent and with the same optical density at the excitation wavelength (405 nm). An integrating sphere for relative and absolute measurements was used in this work as an alternative methodology to obtain the values for the fluorescence emission quantum yields (ΦF) of the adsorbed porphyrin under study. The cytotoxicity evaluation was made in the dark and under irradiation, using four different human tumor cell lines and one non-tumor primary cell culture. RESULTS: In order to establish the useful range of concentrations of the sensitizer for PDT, and due to the use of powdered samples, a special methodology was needed: the variations of the fluorescence lifetimes and fluorescence quantum yields were evaluated as a function of the concentration of the dye, measured by (1-R)*fdye. Both ΦF and τF are constant in the range from 0.002 to about 0.050 µmol g-1, and only after that a concentration quenching effect becomes visible, decreasing both ΦF and τF. This methodology is based in the correlations established between the Remission Function values and ΦF and τF obtained for increasing values of the sensitizer concentrations. CONCLUSIONS: The study of the aggregation effects of P2.1 and P2.2 porphyrins into a PEG matrix allowed us to determine the usable concentration range for photodynamic therapy use, where the aggregation of porphyrins decreases, therefore reducing the PDT action. The use of an integrating sphere for relative and absolute measurements of fluorescence quantum yields and also the lifetime studies as a function of the dye loading confirms the useful range for the use of P2.1 and P2.2 in PEG as powdered samples. The determination of the GI50, the porphyrin concentration which inhibits 50% of the cell growth, evidences that P2.2, the A3B porphyrin overtakes P2.1 (the A4 porphyrin) in terms of PDT efficiency and both porphyrins are much better PDT agents than the unsubstituted porphyrin, TPP. These data clearly show that porphyrins P2.2 and P2.1 exhibit an excellent behaviour in terms of its photocytotoxicity. These results encourage us to pursuit in the study of this family of porphyrins in which a balance of hydrophobic versus hydrophilic substituents in the phenyl group was achieved.


Subject(s)
Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Carriers/chemistry , Drug Evaluation, Preclinical , Humans , Lasers, Gas , Nanoparticles/chemistry , Neoplasms/pathology , Photochemotherapy/instrumentation , Photosensitizing Agents/pharmacokinetics , Polyethylene Glycols/chemistry , Porphyrins/pharmacokinetics , Theranostic Nanomedicine/instrumentation , Theranostic Nanomedicine/methods , Tissue Distribution/radiation effects
12.
Gastric Cancer ; 23(1): 82-94, 2020 01.
Article in English | MEDLINE | ID: mdl-31302791

ABSTRACT

BACKGROUND: Near-infrared photoimmunotherapy (NIR-PIT) constitutes a new class of molecular-targeted theranostics utilizing monoclonal antibody (mAb)-photosensitizer conjugates and NIR light. In this study, we developed a new type of NIR-PIT targeting vascular endothelial growth factor receptor 2 (VEGFR-2) expressed on vascular endothelium in an experimental gastric cancer model and evaluated the feasibility by comparing conventional NIR-PIT targeting cancer cell membrane in vitro and in vivo. METHODS: HER2-positive human gastric cancer cells, NCI-N87, were used for the experiments. Anti-HER2 mAb, trastuzumab and anti-VEGFR-2 mAb, DC101 were conjugated to photosensitizer, IR700. Phototoxicity in response to NIR-PIT were investigated in vitro and in vivo. Microvessel densities, as an indicator of angiogenesis, were counted in harvested xenografts after NIR-PIT to elucidate the mechanism. RESULTS: DC101-IR700 did not induce phototoxic effect in vitro because of the absence of expression of VEGFR-2 in NCI-N87 cancer cells. However, it induced an antitumor effect in NCI-N87 xenograft tumors accompanied with damage in tumor neovasculature as determined by decreasing tumor microvessel density, which represents a different mechanism than that of conventional NIR-PIT targeting antigens expressed on the tumor cell membrane. CONCLUSION: We demonstrated a new approach of NIR-PIT utilizing a target on vascular endothelium, such as VEGFR-2, and this treatment might lead to the development of a new therapeutic strategy for human gastric cancer.


Subject(s)
Immunotherapy/methods , Phototherapy/methods , Stomach Neoplasms/therapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Female , Humans , Mice, Inbred BALB C , Microvessels/drug effects , Microvessels/pathology , Molecular Targeted Therapy , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Receptor, ErbB-2/metabolism , Stomach Neoplasms/blood supply , Stomach Neoplasms/pathology , Tissue Distribution , Trastuzumab/pharmacokinetics , Trastuzumab/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
13.
Macromol Biosci ; 20(2): e1900301, 2020 02.
Article in English | MEDLINE | ID: mdl-31762196

ABSTRACT

In this work, dual-mode antibacterial conjugated polymer nanoparticles (DMCPNs) combined with photothermal therapy (PTT) and photodynamic therapy (PDT) are designed and explored for efficient killing of ampicillin-resistant Escherichia coli (Ampr E. coli). The DMCPNs are self-assembled into nanoparticles with a size of 50.4 ± 0.6 nm by co-precipitation method using the photothermal agent poly(diketopyrrolopyrrole-thienothiophene) (PDPPTT) and the photosensitizer poly[2-methoxy-5-((2-ethylhexyl)oxy)-p-phenylenevinylene] (MEH-PPV) in the presence of poly(styrene-co-maleic anhydride) which makes nanoparticles disperse well in water via hydrophobic interactions. Thus, DMCPNs simultaneously possess photothermal effect and the ability of sensitizing oxygen in the surrounding to generate reactive oxygen species upon the illumination of light, which could easily damage resistant bacteria. Under combined irradiation of near-infrared light (550 mW cm-2 , 5 min) and white light (65 mW cm-2 , 5 min), DMCPNs with a concentration of 9.6 × 10-4 µm could reach a 93% inhibition rate against Ampr E. coli, which is higher than the efficiency treated by PTT or PDT alone. The dual-mode nanoparticles provide potential for treating pathogenic infections induced by resistant microorganisms in clinic.


Subject(s)
Anti-Bacterial Agents , Escherichia coli/growth & development , Hyperthermia, Induced , Nanoparticles/chemistry , Photochemotherapy , Photosensitizing Agents , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics
14.
ACS Nano ; 13(11): 12638-12652, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31625721

ABSTRACT

Photodynamic therapy (PDT) is a clinical cancer treatment modality based on the induction of therapeutic reactive oxygen species (ROS), which can trigger immunogenic cell death (ICD). With the aim of simultaneously improving both PDT-mediated intracellular ROS production and ICD levels, we designed a serum albumin (SA)-coated boehmite ("B"; aluminum hydroxide oxide) organic-inorganic scaffold that could be loaded with chlorin e6 (Ce6), a photosensitizer, and a honey bee venom melittin (MLT) peptide, denoted Ce6/MLT@SAB. Ce6/MLT@SAB was anchored by a boehmite nanorod structure and exhibited particle size of approximately 180 nm. Ce6/MLT@SAB could significantly reduce hemolysis relative to that of free MLT, while providing MLT-enhanced PDT antitumor effects in vitro. Compared with Ce6@SAB, Ce6/MLT@SAB improved Ce6 penetration of cancer cells both in vitro and in vivo, thereby providing enhanced intracellular ROS generation with 660 nm light treatment. Following phototreatment, Ce6/MLT@SAB-treated cells displayed significantly improved levels of ICD and abilities to activate dendritic cells. In the absence of laser irradiation, multidose injection of Ce6/MLT@SAB could delay the growth of subcutaneous murine tumors by more than 60%, compared to controls. When combined with laser irradiation, a single injection and phototreatment with Ce6/MLT@SAB eradicated one-third of subcutaneous tumors in treated mice. The addition of an immune checkpoint blockade to Ce6/MLT@SAB phototreatment further augmented antitumor effects, generating increased numbers of CD4+ and CD8+ T cells in tumors with concomitant reduction of myeloid-derived suppressor cells.


Subject(s)
Antineoplastic Agents , Immunotherapy/methods , Melitten , Nanoparticles/chemistry , Photochemotherapy/methods , Photosensitizing Agents , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Female , Immunogenic Cell Death/drug effects , Melitten/chemistry , Melitten/pharmacokinetics , Melitten/pharmacology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology
15.
Theranostics ; 9(17): 4971-4981, 2019.
Article in English | MEDLINE | ID: mdl-31410195

ABSTRACT

The strategy that combines photodynamic therapy (PDT) and photothermal therapy (PTT) is widely used to achieve strong antitumor efficiency. Since light in the NIR-II window possesses ideal penetration ability, developing NIR-II PTT and NIR-II light triggered photosensitizer release for combined PDT and PTT is very promising in nanomedicine. Methods: We develop a novel nanocarrier (termed AuHNRs-DTPP) by conjugating photosensitizer contained chimeric peptide (DTPP) to Au hollow nanorods (AuHNRs). AuHNRs was obtained by a Te-templated method with the assistance of L-cysteine. The chimeric peptide PpIX-PEG8-GGK(TPP)GRDEVDGC (DTPP) was obtained through a solid-phase peptide synthesis (SPPS) method. Results: Under the 1064 nm laser irradiation, the nanocarrier can accumulate heat quickly for efficient PTT, and then release activated photosensitizer for real-time apoptosis imaging. Thereafter, supplementary PDT can be conducted to kill tumor cells survived from the PTT, and meanwhile the normal tissue can be protected from photo-toxicity. Conclusion: This designed AuHNRs-DTPP nanocarrier with remarkable therapy effect, real-time apoptosis imaging ability and reduced skin damage is of great potential in nanomedicine application.


Subject(s)
Apoptosis , Nanotubes/chemistry , Neoplasms, Experimental/therapy , Peptides/administration & dosage , Photochemotherapy/methods , Theranostic Nanomedicine/methods , Animals , Drug Liberation , Female , Gold/chemistry , HeLa Cells , Humans , Hyperthermia, Induced/methods , Infrared Rays/therapeutic use , Mice , Mice, Nude , Neoplasms, Experimental/diagnostic imaging , Peptides/chemistry , Peptides/pharmacokinetics , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Phototherapy/methods
16.
Nanoscale ; 11(12): 5474-5488, 2019 Mar 21.
Article in English | MEDLINE | ID: mdl-30855625

ABSTRACT

Clinical chemotherapy for cancer is limited by the physiological barrier of tumors, resulting in low drug delivery to tumors, poor efficacy of drugs and inability to block tumor metastasis. Here we developed an intelligent switchable nitric oxide (NO)-releasing nanoparticle, IPH-NO, which loads a photosensitizer (IR780) and the chemotherapy drug paclitaxel (PTX) into NO donor-S-nitrosated human serum albumin (HSA-NO). NO exhibits two effects based on its concentration: enhancement of chemotherapy by increasing the enhanced permeability and retention (EPR) effect at low concentrations and direct killing of cancer cells at high concentrations. IPH-NO can slowly release NO in the presence of glutathione to boost tumor vascular permeability and improve drug accumulation. Near-infrared light irradiation was utilized to induce a quick release of NO that can directly kill cancer cells at high concentrations. This combination of phototherapy and NO gas therapy activated by NIR together with chemotherapy showed significant effects in tumor inhibition. Furthermore, IPH-NO blocked tumor metastasis by inhibiting epithelial mesenchymal transition. PH-NO provides a novel strategy to control NO release at tumor site for drug accumulation and combination therapies, consequently potentiating the anticancer efficacy and inhibiting tumor metastasis.


Subject(s)
Nanomedicine , Nanoparticles/chemistry , Nitric Oxide/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Glutathione/chemistry , Half-Life , Humans , Hypothermia, Induced , Infrared Rays , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/pathology , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Serum Albumin, Human/chemistry , Singlet Oxygen/analysis
17.
ACS Appl Mater Interfaces ; 11(7): 6777-6788, 2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30668088

ABSTRACT

Recently, rodlike nanomaterials with specific aspect ratio for efficient cellular uptake have received enormous attention. For functional nanomaterials, such as photothermal agents, large surface areas for their rod-shaped exterior that increase the amount of light absorbed would lead to a higher absorption coefficient as well as drug-loading property. In this project, we coated rodlike mesoporous silica with gold nanoshells (MSNR@Au hybrid), modifying them with ultrasmall gadolinium (Gd)-chelated supramolecular photosensitizers, TPPS4 (MSNR@Au-TPPS4(Gd)), which could be applied to near-infrared fluorescence/multispectral optoacoustic tomography/computed tomography/magnetic resonance imaging and imaging-guided remotely controlled photothermal (PTT)/photodynamic (PDT) combined antitumor therapy. Gold nanoshells, as a perfect PTT agent, were used to assemble the rodlike mesoporous silica nanoparticles with larger superficial area and higher drug loading, thus obtaining the MSNR@Au hybrid. HS-ß-CD, which was used as the host, was adsorbed on the gold nanoshell (MSNR@Au-ß-CD) to link TPPS4(Gd) through the host-guest reaction, thus forming CD-TPPS4 supramolecular photosensitizers (supraPSs). Compared with conventional PSs, supraPSs have host screens, which could reduce the self-aggregation of TPPS4, and consequently generate 1O2 with high efficiency. The in vivo quadmodal imaging of MSNR@Au-TPPS4(Gd) nanoparticles revealed an intensive tumor uptake effect after injection. The in vivo antitumor efficacy further testified that the synergistic therapy, which was more efficient than any other monotherapy, exhibited an excellent tumor inhibition therapeutic effect. As a result, this encourages to further explore multifunctional theranostic nanoparticles based on gold shells for combined cancer therapy.


Subject(s)
Contrast Media , Gold , Hyperthermia, Induced/methods , Nanoshells , Neoplasms, Experimental , Photochemotherapy , Photosensitizing Agents , Silicon Dioxide , Tomography, X-Ray Computed , Animals , Cell Line, Tumor , Contrast Media/chemistry , Contrast Media/pharmacokinetics , Contrast Media/pharmacology , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Humans , Mice, Inbred BALB C , Mice, Nude , Nanoshells/chemistry , Nanoshells/therapeutic use , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacokinetics , Silicon Dioxide/pharmacology , Theranostic Nanomedicine/methods
18.
Small ; 15(3): e1803926, 2019 01.
Article in English | MEDLINE | ID: mdl-30488638

ABSTRACT

Controlled drug release systems can enhance the safety and availability but avoid the side effect of drugs. Herein, the concept of DNA complementary base pairing rules in biology is used to design and prepare a photothermal-triggered drug release system. Adenine (A) modified polydopamine nanoparticles (A-PDA, photothermal reagent) can effectively bind with thymine (T) modified Zinc phthalocyanine (T-ZnPc, photosensitizer) forming A-PDA = T-ZnPc (PATP) complex based on A = T complementary base pairing rules. Similar to DNA, whose base pairing in double strands will break by heating, T-ZnPc can be effectively released from A-PDA after near infrared irradiation-triggered light-thermal conversion to obtain satisfactory photodynamic-photothermal synergistic tumor treatment. In addition, PDA can carry abundant Gd3+ to provide magnetic resonance imaging guided delivery and theranostic function.


Subject(s)
Base Pairing/physiology , Delayed-Action Preparations , Drug Delivery Systems/methods , Drug Liberation , Hyperthermia, Induced/methods , Neoplasms/therapy , Photochemotherapy/methods , Adenine/chemistry , Animals , Cell Line, Tumor , Combined Modality Therapy , DNA, Complementary/chemistry , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Liberation/genetics , Drug Synergism , Female , Humans , Indoles/chemistry , Isoindoles , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Organometallic Compounds/chemistry , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Phototherapy/methods , Polymers/chemistry , Xenograft Model Antitumor Assays , Zinc Compounds
19.
Photodiagnosis Photodyn Ther ; 25: 87-91, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30447412

ABSTRACT

INTRODUCTION: Herpes simplex virus (HSV) is among the most common viruses in humans. HSV1 is often responsible for oral and perioral herpetic lesions. Photodynamic therapy (PDT) is a novel antimicrobial modality that involves the use of laser and a photosensitizer with a specific wavelength. This study aimed to assess and compare the effect of PDT with 810 and 940 nm diode laser and indocyanine green (ICG) photosensitizer on HSV1. METHODS: In this in vitro study, HSV1 isolated from herpes labialis and there were 6 experimental groups.The irradiation parameters were the same for all groups. Number of remaining viruses per milliliter in each group was determined using real-time polymerase chain reaction (PCR) and statistically analyzed by ANOVA. RESULTS: The virus count in all groups significantly decreased compared to the control group (P < 0.05) except in group ICG- without irradiation (P > 0.05). Comparison of groups 810- and 940- (use of each laser alone) with groups 810+ and 940+ (use of each laser plus ICG) revealed that reduction in virus count in groups 810+ and 940+ was significantly greater than that in groups 810- and 940-. CONCLUSION: 810 nm diode laser irradiation and ICG causes the greatest reduction in number of HSV1 compared to all the other groups. ICG without laser irradiation has not significant efficacy on reduction of virus count.


Subject(s)
Herpesvirus 1, Human/drug effects , Indocyanine Green/pharmacology , Lasers, Semiconductor/therapeutic use , Low-Level Light Therapy/methods , Photochemotherapy/methods , Photosensitizing Agents/pharmacokinetics , Humans
20.
ACS Appl Mater Interfaces ; 11(1): 417-429, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30537815

ABSTRACT

Environmental stimuli, including pH, light, and temperature, have been utilized for activating controlled drug delivery to achieve efficient antitumor therapeutics while minimizing undesirable side effects. In this study, a multifunctional nanoplatform based on hollow mesoporous copper sulfide nanoparticles (H-CuS NPs) was developed by loading the interior cavity of the NPs with a drug-loaded phase-change material (PCM, 1-tetradecanol). Doxorubicin (DOX) and chlorin e6 (Ce6) were selected as the model chemotherapeutic drug and photosensitizer, respectively, which were encapsulated in H-CuS NPs via the PCM to form H-CuS@PCM/DOX/Ce6 (HPDC) NPs. When exposed to near infrared laser irradiation, this nanocomplex could produce a strong photothermic effect and thus induce the controlled release of DOX and Ce6 from the melting PCM. Subsequently, the DOX-mediated chemotherapeutic effect and Ce6-mediated photodynamic effect further contributed to enhanced tumor eradication. The efficacy of this multimodal cancer treatment combining chemo-, photothermal, and photodynamic therapies was systematically evaluated both in vitro and in vivo using a 4T1 mouse mammary tumor cell line and a mouse model bearing breast cancer. Moreover, this nanoplatform exhibited minimal systemic toxicity and good hemocompatibility and may provide an effective strategy for the delivery of multiple therapeutic agents and application of multimodal cancer treatments.


Subject(s)
Breast Neoplasms , Doxorubicin , Drug Delivery Systems , Hyperthermia, Induced , Mammary Neoplasms, Experimental , Nanoparticles , Photochemotherapy , Photosensitizing Agents , Porphyrins , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Chlorophyllides , Copper/chemistry , Copper/pharmacokinetics , Copper/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Female , Infrared Rays , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Porphyrins/pharmacology , Sulfides , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL