Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Nanotechnology ; 35(29)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38593752

ABSTRACT

Melanoma is one of the most aggressive and lethal types of cancer owing to its metastatic propensity and chemoresistance property. An alternative therapeutic option is photodynamic and photothermal therapies (PDT/PTT), which employ near-infrared (NIR) light to generate heat and reactive oxygen species (ROS). As per previous reports, Melanin (Mel), and its synthetic analogs (i.e. polydopamine nanoparticles) can induce NIR light-mediated heat energy, thereby selectively targeting and ameliorating cancer cells. Similarly, chlorin e6 (Ce6) also has high ROS generation ability and antitumor activity against various types of cancer. Based on this tenet, In the current study, we have encapsulated Mel-Ce6 in a polydopamine (PDA) nanocarrier (MCP NPs) synthesized by the oxidation polymerization method. The hydrodynamic diameter of the synthesized spherical MCP NPs was 139 ± 10 nm. The MCP NPs, upon irradiation with NIR 690 nm laser for 6 min, showed photothermal efficacy of more than 50 °C. Moreover, the red fluorescence in the MCP NPs due to Ce6 can be leveraged for diagnostic purposes. Further, the MCP NPs exhibited considerable biocompatibility with the L929 cell line and exerted nearly 70% ROS-mediated cytotoxicity on the B16 melanoma cell line after the laser irradiation. Thus, the prepared MCP NPs could be a promising theranostic agent for treating the B16 melanoma cancer.


Subject(s)
Chlorophyllides , Indoles , Melanins , Melanoma, Experimental , Nanoparticles , Polymers , Porphyrins , Indoles/chemistry , Indoles/pharmacology , Polymers/chemistry , Polymers/pharmacology , Nanoparticles/chemistry , Animals , Mice , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Cell Line, Tumor , Porphyrins/chemistry , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Phototherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photochemotherapy/methods , Photothermal Therapy
2.
Biomacromolecules ; 25(4): 2302-2311, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38507248

ABSTRACT

Photodynamic therapy (PDT) employs photosensitizers to convert nearby oxygen into toxic singlet oxygen (1O2) upon laser light irradiation, showing great potential as a noninvasive approach for tumor ablation. However, the therapeutic efficacy of PDT is essentially impeded by π-π stacking and the aggregation of photosensitizers. Herein, we propose a tumor microenvironment-triggered self-adaptive nanoplatform to weaken the aggregation of photosensitizers by selenium-based oxidation at the tumor site. The selenide units in a selenium-based porphyrin-containing amphiphilic copolymer (PSe) could be oxidized into hydrophilic selenoxide units, leading to the nanoplatform self-expansion and stretching of the distance between intramolecular porphyrin units. This process could provide a better switch to greatly reduce the aggregation of photosensitive porphyrin units, generating more 1O2 upon laser irradiation. As verified in a series of in vitro and in vivo studies, PSe could be efficiently self-adapted at tumor sites, thus significantly enhancing the PDT therapeutic effect against solid tumors and minimizing side effects.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Porphyrins , Selenium , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Tumor Microenvironment , Selenium/therapeutic use , Nanoparticles/therapeutic use , Oxygen , Neoplasms/drug therapy , Neoplasms/pathology , Polymers/therapeutic use , Porphyrins/pharmacology , Cell Line, Tumor
3.
Biomater Sci ; 12(7): 1864-1870, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38411494

ABSTRACT

Hepatocellular carcinoma (HCC) is characterized by a high degree of malignancy and mortality. Sorafenib (SOR), a multi-kinase inhibitor, is clinically used in the treatment of HCC. However, SOR suffers from serious side effects and drug resistance. The development of novel therapeutic strategies for HCC therapy is urgently needed. Sonodynamic therapy (SDT) has unique advantages in treating deep tumors due to the merits of deep tissue penetration, low side effects, and the absence of drug resistance. Here, we developed multifunctional nanoparticles (NPs) termed SOR-TCPP@PEG-FA by assembling SOR, tetrakis (4-carboxyphenyl) porphyrin (TCPP), and folic acid (FA)-modified DSPE-PEG. The FA group enhances the tumor targeting capability of these NPs, while TCPP generates ROS under ultrasound (US) irradiation, which are toxic to tumor cells, and SOR with chemotherapeutic effects is released, thus realizing the synergistic SDT and chemotherapy of tumors.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Porphyrins , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Sorafenib , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Porphyrins/pharmacology , Porphyrins/therapeutic use , Cell Line, Tumor
4.
ACS Nano ; 18(1): 713-727, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38117769

ABSTRACT

Porphyrins and their derivatives are widely used as photosensitizers and sonosensitizers in tumor treatment. Nevertheless, their poor water solubility and low chemical stability reduce their singlet oxygen (1O2) yield and, consequently, their photodynamic therapy (PDT) and sonodynamic therapy (SDT) efficiency. Although strategies for porphyrin molecule assembly have been developed to augment 1O2 generation, there is scope for further improving PDT and SDT efficiencies. Herein, we synthesized ordered manganese porphyrin (SM) nanoparticles with well-defined self-assembled metalloporphyrin networks that enabled efficient energy transfer for enhanced photocatalytic and sonocatalytic activity in 1O2 production. Subsequently, Au nanoparticles were grown in situ on the SM surface by anchoring the terminal alkynyl of porphyrin to form plasmonic SMA heterostructures, which showed the excellent near-infrared-II (NIR-II) region absorption and photothermal properties, and facilitated electron-hole pair separation and transfer. With the modification of hyaluronic acid (HA), SMAH heterostructure nanocomposites exhibited good water solubility and were actively targeted to cancer cells. Under NIR-II light and ultrasound (US) irradiation, the SMAH generates hyperthermia, and a large amount of 1O2, inducing cancer cell damage. Both in vitro and in vivo studies confirmed that the SMAH nanocomposites effectively suppressed tumor growth by decreasing GSH levels in SDT-augmented PDT/PTT. Moreover, by utilizing the strong absorption in the NIR-II window, SMAH nanocomposites can achieve NIR-II photoacoustic imaging-guided combined cancer treatment. This work provides a paradigm for enhancing the 1O2 yield of metalloporphyrins to improve the synergistic therapeutic effect of SDT/PDT/PTT.


Subject(s)
Metal Nanoparticles , Nanoparticles , Neoplasms , Photoacoustic Techniques , Photochemotherapy , Porphyrins , Humans , Manganese , Porphyrins/pharmacology , Porphyrins/therapeutic use , Gold/chemistry , Metal Nanoparticles/therapeutic use , Metal Nanoparticles/chemistry , Phototherapy , Neoplasms/therapy , Water , Cell Line, Tumor
5.
J Mater Chem B ; 11(46): 11094-11102, 2023 11 29.
Article in English | MEDLINE | ID: mdl-37987617

ABSTRACT

Covalent organic frameworks (COFs) have promising applications in enhanced phototherapy. However, COFs that can sustainably play a role in phototherapy without continuous irradiation are extremely scarce. Herein, we report the fabrication of porphyrin-anthracene multifunctional COFs (Por-DPA) for sustainable photosterilization and bacterial-infected wound healing. A porphyrin photosensitizer, as one of the monomers, was used to provide photothermal and photodynamic activities under irradiation. An anthracene derivative, a good chemical source of singlet oxygen (1O2), was selected as another monomer to capture 1O2 and release it continuously via cycloreversion in the dark. The prepared Por-DPA COF prevents the self-aggregation quenching of the photosensitizer and thermal damage caused by continuous exposure to external light sources. Besides, Por-DPA exhibits good photothermal conversion performance and efficient 1O2 production capacity through dual pathways of photosensitization and cycloreversion. The developed sustainable photosterilization platform not only has good bactericidal effects on Escherichia coli and Staphylococcus aureus, but also promotes wound healing without obvious side effects, and is expected to be a novel efficient bactericide.


Subject(s)
Metal-Organic Frameworks , Porphyrins , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Metal-Organic Frameworks/pharmacology , Metal-Organic Frameworks/chemistry , Porphyrins/pharmacology , Porphyrins/chemistry , Phototherapy , Singlet Oxygen/metabolism
6.
Adv Healthc Mater ; 12(32): e2301728, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37602576

ABSTRACT

Notwithstanding that immunotherapy has made eminent clinical breakthroughs, activating the immunogenicity and breaking the immunosuppressive tumor microenvironment (ITME) remains tempting yet challenging. Herein, a customized-designed immunostimulant is engineered for attenuating ITME and eliciting an immune response to address this challenge head-on. This immunostimulant is equipped with dual silica layers coated upconversion nanoparticles (UCNPs) as nanocarriers modified with endoplasmic reticulum (ER)-targeted molecular N-p-Tosylglycine, in which the dense silica for chlorin e6 (Ce6) and the glutathione (GSH)-responsive degradable silica for loading resveratrol (RES) - (UCSMRER ). On the one hand, this precise ER-targeted photodynamic therapy (PDT) can generate reactive oxygen species (ROS) in situ under the 980 nm laser irradiation, which not only induced severe cell death directly but also caused intense ER stress-based immunogenic cell death (ICD). On the other hand, tumor hypoxia aggravated by the PDT is alleviated by RES released on-demand, which reduced oxygen consumption by impairing the mitochondrial electron transport chain (ETC). This integrated precise ER-targeted and oxygen-compensated strategy maximized the PDT effect and potentiated ICD-associated immunotherapy, which availed to attenuate ITME, activate tumor immunogenicity, and further magnify the anti-tumor effect. This innovative concept about PDT and immunotherapy sheds light on cancer-related clinical application.


Subject(s)
Nanoparticles , Photochemotherapy , Porphyrins , Oxygen , Cell Line, Tumor , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Nanoparticles/therapeutic use , Reactive Oxygen Species/metabolism , Silicon Dioxide , Endoplasmic Reticulum/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Porphyrins/pharmacology
7.
Adv Healthc Mater ; 12(25): e2300752, 2023 10.
Article in English | MEDLINE | ID: mdl-37306666

ABSTRACT

The combination of phototherapy and chemotherapy holds great potential for cancer treatment, while hypoxia in tumor as well as unexpected drug release largely restricts anticancer therapy. Inspired by the natural intelligence, herein, for the first time, a "bottom-up" protein self-assembly strategy mediated by near-infrared (NIR) quantum dots (QDs) with multicharged electrostatic interactions is presented to develop a tumor microenvironment (TME)-responsive theranostic nanoplatform for imaging-guided synergistic photodynamic therapy (PDT)/photothermal therapy (PTT)/chemotherapy. Catalase (CAT) possesses diverse surface charge distribution under different pH conditions. After modification by chlorin e6 (Ce6), the formulated CAT-Ce6 with patchy negative charges can be assembled with NIR Ag2 S QDs by regulating their electrostatic interactions, allowing for effective incorporation of specific anticancer drug oxaliplatin (Oxa). Such Ag2 S@CAT-Ce6@Oxa nanosystems are able to visualize nanoparticle (NP) accumulation to guide subsequent phototherapy, together with significant alleviation of tumor hypoxia to further enhance PDT. Moreover, the acidic TME triggers controllable disassembly through weakening the CAT surface charge to disrupt electrostatic interactions, allowing for sustained drug release. Both in vitro and in vivo results demonstrate remarkable inhibition of colorectal tumor growth with a synergistic effect. Overall, this multicharged electrostatic protein self-assembly strategy provides a versatile platform for realizing TME-specific theranostics with high efficiency and safety, promising for clinical translation.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Porphyrins , Quantum Dots , Humans , Photothermal Therapy , Phototherapy/methods , Neoplasms/drug therapy , Hypoxia/drug therapy , Porphyrins/pharmacology , Porphyrins/therapeutic use , Photochemotherapy/methods , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Tumor Microenvironment
8.
ACS Appl Mater Interfaces ; 15(21): 25369-25381, 2023 May 31.
Article in English | MEDLINE | ID: mdl-37199535

ABSTRACT

Photodynamic therapy (PDT) is a promising strategy for cancer treatment. However, its efficiency is hindered by three key parameters, namely, limited penetration depth of external light, tumor hypoxia, and self-aggregation of photosensitizers. Herein, we fabricated a novel "all-in-one" chemiluminescence-PDT nanosystem through the integration of an oxygen-supplying protein (hemoglobin, Hb) and a luminescent donor (luminol, Lum) in hierarchically engineered mesoporous porphyrinic metal-organic framework (MOF) nanoparticles. Mechanistically, the in situ chemiluminescence of Lum is activated by the high concentration of H2O2 in 4T1 cancer cells and further catalyzed by Hb and then absorbed by the porphyrin ligands in MOF nanoparticles through chemiluminescence resonance energy transfer. The excited porphyrins then sensitize oxygen supplied by Hb to produce sufficient reactive oxygen species that kill cancer cells. The MOF-based nanocomposite demonstrates excellent anticancer activity both in vitro and in vivo, with eventually a 68.1% tumor inhibition rate after intravenous injections without external light irradiation. This self-illuminating, oxygen-self-supplying nanosystem integrates all essential components of PDT into one simple nanoplatform, demonstrating great potential for the selective phototherapy of deep-seated cancer.


Subject(s)
Metal-Organic Frameworks , Nanocomposites , Neoplasms , Photochemotherapy , Porphyrins , Humans , Metal-Organic Frameworks/pharmacology , Luminescence , Hydrogen Peroxide , Tumor Microenvironment , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Oxygen , Neoplasms/drug therapy , Nanocomposites/therapeutic use , Porphyrins/pharmacology , Cell Line, Tumor
9.
J Nanobiotechnology ; 21(1): 146, 2023 May 04.
Article in English | MEDLINE | ID: mdl-37143039

ABSTRACT

BACKGROUND: Uveal melanoma (UM) is adults' most common primary intraocular malignant tumor, prone to metastasis and high mortality. Eyeball enucleation commonly used in the clinic will lead to permanent blindness and mental disorders. Thus, new methods are urgently needed to diagnose and treat UM early to preserve patients' vision. METHODS AND RESULTS: Herein, multifunctional nanoparticles (NPs) were synthesized by loading chlorin e6 (Ce6) in poly-lactic-co-glycolic acid (PLGA) NPs and wrapping FeIII-tannic acid (FeIII-TA) on the outside (FeIII-TA/PLGA/Ce6, designated as FTCPNPs). Then, the synergistic photothermal therapy (PTT) and photodynamic therapy (PDT) antitumor effects of FTCPNPs excited by near-infrared (NIR) laser were evaluated. Moreover, we verified the mechanism of synergistic PTT/PDT leading to mitochondrial dysfunction and inducing tumor cell apoptosis. Additionally, FTCPNPs can be used as excellent magnetic resonance (MR)/photoacoustic (PA) imaging contrast agents, enabling imaging-guided cancer treatment. Finally, The NPs have good biological safety. CONCLUSION: This noninvasive NIR light-triggered cooperative phototherapy can easily penetrate eye tissue and overcome the disadvantage of limited penetration of phototherapy. Therefore, cooperative phototherapy is expected to be used in fundus tumors. This treatment model is applied to UM for the first time, providing a promising strategy and new idea for integrating the diagnosis and treatment of UM.


Subject(s)
Melanoma , Nanoparticles , Photochemotherapy , Porphyrins , Humans , Ferric Compounds , Cell Line, Tumor , Phototherapy/methods , Melanoma/diagnostic imaging , Melanoma/therapy , Porphyrins/therapeutic use , Porphyrins/pharmacology , Nanoparticles/therapeutic use
10.
J Colloid Interface Sci ; 645: 122-132, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37146376

ABSTRACT

Multifunctional nanoagents with photodynamic therapy (PDT) and photothermal therapy (PTT) functions have shown great promise for cancer treatment, while the design and synthesis of efficient nanoagents remain a challenge. To realize nanozyme-enhanced PDT-PTT combined therapy, herein we have synthesized the Ce6@CuS-Pt/PEG nanoplatforms as a model of efficient nanoagents. Hollow CuS nanospheres with an average diameter of âˆ¼ 200 nm are first synthesized through vulcanization using Cu2O as the precursor. Subsequently, CuS nanospheres are surface-decorated with Pt nanoparticles (NPs) as nanozyme via an in-situ reduction route, followed by modifying the DSPE-PEG5000 and loading the photosensitizer Chlorin e6 (Ce6). The obtained Ce6@CuS-Pt/PEG NPs exhibit high photothermal conversion efficiency (43.08%), good singlet oxygen (1O2) generation ability, and good physiological stability. In addition, Ce6@CuS-Pt/PEG NPs show good catalytic performance due to the presence of Pt nanozyme, which can effectively convert H2O2 to O2 and significantly enhance the production of cytotoxic 1O2. When Ce6@CuS-Pt/PEG NPs dispersion is injected into mice, the tumors can be wholly suppressed owing to nanozyme-enhanced PDT-PTT combined therapy, providing better therapeutic effects compared to single-mode phototherapy. Thus, the present Ce6@CuS-Pt/PEG NPs can act as an efficient multifunctional nanoplatform for tumor therapy.


Subject(s)
Nanoparticles , Nanospheres , Photochemotherapy , Porphyrins , Animals , Mice , Photothermal Therapy , Precision Medicine , Hydrogen Peroxide , Photosensitizing Agents , Nanoparticles/therapeutic use , Polyethylene Glycols , Cell Line, Tumor , Porphyrins/pharmacology
11.
Acta Biomater ; 161: 112-133, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36907234

ABSTRACT

Wound treatment is largely influenced by pre-existing hypoxic microenvironments and biofilms, which can severely diminish the efficacy of phototherapy, suggesting the importance of multifunctional nanoplatforms for synergistic treatment of wound infections. Here, we developed a multifunctional injectable hydrogel (PSPG hydrogel) by loading photothermal sensitive sodium nitroprusside (SNP) into Pt-modified porphyrin metal organic framework (PCN) and in situ modification of gold particles to form a near-infrared (NIR) light-triggered all-in-one phototherapeutic nanoplatform. The Pt-modified nanoplatform exhibits a remarkable catalase-like behavior and promotes the continuous decomposition of endogenous H2O2 into O2, thereby enhancing the photodynamic therapy (PDT) effect under hypoxia. Under dual NIR irradiation, PSPG hydrogel can not only produce hyperthermia (η=89.21%) but also generate reactive oxygen species and trigger NO release, contributing jointly to removal of biofilms and disruption of the cell membranes of methicillin-resistant Staphylococcus aureus (MRSA) and Escherichia coli (E. coli). In vivo experiments demonstrated a 99.9% reduction in bacterial burden on wounds. Additionally, PSPG hydrogel can accelerate MRSA-infected and Pseudomonas aeruginosa-infected (P. aeruginosa-infected) wound healing by promoting angiogenesis, collagen deposition, and suppressing inflammatory responses. Furthermore, in vitro and in vivo experiments revealed that PSPG hydrogel has good cytocompatibility. Overall, we proposed an antimicrobial strategy to eliminate bacteria through the synergistic effects of gas-photodynamic-photothermal killing, alleviating hypoxia in the bacterial infection microenvironment, and inhibiting biofilms, offering a new way against antimicrobial resistance and biofilm-associated infections. STATEMENT OF SIGNIFICANCE: The NIR light-triggered multifunctional injectable hydrogel nanoplatform (PSPG hydrogel) based on Pt-decorated gold nanoparticles with sodium nitroprusside (SNP)-loading porphyrin metal organic framework (PCN) as inner templates can efficiently perform photothermal conversion (η=89.21%) to trigger NO release from SNP, while continuously regulating the hypoxic microenvironment at the bacterial infection site through Pt-induced self-oxygenation, achieving efficient sterilization and removal of biofilm by synergistic PDT and PTT phototherapy. In vivo and in vitro experiments demonstrated that the PSPG hydrogel has significant anti-biofilm, antibacterial, and inflammatory regulatory functions. This study proposed an antimicrobial strategy to eliminate bacteria through the synergistic effects of gas-photodynamic-photothermal killing, alleviating hypoxia in the bacterial infection microenvironment, and inhibiting biofilms.


Subject(s)
Metal Nanoparticles , Metal-Organic Frameworks , Methicillin-Resistant Staphylococcus aureus , Porphyrins , Humans , Hydrogels/pharmacology , Hydrogen Peroxide/pharmacology , Escherichia coli , Gold/pharmacology , Nitroprusside/pharmacology , Wound Healing , Hypoxia , Porphyrins/pharmacology , Anti-Bacterial Agents/pharmacology , Biofilms
12.
Microb Pathog ; 178: 106081, 2023 May.
Article in English | MEDLINE | ID: mdl-36948363

ABSTRACT

Infectious bovine keratoconjunctivitis (IBK) is the most important eye disease in ruminants worldwide. Moraxella bovis and Moraxella bovoculi can form biofilm and are frequently isolated from affected animals. Antimicrobials are used worldwide to treat clinical cases of IBK, although they have limited success in clearing the infection. Therefore, photodynamic therapy using porphyrins as photosensitizing molecules is an alternative method to eliminate microorganisms, including biofilms. We evaluated the antibacterial activity of a zinc(II) metalloporphyrin (ZnTMeP) against M. bovis and M. bovoculi biofilms since this compound can efficiently inactivate planktonic Moraxella spp. This study was carried out with two reference strains of Moraxella spp. (M. bovis: ATCC® 10900 and M. bovoculli: ATCC® BAA1259). The antibacterial activity of 4.0 µM of the ZnTMeP porphyrin was evaluated on forming and consolidate biofilms with three 30-min cycles of white-light exposure for three days. The ZnTMeP porphyrin reduced M. bovis and M. bovoculi biofilm formation. In addition, ZnTMeP partially destroyed consolidated M. bovoculi biofilms in the second white-light irradiation cycle, although the porphyrin had no effect against the consolidated biofilm of M. bovis. Despite the biofilm still not being completely inactivated, our findings are promising and encourage further experiments using the phototherapy protocol.


Subject(s)
Cattle Diseases , Keratoconjunctivitis, Infectious , Moraxellaceae Infections , Photochemotherapy , Porphyrins , Cattle , Animals , Porphyrins/pharmacology , Keratoconjunctivitis, Infectious/microbiology , Cattle Diseases/drug therapy , Cattle Diseases/microbiology , Moraxella , Anti-Bacterial Agents/pharmacology , Moraxellaceae Infections/microbiology , Moraxellaceae Infections/veterinary , Biofilms
13.
Molecules ; 28(5)2023 Feb 22.
Article in English | MEDLINE | ID: mdl-36903314

ABSTRACT

Sulfonamides are a conventional class of antibiotics that are well-suited to combat infections. However, their overuse leads to antimicrobial resistance. Porphyrins and analogs have demonstrated excellent photosensitizing properties and have been used as antimicrobial agents to photoinactivate microorganisms, including multiresistant Staphylococcus aureus (MRSA) strains. It is well recognized that the combination of different therapeutic agents might improve the biological outcome. In this present work, a novel meso-arylporphyrin and its Zn(II) complex functionalized with sulfonamide groups were synthesized and characterized and the antibacterial activity towards MRSA with and without the presence of the adjuvant KI was evaluated. For comparison, the studies were also extended to the corresponding sulfonated porphyrin TPP(SO3H)4. Photodynamic studies revealed that all porphyrin derivatives were effective in photoinactivating MRSA (>99.9% of reduction) at a concentration of 5.0 µM upon white light radiation with an irradiance of 25 mW cm-2 and a total light dose of 15 J cm-2. The combination of the porphyrin photosensitizers with the co-adjuvant KI during the photodynamic treatment proved to be very promising allowing a significant reduction in the treatment time and photosensitizer concentration by six times and at least five times, respectively. The combined effect observed for TPP(SO2NHEt)4 and ZnTPP(SO2NHEt)4 with KI seems to be due to the formation of reactive iodine radicals. In the photodynamic studies with TPP(SO3H)4 plus KI, the cooperative action was mainly due to the formation of free iodine (I2).


Subject(s)
Iodine , Methicillin-Resistant Staphylococcus aureus , Photochemotherapy , Porphyrins , Staphylococcal Infections , Humans , Photosensitizing Agents/pharmacology , Staphylococcus aureus , Porphyrins/pharmacology , Anti-Bacterial Agents/pharmacology , Sulfanilamide/pharmacology , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/pharmacology , Iodine/pharmacology
14.
Small ; 19(23): e2300594, 2023 06.
Article in English | MEDLINE | ID: mdl-36755191

ABSTRACT

A primary concern about photodynamic therapy (PDT) is its inability to regulate the generation levels of reactive oxidative species (ROS) based on the complex microenvironment, resulting in the impairment toward normal tissues and immunosuppression. Besides, tumor metastasis also compromises PDT's efficacy and drives mortality. However, it is very challenging to achieve such two goals within one nanosystem. Here, the nanoassembly (CPR) with self-regulated photodynamic and antimetastasis properties comprises three parts: chlorin e6-conjugated ß-cyclodextrin (CD-Ce6) acts as the main PDT agent and ferrocene (Fc)-terminated phenylboronic acid-containing conjugates entering into the cavity of CD-Ce6, as well as rosmarinic acid (RA)-boronic acid crosslinked shell. Compared with non-crosslinked counterpart, CPR displays better stability and enhanced tumor accumulation. Under laser irradiation, CPR generates plenty of ROS to damage tumor cells and induce immunogenic cell death. Mildly acidic TME partly cleaves the crosslinkers to dissociate antioxidant RAs from micelles, which together with Fc in CPR scavenge PDT-induced ROS in the TME. By contrast, under acidic lysosomal conditions, Fc catalyzes abundant H2 O2 in tumor cells to produce highly cytotoxic •OH, while RA continuously reduces ferroptosis-generated Fc+ into Fc, both to augment the PDT efficacy in tumor cells. CPR also remarkably hinders the epithelial-mesenchymal transition to prevent the lung metastasis.


Subject(s)
Nanoparticles , Photochemotherapy , Porphyrins , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Phototherapy , Cinnamates/pharmacology , Porphyrins/pharmacology , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Rosmarinic Acid
15.
Biomater Sci ; 11(5): 1776-1784, 2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36648453

ABSTRACT

Porphyrinic covalent organic frameworks (COFs) have emerged as prospective materials in photodynamic and photothermal sterilization. However, it is still a great challenge to construct an efficient COF-based sterilizing agent with good photothermal and photodynamic properties and bacterial targeting ability. Herein, we report a multifunctional porphyrin-COF for bacterial-targeted and reaction-enhanced synergistic phototherapy/chemotherapy for sterilization and wound healing. The ordered crystal structure of the porphyrin-COF not only effectively avoids the self-aggregation-induced quenching of the porphyrin monomer, but also facilitates the storage and transport of singlet oxygen. The acrylate substituent in the other monomer serves as a bacterial targeting moiety and the in situ reaction site with the sulfhydryl group of the bacterial surface protein via a Michael addition reaction, thus fixing the bacteria on the surface of COF and making them lose the colonization ability. Furthermore, the bonding of COF and bacteria further amplifies the therapeutic efficiency of phototherapy. Therefore, the developed multifunctional sterilization platform not only provides a new strategy for the design of novel bactericidal materials but also broadens the biological applications of COF-based materials.


Subject(s)
Metal-Organic Frameworks , Porphyrins , Metal-Organic Frameworks/pharmacology , Metal-Organic Frameworks/chemistry , Porphyrins/pharmacology , Porphyrins/chemistry , Phototherapy , Bacteria , Wound Healing
16.
Mol Pharm ; 20(2): 875-885, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36689197

ABSTRACT

Chlorin e6 (Ce6) has been extensively researched and developed as an antitumor therapy. Ce6 is a highly effective photosensitizer and sonosensitizer with promising future applications in photodynamic therapy, dynamic acoustic therapy, and combined acoustic and light therapy for tumors. Ce6 is also being studied for other applications in fluorescence navigation, antibacterials, and plant growth regulation. Here we review the role and research status of Ce6 in tumor therapy and the problems and challenges of its clinical application. Other biomedical effects of Ce6 are also briefly discussed. Despite the difficulties in clinical application, Ce6 has significant advantages in photodynamic therapy (PDT)/sonodynamic therapy (SDT) against cancer and offers several possibilities in clinical utility.


Subject(s)
Chlorophyllides , Neoplasms , Photochemotherapy , Porphyrins , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Neoplasms/drug therapy , Porphyrins/pharmacology , Porphyrins/therapeutic use , Cell Line, Tumor
17.
Biomater Sci ; 11(1): 108-118, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36468355

ABSTRACT

The application of photodynamic therapy (PDT) is limited by tumor hypoxia. To overcome hypoxia, catalase-like nanozymes are often used to catalyze endogenous H2O2 enriched in tumor tissues to O2. Nonetheless, the catalase activity may not be optimal at body temperature and the O2 supply may not meet the rapid O2 consumption of PDT. Herein, we provide a two-pronged strategy to alleviate tumor hypoxia based on hollow mesoporous Prussian blue nanoparticles (HMPB NPs). HMPB NPs can efficiently load the photosensitizer chlorin e6 (Ce6) and exhibit photothermal capability and temperature-dependent catalase activity. Under 808 nm laser irradiation, the photothermal effect of HMPB NPs elevated the catalase activity of HMPB NPs for O2 production. Furthermore, mild hyperthermia reduced cancer associated fibroblasts (CAFs) and induced extracellular matrix (ECM) degradation. The reduction of CAFs and the ECM decreased the solid stress of tumor tissues and normalized the tumor vasculature, which was beneficial for the external supplementation of O2 to tumors. Thereafter, under 606 nm laser irradiation, Ce6-mediated PDT generated excessive reactive oxygen species (ROS) that induced tumor cell apoptosis and achieved a high tumor inhibition rate of 92.2% in 4T1 breast tumors. Our work indicated that the alleviation of tumor hypoxia from both internal and external pathways significantly enhanced Ce6-mediated PDT against breast cancers.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Photochemotherapy , Porphyrins , Humans , Catalase , Hydrogen Peroxide , Tumor Hypoxia , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Porphyrins/pharmacology
18.
Small ; 18(52): e2204951, 2022 12.
Article in English | MEDLINE | ID: mdl-36333122

ABSTRACT

Photodynamic therapy (PDT) has been showing great potential in cancer treatment. However, the efficacy of PDT is always limited by the intrinsic hypoxic tumor microenvironment (TME) and the low accumulation efficiency of photosensitizers in tumors. To address the issue, a multifunctional hollow multilayer nanoplatform (H-MnO2 @TPyP@Bro) comprising manganese dioxide, porphyrin (TPyP) and bromelain (Bro), is developed for enhanced photodynamic therapy. MnO2 catalyzes the intracellular hydrogen peroxide (H2 O2 ) to produce oxygen (O2 ), reversing the hypoxic TME in vivo. The generated O2 is converted into singlet oxygen (1 O2 ) by the TPyP shell under near-infrared light, which can inhibit tumor proliferation. Meanwhile, the Bro can digest collagen in the extracellular matrix around the tumor, and can promote the accumulation of H-MnO2 @TPyP@Bro in the deeper tumor tissue, further improving the therapeutic effect of PDT. In addition, MnO2 can react with the overexpressed glutathione in TME to release Mn2+ . Consequently, Mn2+ not only induces chemo-dynamic therapy based on Fenton reaction by converting H2 O2 into hydroxyl radicals, but also activates the Mn2+ -based magnetic resonance imaging. Therefore, the developed H-MnO2 @TPyP@Bro nanoplatform can effectively modulate the unfavorable TME and overcome the limitations of conventional PDT for cancer diagnostic and therapeutic.


Subject(s)
Neoplasms , Photochemotherapy , Porphyrins , Humans , Photochemotherapy/methods , Manganese Compounds , Porphyrins/pharmacology , Porphyrins/therapeutic use , Bromelains/pharmacology , Bromelains/therapeutic use , Oxides/pharmacology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Oxygen/pharmacology , Neoplasms/therapy , Hydrogen Peroxide/pharmacology , Tumor Microenvironment
19.
Int J Biol Macromol ; 221: 121-134, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36049568

ABSTRACT

Photothermal and photodynamic therapies (PTT/PDT) have been widely accepted as noninvasive therapeutic methods for cancer treatment. However, tumor hypoxia and insufficient delivery of photoactive compounds to cancer cells can reduce the efficacy of phototherapy. Herein, we first synthesized thiolated hyaluronic acid (THA) and then conjugated it with catalase (CAT) onto chlorin e6 (Ce6)-adsorbed small gold nanorods (Ce6@sAuNRs) with near-infrared (NIR)/visible light activated photothermal/photodynamic effects. The conjugation of THA and CAT on Ce6@sAuNRs resulted in a red-shift of the longitudinal LSPR absorption band of sAuNRs up to 1000 nm and maintained the excellent enzymatic activity of catalase. Modification of Ce6@sAuNRs with THA resulted in efficient internalization of the nanocomposite into MCF-7/ADR multidrug-resistant (MDR) breast cancer cells (CD44+), thereby significantly enhancing the intracellular accumulation of the photosensitizer Ce6. CAT endows Ce6@sAuNRs with self-supporting oxygen production, which enables them to efficiently generate singlet oxygen (1O2) under 660 nm laser irradiation and enhances the photodynamic effect against hypoxic breast cancer cells. The results highlight the prospect of this novel multi-functional nanoplatform integrating active biological macromolecules (THA and CAT) into photosensitizer/photothermal gold nanocomposites in overcoming the limitations of hypoxic MDR breast cancer cell treatment.


Subject(s)
Breast Neoplasms , Photochemotherapy , Porphyrins , Catalase , Gold/pharmacology , Hyaluronic Acid/pharmacology , Oxygen , Photochemotherapy/methods , Photosensitizing Agents , Porphyrins/pharmacology , Breast Neoplasms/drug therapy , Humans , Hyaluronan Receptors , Nanotubes , MCF-7 Cells
20.
J Nanobiotechnology ; 20(1): 410, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36109814

ABSTRACT

Photodynamic therapy (PDT) has emerged as a promising tumor treatment method via light-triggered generation of reactive oxygen species (ROS) to kill tumor cells. However, the efficacy of PDT is usually restricted by several biological limitations, including hypoxia, excess glutathione (GSH) neutralization, as well as tumor resistance. To tackle these issues, herein we developed a new kind of DNA nanozyme to realize enhanced PDT and synergistic tumor ferroptosis. The DNA nanozyme was constructed via rolling circle amplification, which contained repeat AS1411 G quadruplex (G4) units to form multiple G4/hemin DNAzymes with catalase-mimic activity. Both hemin, an iron-containing porphyrin cofactor, and chlorine e6 (Ce6), a photosensitizer, were facilely inserted into G4 structure with high efficiency, achieving in-situ catalytic oxygenation and photodynamic ROS production. Compared to other self-oxygen-supplying tools, such DNA nanozyme is advantageous for high biological stability and compatibility. Moreover, the nanostructure could achieve tumor cells targeting internalization and intranuclear transport of Ce6 by virtue of specific nucleolin binding of AS1411. The nanozyme could catalyze the decomposition of intracellular H2O2 into oxygen for hypoxia relief as evidenced by the suppression of hypoxia-inducible factor-1α (HIF-1α), and moreover, GSH depletion and cell ferroptosis were also achieved for synergistic tumor therapy. Upon intravenous injection, the nanostructure could effectively accumulate into tumor, and impose multi-modal tumor therapy with excellent biocompatibility. Therefore, by integrating the capabilities of O2 generation and GSH depletion, such DNA nanozyme is a promising nanoplatform for tumor PDT/ferroptosis combination therapy.


Subject(s)
DNA, Catalytic , Ferroptosis , Photochemotherapy , Porphyrins , Catalase , DNA , Glutathione/metabolism , Hemin , Humans , Hydrogen Peroxide , Hypoxia/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit , Iron , Oxygen , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL