Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.869
Filter
1.
Bull Math Biol ; 86(7): 83, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38842602

ABSTRACT

5-Aminolevulinic Acid (5-ALA) is the only fluorophore approved by the FDA as an intraoperative optical imaging agent for fluorescence-guided surgery in patients with glioblastoma. The dosing regimen is based on rodent tests where a maximum signal occurs around 6 h after drug administration. Here, we construct a computational framework to simulate the transport of 5-ALA through the stomach, blood, and brain, and the subsequent conversion to the fluorescent agent protoporphyrin IX at the tumor site. The framework combines compartmental models with spatially-resolved partial differential equations, enabling one to address questions regarding quantity and timing of 5-ALA administration before surgery. Numerical tests in two spatial dimensions indicate that, for tumors exceeding the detection threshold, the time to peak fluorescent concentration is 2-7 h, broadly consistent with the current surgical guidelines. Moreover, the framework enables one to examine the specific effects of tumor size and location on the required dose and timing of 5-ALA administration before glioblastoma surgery.


Subject(s)
Aminolevulinic Acid , Brain Neoplasms , Computer Simulation , Glioblastoma , Mathematical Concepts , Models, Biological , Protoporphyrins , Surgery, Computer-Assisted , Glioblastoma/surgery , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/diagnostic imaging , Aminolevulinic Acid/administration & dosage , Humans , Brain Neoplasms/surgery , Protoporphyrins/administration & dosage , Protoporphyrins/metabolism , Surgery, Computer-Assisted/methods , Animals , Photosensitizing Agents/administration & dosage , Optical Imaging/methods , Fluorescent Dyes/administration & dosage
2.
J Drugs Dermatol ; 23(5): 332-337, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38709699

ABSTRACT

BACKGROUND: Peer-reviewed, clinical studies measuring the efficacy and usability of skin care products enhance their integrity and may guide experts in the field in providing recommendations. A single-blind, prospective clinical study was designed to assess the subject satisfaction, clinical benefit, and safety of three photodynamic topical formulations referred to as MMSRepose (MMSRep), MMSRevive (MMSRev), and MMSBalance (MMSB).  Methods: Thirteen male and female patients (mean age 49 +/- 17.8 years) applied one of the three topical serums twice daily over a period of 12 weeks. Subjects returned for photography, and blinded investigator evaluation of rhytides (fine lines) and dyspigmentation were measured on a 6- and 4-point scale, respectively. Patient-perceived efficacy of multiple clinical outcomes was measured on a 5-point scale.  Results: 100% of subjects reported at least a 1-grade improvement in global aesthetic at the conclusion of the study. Investigator assessment revealed an overall 53.3% decrease in rhytides, correlating to a mean point reduction from 1.65 +/- 0.77 to 0.77 +/- 0.53 (P<0.001) from baseline to week 12. Investigator assessment of dyspigmentation revealed a 62.7% decrease, correlating to a mean point reduction of 1.85 +/- 0.68 from week 1 to 0.69 +/- 0.48 at week 12 (P<0.001). CONCLUSION: Photodynamic serums demonstrate clinical efficacy in skin rejuvenation and high user satisfaction. There were no serious adverse events. This study is limited by the inability to randomize to placebo due to the small sample size, as subject retention was heavily impacted by the SARS-CoV-2 pandemic. Future studies may be indicated to undergo comparison with a larger cohort.  J Drugs Dermatol. 2024;23(5):332-337. doi:10.36849/JDD.7167.


Subject(s)
Patient Satisfaction , Photochemotherapy , Skin Aging , Humans , Prospective Studies , Female , Male , Middle Aged , Photochemotherapy/methods , Photochemotherapy/adverse effects , Skin Aging/drug effects , Single-Blind Method , Adult , Aged , Treatment Outcome , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/adverse effects , Skin Care/methods , Administration, Cutaneous , Rejuvenation
3.
Int J Nanomedicine ; 19: 3737-3751, 2024.
Article in English | MEDLINE | ID: mdl-38699684

ABSTRACT

Background: Chemo-photodynamic combination therapy has demonstrated significant potential in the treatment of cancer. Triptolide (TPL), a naturally derived anticancer agent, when combined with the photosensitizer Chlorin e6 (Ce6), has shown to provide enhanced anti-tumor benefits. However, the development of stimuli-responsive nanovehicles for the co-delivery of TPL and Ce6 could further enhance the efficacy of this combination therapy. Methods: In this study, we synthesized a pH/ROS dual-responsive mPEG-TK-PBAE copolymer, which contains a pH-sensitive PBAE moiety and a ROS-sensitive thioketal (TK) linkage. Through a self-assembly process, TPL and Ce6 were successfully co-loaded into mPEG-TK-PBAE nanoparticles, hereafter referred to as TPL/Ce6 NPs. We evaluated the pH- and ROS-sensitive drug release and particle size changes. Furthermore, we investigated both the in vitro suppression of cellular proliferation and induction of apoptosis in HepG2 cells, as well as the in vivo anti-tumor efficacy of TPL/Ce6 NPs in H22 xenograft nude mice. Results: The mPEG-TK-PBAE copolymer was synthesized through a one-pot Michael-addition reaction and successfully co-encapsulated both TPL and Ce6 by self-assembly. Upon exposure to acid pH values and high ROS levels, the payloads in TPL/Ce6 NPs were rapidly released. Notably, the abundant ROS generated by the released Ce6 under laser irradiation further accelerated the degradation of the nanosystem, thereby amplifying the tumor microenvironment-responsive drug release and enhancing anticancer efficacy. Consequently, TPL/Ce6 NPs significantly increased PDT-induced oxidative stress and augmented TPL-induced apoptosis in HepG2 cells, leading to synergistic anticancer effects in vitro. Moreover, administering TPL/Ce6 NPs (containing 0.3 mg/kg of TPL and 4 mg/kg of Ce6) seven times, accompanied by 650 nm laser irradiation, efficiently inhibited tumor growth in H22 tumor-bearing mice, while exhibiting lower systemic toxicity. Conclusion: Overall, we have developed a tumor microenvironment-responsive nanosystem for the co-delivery of TPL and Ce6, demonstrating amplified synergistic effects of chemo-photodynamic therapy (chemo-PDT) for hepatocellular carcinoma (HCC) treatment.


Subject(s)
Apoptosis , Chlorophyllides , Diterpenes , Liver Neoplasms , Mice, Nude , Phenanthrenes , Photochemotherapy , Photosensitizing Agents , Porphyrins , Reactive Oxygen Species , Animals , Humans , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Hep G2 Cells , Liver Neoplasms/drug therapy , Porphyrins/chemistry , Porphyrins/pharmacology , Porphyrins/administration & dosage , Porphyrins/pharmacokinetics , Diterpenes/chemistry , Diterpenes/pharmacology , Diterpenes/pharmacokinetics , Diterpenes/administration & dosage , Hydrogen-Ion Concentration , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Apoptosis/drug effects , Mice , Carcinoma, Hepatocellular/drug therapy , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Epoxy Compounds/administration & dosage , Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Drug Liberation , Cell Proliferation/drug effects , Polyethylene Glycols/chemistry , Combined Modality Therapy
4.
Int J Nanomedicine ; 19: 4679-4699, 2024.
Article in English | MEDLINE | ID: mdl-38803997

ABSTRACT

Background: Breast cancer is a heterogeneous disease globally accounting for approximately 1 million new cases annually. Chemotherapy remains the main therapeutic option, but the antitumor efficacy needs to be improved. Methods: Two multifunctional nanoparticles were developed in this paper using oleic acid and mPEG2k-PCL2k as the drug carriers. Squamocin (Squ) was employed as a chemotherapeutic agent. Resiquimod (R848) or ginsenoside Rh2 was co-encapsulated in the nanoparticles to remold the immunosuppressive tumor microenvironment, and IR780 was coloaded as a photosensitizer to realize photothermal therapy. Results: The obtained Squ-R848-IR780 nanoparticles and Squ-Rh2-IR780 nanoparticles were uniformly spherical and approximately (162.200 ± 2.800) nm and (157.300 ± 1.1590) nm, respectively, in average diameter, with good encapsulation efficiency (above 85% for each drug), excellent stability in various physiological media and high photothermal conversion efficiency (24.10% and 22.58%, respectively). After intravenous administration, both nanoparticles quickly accumulated in the tumor and effectively enhanced the local temperature of the tumor to over 45 °C when irradiated by an 808 nm laser. At a low dose of 0.1 mg/kg, Squ nanoparticles treatment alone displayed a tumor inhibition rate of 55.28%, pulmonary metastasis inhibition rate of 59.47% and a mean survival time of 38 days, which were all higher than those of PTX injection (8 mg/kg) (43.64%, 25 days and 37.25%), indicating that Squ was a potent and effective antitumor agent. Both multifunctional nanoparticles, Squ-Rh2-IR780 nanoparticles and Squ-R848-IR780 nanoparticles, demonstrated even better therapeutic efficacy, with tumor inhibition rates of 90.02% and 97.28%, pulmonary metastasis inhibition rates of 95.42% and 98.09, and mean survival times of 46 days and 52 days, respectively. Conclusion: The multifunctional nanoparticles coloaded with squamocin, R848 and IR 780 achieved extraordinary therapeutic efficacy and excellent antimetastasis activity and are thus promising in the future treatment of breast tumors and probably other tumors.


Subject(s)
Breast Neoplasms , Indoles , Nanoparticles , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Animals , Nanoparticles/chemistry , Humans , Indoles/chemistry , Indoles/pharmacology , Cell Line, Tumor , Mice , Drug Carriers/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Photothermal Therapy/methods , Mice, Inbred BALB C , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Imidazoles/chemistry , Imidazoles/pharmacology , Imidazoles/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Tumor Microenvironment/drug effects
5.
Int J Nanomedicine ; 19: 4701-4717, 2024.
Article in English | MEDLINE | ID: mdl-38808148

ABSTRACT

Purpose: Numerous failures in melanoma treatment as a highly aggressive form of skin cancer with an unfavorable prognosis and excessive resistance to conventional therapies are prompting an urgent search for more effective therapeutic tools. Consequently, to increase the treatment efficiency and to reduce the side effects of traditional administration ways, herein, it has become crucial to combine photodynamic therapy as a promising therapeutic approach with the selectivity and biocompatibility of a novel colloidal transdermal nanoplatform for effective delivery of hybrid cargo with synergistic effects on melanoma cells. Methods: The self-assembled bilosomes, co-stabilized with L-α-phosphatidylcholine, sodium cholate, Pluronic® P123, and cholesterol, were designated, and the stability of colloidal vesicles was studied using dynamic and electrophoretic light scattering, also provided in cell culture medium (Dulbecco's Modified Eagle's Medium). The hybrid compounds - a classical photosensitizer (Methylene Blue) along with a complementary natural polyphenolic agent (curcumin), were successfully co-loaded, as confirmed by UV-Vis, ATR-FTIR, and fluorescent spectroscopies. The biocompatibility and usefulness of the polymer functionalized bilosome with loaded double cargo were demonstrated in vitro cyto- and phototoxicity experiments using normal keratinocytes and melanoma cancer cells. Results: The in vitro bioimaging and immunofluorescence study upon human skin epithelial (A375) and malignant (Me45) melanoma cell lines established the protective effect of the PEGylated bilosome surface. This effect was confirmed in cytotoxicity experiments, also determined on human cutaneous (HaCaT) keratinocytes. The flow cytometry experiments indicated the enhanced uptake of the encapsulated hybrid cargo compared to the non-loaded MB and CUR molecules, as well as a selectivity of the obtained nanocarriers upon tumor cell lines. The phyto-photodynamic action provided 24h-post irradiation revealed a more significant influence of the nanoplatform on Me45 cells in contrast to the A375 cell line, causing the cell viability rate below 20% of the control. Conclusion: As a result, we established an innovative and effective strategy for potential metastatic melanoma treatment through the synergism of phyto-photodynamic therapy and novel bilosomal-origin nanophotosensitizers.


Subject(s)
Curcumin , Melanoma , Nanomedicine , Photochemotherapy , Photosensitizing Agents , Skin Neoplasms , Humans , Skin Neoplasms/drug therapy , Melanoma/drug therapy , Photochemotherapy/methods , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/administration & dosage , Curcumin/chemistry , Curcumin/pharmacology , Cell Survival/drug effects , Liposomes/chemistry , Liposomes/pharmacology , Cholesterol/chemistry , Phosphatidylcholines/chemistry , Phosphatidylcholines/pharmacology , Sodium Cholate/chemistry , Drug Delivery Systems/methods , Poloxalene/chemistry , Poloxalene/pharmacology
6.
Lasers Med Sci ; 39(1): 141, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801600

ABSTRACT

PURPOSE: Conventional approaches for enhancing wound healing may not always yield satisfactory results. Instead, we test the effectiveness of a newly developed photodynamic therapy (PDT) that uses methylene blue (MB) loaded with polyethylene glycol (PEG) (MB-PEG) hydrogel to accelerate wound healing process in mice. METHODS: A dorsal skin incision with 6 mm punch which topically subjected to MB-PEG hydrogel and a low-level laser light of red light to assess the regeneration process of wounded skin. A total of 63 adult male CD1 mice divided into normal group (no treatment) and other wound groups received different treatments of laser (650 ± 5 nm and power intensity of 180 mW/cm2), MB-PEG, or PDT (MB-PEG followed by laser). The wound healing parameters were investigated by histological examination of the skin and measuring of proinflammatory cytokines at the early stage (48 h) and a late one on day 21. RESULTS: at 48 h, the score of tissue granulation, inflammation, and angiogenesis process were markedly improved in wounded groups that received MB + PEG combined with laser compared to the group treated with laser alone. On day 21, a significant improvement of the inflammation was detected in the group treated with MB + PEG plus laser compared to the other groups. At 48 h, the upregulated serum levels of tumor necrosis factor (TNF)-α and interleukin (IL)-1ß in the wound group were significantly (P < 0.001) reduced in the group treated with MB + PEG combined with laser. CONCLUSION: MB-PEG based hydrogel improves and accelerates wound closure in the context of laser compared to either single treatment.


Subject(s)
Methylene Blue , Photochemotherapy , Polyethylene Glycols , Skin , Wound Healing , Animals , Wound Healing/drug effects , Wound Healing/radiation effects , Mice , Photochemotherapy/methods , Methylene Blue/pharmacology , Male , Skin/radiation effects , Skin/drug effects , Skin/injuries , Hydrogels , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacology , Cytokines/metabolism
7.
Int J Nanomedicine ; 19: 4163-4180, 2024.
Article in English | MEDLINE | ID: mdl-38751660

ABSTRACT

Purpose: The study aimed to address the non-specific toxicity of cytotoxins (CTX) in liver cancer treatment and explore their combined application with the photosensitizer Ce6, co-loaded into carbonized Zn/Co bimetallic organic frameworks. The goal was to achieve controlled CTX release and synergistic photodynamic therapy, with a focus on evaluating anti-tumor activity against human liver cancer cell lines (Hep G2). Methods: Purified cobra cytotoxin (CTX) and photosensitizer Ce6 were co-loaded into carbonized Zn/Co bimetallic organic frameworks, resulting in RGD-PDA@C-ZIF@(CTX+Ce6). The formulation was designed with surface-functionalization using polydopamine and tumor-penetrating peptide RGD. This approach aimed to facilitate controlled CTX release and enhance the synergistic effect of photodynamic therapy. The accumulation of RGD-PDA@C-ZIF@(CTX+Ce6) at tumor sites was achieved through RGD's active targeting and the enhanced permeability and retention (EPR) effect. In the acidic tumor microenvironment, the porous structure of the metal-organic framework disintegrated, releasing CTX and Ce6 into tumor cells. Results: Experiments demonstrated that RGD-PDA@C-ZIF@(CTX+Ce6) nanoparticles, combined with near-infrared laser irradiation, exhibited optimal anti-tumor effects against human liver cancer cells. The formulation showcased heightened anti-tumor activity without discernible systemic toxicity. Conclusion: The study underscores the potential of utilizing metal-organic frameworks as an efficient nanoplatform for co-loading cytotoxins and photodynamic therapy in liver cancer treatment. The developed formulation, RGD-PDA@C-ZIF@(CTX+Ce6), offers a promising avenue for advancing the clinical application of cytotoxins in oncology, providing a solid theoretical foundation for future research and development.


Subject(s)
Indoles , Liver Neoplasms , Metal-Organic Frameworks , Photochemotherapy , Photosensitizing Agents , Zinc , Humans , Photochemotherapy/methods , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Liver Neoplasms/drug therapy , Zinc/chemistry , Zinc/pharmacology , Indoles/chemistry , Indoles/pharmacology , Indoles/administration & dosage , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/administration & dosage , Animals , Hep G2 Cells , Cobalt/chemistry , Cobalt/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Oligopeptides/pharmacokinetics , Polymers/chemistry , Mice , Cytotoxins/chemistry , Cytotoxins/pharmacology , Cytotoxins/pharmacokinetics , Mice, Nude , Mice, Inbred BALB C , Cell Survival/drug effects
8.
Int J Mol Sci ; 25(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38791462

ABSTRACT

Small interfering RNA (siRNA) has significant potential as a treatment for cancer by targeting specific genes or molecular pathways involved in cancer development and progression. The addition of siRNA to other therapeutic strategies, like photodynamic therapy (PDT), can enhance the anticancer effects, providing synergistic benefits. Nevertheless, the effective delivery of siRNA into target cells remains an obstacle in cancer therapy. Herein, supramolecular nanoparticles were fabricated via the co-assembly of natural histone and hyaluronic acid for the co-delivery of HMGB1-siRNA and the photosensitizer chlorin e6 (Ce6) into the MCF-7 cell. The produced siRNA-Ce6 nanoparticles (siRNA-Ce6 NPs) have a spherical morphology and exhibit uniform distribution. In vitro experiments demonstrate that the siRNA-Ce6 NPs display good biocompatibility, enhanced cellular uptake, and improved cytotoxicity. These outcomes indicate that the nanoparticles constructed by the co-assembly of histone and hyaluronic acid hold enormous promise as a means of siRNA and photosensitizer co-delivery towards synergetic therapy.


Subject(s)
Histones , Hyaluronic Acid , Nanoparticles , Photosensitizing Agents , RNA, Small Interfering , Hyaluronic Acid/chemistry , Humans , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Nanoparticles/chemistry , Histones/metabolism , MCF-7 Cells , Photochemotherapy/methods , Porphyrins/chemistry , Porphyrins/pharmacology , Chlorophyllides , Cell Survival/drug effects
9.
Biomacromolecules ; 25(6): 3671-3684, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38720431

ABSTRACT

Reactive oxygen species (ROS) have emerged as a promising treatment option for antibacterial and biofilm eradication. However, their therapeutic efficacy is significantly hampered by the unique microenvironments of diabetic wounds. In this study, we designed and synthesized porphyrin-based Fe covalent organic frameworks (Fe-COF) through a Schiff base condensation reaction. Subsequently, Fe-COF were encapsulated with hyaluronic acid (HA) through electrostatic adsorption, resulting in a novel formulation named HA-Fe-COF for diabetic wound healing. HA-Fe-COF were engineered to respond to hyaluronidase in the infected wound, leading to the controlled release of Fe-COF. Those released Fe-COF served a dual role as photosensitizers, generating singlet oxygen and localized heating when exposed to dual light sources. Additionally, they acted as peroxidase-like nanozymes, facilitating the production of ROS through enzymatic reactions. This innovative approach enabled a synergistic therapeutic effect combining photodynamic, photothermal, and chemodynamic modalities. Furthermore, the sustained release of HA from HA-Fe-COF promoted angiogenesis, collagen deposition, and re-epithelialization during the diabetic wound healing process. This "all-in-one" strategy offers a novel approach for the development of antimicrobial and biofilm eradication strategies that minimize damage to healthy tissues in vivo.


Subject(s)
Hyaluronic Acid , Metal-Organic Frameworks , Porphyrins , Wound Healing , Wound Healing/drug effects , Animals , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology , Mice , Reactive Oxygen Species/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Skin/drug effects , Humans , Wound Infection/drug therapy , Wound Infection/microbiology , Iron/chemistry , Photochemotherapy/methods , Hyaluronoglucosaminidase
10.
Biomater Adv ; 161: 213891, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38781738

ABSTRACT

An antitumour chemo-photodynamic therapy nanoplatform was constructed based on phospholipid-coated NaYF4: Yb/Er upconversion nanoparticles (UCNPs). In this work, the amphiphilic block copolymer DSPE-PEG2000 was combined with the surface ligand oleic acid of the UCNPs through hydrophobic interaction to form liposomes with a dense hydrophobic layer in which the photosensitizer hypocrellin B (HB) was assembled. The coated HB formed J-aggregates, which caused a large redshift in the absorption spectrum and improved the quantum efficiency of energy transfer. Furthermore, MnO2 nanosheets grew in-situ on the liposomes through OMn coordination. Therefore, a multifunctional tumour microenvironment (TME)-responsive theranostic nanoplatform integrating photodynamic therapy (PDT) and chemodynamic therapy (CDT) was successfully developed. The results showed that this NIR-mediated chemo-photodynamic therapy nanoplatform was highly efficient for oncotherapy.


Subject(s)
Manganese Compounds , Nanoparticles , Oxides , Perylene , Photochemotherapy , Photosensitizing Agents , Quinones , Photochemotherapy/methods , Perylene/analogs & derivatives , Perylene/pharmacology , Perylene/chemistry , Perylene/administration & dosage , Humans , Quinones/chemistry , Quinones/pharmacology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Oxides/chemistry , Oxides/pharmacology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/administration & dosage , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Animals , Phenol/chemistry , Phenol/pharmacology , Liposomes/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Mice , Cell Line, Tumor , Tumor Microenvironment/drug effects
11.
Lasers Med Sci ; 39(1): 133, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38771549

ABSTRACT

BACKGROUND: Tooth discoloration is a common concern in antimicrobial photodynamic therapy (aPDT) using various photosensitizers (PS). Toluidine Blue (TB), Methylene Blue (MB), Phthalocyanine (Pc), and 2-mercaptopyridine-substituted zinc phthalocyanine (TM-ZnPc) are among those studied, but their relative impacts on tooth discoloration remain unclear. AIM: This study aimed to compare the effects of TB, MB, Pc, and TM-ZnPc in aPDT on tooth discoloration, utilizing a controlled experimental setup. MATERIALS AND METHODS: The study comprised seventy-five single-rooted incisors with root canals. Following meticulous preparation, a standardized area on the crown surface was designated for examination, and precise measurements of the initial tooth colors were recorded. Samples were randomly divided into five groups: Negative control, MB, TM, Pc, and TM-ZnPc. Photoactivation was performed using LED light, and color measurements were taken at multiple time points up to 90 days. Data were converted to Lab* color values of the CIE Lab* color system (International Commission on Illumination, Vienna, Austria), and ΔE values were calculated. Statistical analysis was performed using Two-way ANOVA and Post-Hoc Tukey tests (p < 0.05). RESULTS: At day 7 and 30, TM-ZnPc and Pc caused less discoloration compared to MB and TB. TM-ZnPc caused more tooth discoloration compared to Pc (p < 0.05). Compared to baseline, MB and TM-ZnPc caused more tooth discoloration at 30 days and TB caused more tooth discoloration at 90 days (p < 0.05). No significant difference was observed in terms of tooth discoloration at all periods evaluated after Pc application (p > 0.05). All photosensitizers tested in the study caused tooth coloration. CONCLUSION: All PS induced clinically detectable tooth discoloration, with TB and MB causing more significant discoloration compared to Pc and TM-ZnPc at certain time points. TM-ZnPc and Pc demonstrated more stable coloration levels over time, suggesting their potential reliability in aPDT applications. This study highlights the importance of selecting appropriate PS to minimize tooth discoloration in aPDT, with Pc showing promise in this regard.


Subject(s)
Isoindoles , Methylene Blue , Photochemotherapy , Photosensitizing Agents , Spectrophotometry , Tolonium Chloride , Tooth Discoloration , Photochemotherapy/methods , Photochemotherapy/adverse effects , Photosensitizing Agents/administration & dosage , Humans , Tooth Discoloration/chemically induced , Methylene Blue/administration & dosage , Zinc Compounds , Indoles/adverse effects , Indoles/administration & dosage , Organometallic Compounds/administration & dosage , Organometallic Compounds/adverse effects
12.
J Colloid Interface Sci ; 670: 279-287, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38763024

ABSTRACT

Nanomedicines that combine reactive oxygen species (ROS)-responsive polyprodrug and photodynamic therapy have shown great potential for improving treatment efficacy. However, the consumption of ROS by overexpressed glutathione in tumor cells is a major obstacle for achieving effective ROS amplification and prodrug activation. Herein, we report a polyprodrug-based nanoparticle that can realize ROS amplification and cascaded drug release. The nanoparticle can respond to the high level of hydrogen peroxide in tumor microenvironment, achieving self-destruction and release of quinone methide. The quinone methide depletes intracellular glutathione and thus decreases the antioxidant capacity of cancer cells. Under laser irradiation, a large amount of ROS will be generated to induce cell damage and prodrug activation. Therefore, the glutathione-depleting polyprodrug nanoparticles can efficiently inhibit tumor growth by enhanced photodynamic therapy and cascaded locoregional chemotherapy.


Subject(s)
Antineoplastic Agents , Glutathione , Nanoparticles , Photochemotherapy , Prodrugs , Reactive Oxygen Species , Glutathione/metabolism , Glutathione/chemistry , Nanoparticles/chemistry , Prodrugs/pharmacology , Prodrugs/chemistry , Humans , Reactive Oxygen Species/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Animals , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Mice , Drug Screening Assays, Antitumor , Particle Size , Hydrogen Peroxide/metabolism , Cell Survival/drug effects , Cell Proliferation/drug effects , Surface Properties , Cell Line, Tumor , Drug Liberation , Tumor Microenvironment/drug effects , Indolequinones
13.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 433-440, 2024 Mar 20.
Article in Chinese | MEDLINE | ID: mdl-38645856

ABSTRACT

Objective: Port-wine stains are a kind of dermatological disease of congenital capillary malformation. Based on the biological characteristics of port-wine stains and the advantages of microneedle transdermal administration, we intend to construct a nanodrug co-loaded with rapamycin (RPM), an anti-angiogenesis drug, and photochlor (HPPH), a photosensitizer, and integrate the nanodrug with dissolvable microneedles (MN) to achieve anti-angiogenesis and photodynamic combination therapy for port-wine stains. Methods: First, RPM and HPPH co-loaded nanoparticles (RPM-HPPH NP) were prepared by the emulsification solvent-volatilization method, and its ability to generate reactive oxygen species (ROS) was investigated under 660 nm laser irradiation. Mouse hemangioendothelioma endothelial cells (EOMA) were used as the subjects of the study. The cellular uptake behaviors were examined by fluorescence microscopy and flow cytometry. The cytotoxicity effects of RPM-HPPH NP with or without 660 nm laser irradiation on EOMA cells were examined by MTT assays (with free RPM serving as the control). Then, hyaluronic acid (HA) dissolvable microneedles loaded with RPM-HPPH NP (RPM-HPPH NP@HA MN) were obtained by compounding the nanodrug with HA dissolvable microneedle system through the molding method. The morphological characteristics and mechanical properties of RPM-HPPH NP@HA MN were investigated by scanning electron microscope and electronic universal testing machine. The penetration ability of RPM-HPPH NP@HA MN on the skin of nude mice was evaluated by trypan blue staining and H&E staining experiment. Results: The RPM-HPPH NP prepared in the study had a particle size of 150 nm and generated large amounts of ROS under laser irradiation. At the cellular level, RPM-HPPH NP was taken up by EOMA cells in a time-dependent manner. The cytotoxicity of RPM-HPPH NP was higher than that of free RPM with or without laser irradiation. Under laser irradiation, RPM-HPPH NP exhibited stronger cytotoxic effects and the difference was statistically significant (P<0.05). The height of the needle tip of RPM-HPPH NP@HA MN was 600 µm and the mechanical property of a single needle was 0.75048 N. Trypan blue staining and HE staining showed that pressing on the microneedles could produce pores on the skin surface and penetration of the stratum corneum. Conclusion: RPM-HPPH NP@HA MN can deliver RPM-HPPH NP percutaneously to the lesion tissue and realize the synergistic treatment of port-wine stains with anti-angiogenic therapy and photodynamic therapy, providing a new strategy for the construction of nanodrug-loaded microneedle delivery system and the clinical treatment of port-wine stains.


Subject(s)
Nanoparticles , Needles , Port-Wine Stain , Sirolimus , Animals , Mice , Nanoparticles/chemistry , Port-Wine Stain/drug therapy , Sirolimus/administration & dosage , Photosensitizing Agents/administration & dosage , Administration, Cutaneous , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Endothelial Cells/drug effects , Drug Delivery Systems , Angiogenesis Inhibitors/administration & dosage , Hemangioendothelioma/drug therapy
14.
Biomater Sci ; 12(10): 2639-2647, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38563394

ABSTRACT

Triple negative breast cancer (TNBC) exhibits limited responsiveness to immunotherapy owing to its immunosuppressive tumor microenvironment (TME). Here, a reactive oxygen species (ROS)-labile nanodrug encapsulating the photosensitizer Ce6 and Bcl-2 inhibitor ABT-737 was developed to provoke a robust immune response via the synergistic effect of photodynamic therapy (PDT) and the reversal of apoptosis resistance. Upon exposure to first-wave near-infrared laser irradiation, the generated ROS triggers PEG cleavage, facilitating the accumulation of the nanodrug at tumor region and endocytosis by tumor cells. Further irradiation leads to the substantial generation of cytotoxic ROS, initiating an immunogenic cell death (ICD) cascade, which prompts the maturation of dendritic cells (DCs) as well as the infiltration of T cells into the tumor site. Meanwhile, Bcl-2 inhibition counteracts apoptosis resistance, thereby amplifying PDT-induced ICD and bolstering antitumor immunity. As a result, the ROS-sensitive nanodrug demonstrates a potent inhibitory effect on tumor growth.


Subject(s)
Apoptosis , Biphenyl Compounds , Immunotherapy , Photochemotherapy , Photosensitizing Agents , Reactive Oxygen Species , Sulfonamides , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Humans , Apoptosis/drug effects , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/administration & dosage , Female , Reactive Oxygen Species/metabolism , Animals , Mice , Biphenyl Compounds/pharmacology , Biphenyl Compounds/chemistry , Sulfonamides/pharmacology , Sulfonamides/chemistry , Chlorophyllides , Cell Line, Tumor , Piperazines/pharmacology , Piperazines/chemistry , Nitrophenols/pharmacology , Nitrophenols/chemistry , Nanoparticles/chemistry , Porphyrins/pharmacology , Porphyrins/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry
15.
Biomed Pharmacother ; 174: 116586, 2024 May.
Article in English | MEDLINE | ID: mdl-38626516

ABSTRACT

Cancer treatment is presently a significant challenge in the medical domain, wherein the primary modalities of intervention include chemotherapy, radiation therapy and surgery. However, these therapeutic modalities carry side effects. Photothermal therapy (PTT) and photodynamic therapy (PDT) have emerged as promising modalities for the treatment of tumors in recent years. Phototherapy is a therapeutic approach that involves the exposure of materials to specific wavelengths of light, which can subsequently be converted into either heat or Reactive Oxygen Species (ROS) to effectively eradicate cancer cells. Due to the hydrophobicity and lack of targeting of many photoresponsive materials, the use of nano-carriers for their transportation has been extensively explored. Among these nanocarriers, liposomes have been identified as an effective drug delivery system due to their controllability and availability in the biomedical field. By binding photoresponsive materials to liposomes, it is possible to reduce the cytotoxicity of the material and regulate drug release and accumulation at the tumor site. This article provides a comprehensive review of the progress made in cancer therapy using photoresponsive materials loaded onto liposomes. Additionally, the article discusses the potential synergistic treatment through the combination of phototherapy with chemo/immuno/gene therapy using liposomes.


Subject(s)
Liposomes , Neoplasms , Photochemotherapy , Humans , Neoplasms/therapy , Neoplasms/drug therapy , Animals , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Drug Delivery Systems/methods , Phototherapy/methods , Photothermal Therapy/methods
16.
J Control Release ; 370: 168-181, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643936

ABSTRACT

The high prevalence and severity of hepatocellular carcinoma (HCC) present a significant menace to human health. Despite the significant advancements in nanotechnology-driven antineoplastic agents, there remains a conspicuous gap in the development of targeted chemotherapeutic agents specifically designed for HCC. Consequently, there is an urgent need to explore potent drug delivery systems for effective HCC treatment. Here we have exploited the interplay between HCC and adipocyte to engineer a hybrid adipocyte-derived exosome platform, serving as a versatile vehicle to specifically target HCC and exsert potent antitumor effect. A lipid-like prodrug of docetaxel (DSTG) with a reactive oxygen species (ROS)-cleavable linker, and a lipid-conjugated photosensitizer (PPLA), spontaneously co-assemble into nanoparticles, functioning as the lipid cores of the hybrid exosomes (HEMPs and NEMPs). These nanoparticles are further encapsuled within adipocyte-derived exosome membranes, enhancing their affinity towards HCC cancer cells. As such, cancer cell uptakes of hybrid exosomes are increased up to 5.73-fold compared to lipid core nanoparticles. Our in vitro and in vivo experiments have demonstrated that HEMPs not only enhance the bioactivity of the prodrug and extend its circulation in the bloodstream but also effectively inhibit tumor growth by selectively targeting hepatocellular carcinoma tumor cells. Self-facilitated synergistic drug release subsequently promoting antitumor efficacy, inducing significant inhibition of tumor growth with minimal side effects. Our findings herald a promising direction for the development of targeted HCC therapeutics.


Subject(s)
Adipocytes , Antineoplastic Agents , Carcinoma, Hepatocellular , Docetaxel , Exosomes , Liver Neoplasms , Nanoparticles , Exosomes/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Animals , Liver Neoplasms/drug therapy , Liver Neoplasms/therapy , Liver Neoplasms/pathology , Humans , Docetaxel/administration & dosage , Docetaxel/pharmacology , Docetaxel/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Adipocytes/drug effects , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Prodrugs/administration & dosage , Prodrugs/therapeutic use , Cell Line, Tumor , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Mice, Nude , Phototherapy/methods , Drug Delivery Systems , Mice , Reactive Oxygen Species/metabolism , Mice, Inbred BALB C
17.
Int J Pharm ; 657: 124170, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38679244

ABSTRACT

Improving the anticancer efficacy of chemotherapeutic drugs and photosensitizers requires innovative multifunctional nanoplatforms. This study introduces a chemo- and phototherapeutic drug delivery system (DDS) based on poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs), both PEGylated and non-PEGylated, with a mean size of 200 ± 75 nm. Colchicine (Colch) and purpurin18 (P18) were co-encapsulated into these NPs, and their in vitro drug release profiles were investigated. The anticancer potential of these systems was evaluated across various cell lines (i.e., CaCo-2, PC-3, MCF-7, and MRC-5 cells), demonstrating enhanced NP uptake by cancer cells compared to free drugs. Co-administration of Colch and P18 in 2D and 3D cell line models exhibited a synergistic effect, harnessing both chemotherapeutic and photodynamic effects, leading to higher cancer cell elimination efficacy. This newly developed multifunctional DDS presents a promising platform for combined chemo- and photodynamic therapy in cancer treatment.


Subject(s)
Colchicine , Drug Carriers , Drug Liberation , Polylactic Acid-Polyglycolic Acid Copolymer , Humans , Colchicine/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Drug Carriers/chemistry , Cell Line, Tumor , Spheroids, Cellular/drug effects , Nanoparticles/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Neoplasms/drug therapy , Cell Survival/drug effects , Drug Delivery Systems/methods
18.
J Control Release ; 370: 302-309, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38663752

ABSTRACT

Displaying antibodies on carrier surfaces facilitates precise targeting and delivery of drugs to diseased cells. Here, we report the synthesis of antibody-lipid conjugates (ALCs) through site-selective acetylation of Lys 248 in human Immunoglobulin G (IgG) and the development of antibody-functionalized red blood cells (immunoRBC) for targeted drug delivery. ImmunoRBC with the HER2-selective antibody trastuzumab displayed on the surface (called Tras-RBC) was constructed following a three-step procedure. First, a peptide-guided, proximity-induced reaction transferred an azidoacetyl group to the ε-amino group of Lys 248 in the Fc domain. Second, the azide-modified IgG was subsequently conjugated with dibenzocyclooctyne (DBCO)-functionalized lipids via strain-promoted azide-alkyne cycloaddition (SPAAC) to result in ALCs. Third, the lipid portion of ALCs was then inserted into the cell membranes, and IgGs were displayed on red blood cells (RBCs) to construct immunoRBCs. We then loaded Tras-RBC with a photosensitizer (PS), Zinc phthalocyanine (ZnPc), to selectively target HER2-overexpressing cells, release ZnPc into cancer cells following photolysis, and induce photodynamic cytotoxicity in the cancer cells. This work showcases assembling immunoRBCs following site-selective lipid conjugation on therapeutic antibodies and the targeted introduction of PS into cancer cells. This method could apply to the surface functionalization of other membrane-bound vesicles or lipid nanoparticles for antibody-directed drug delivery.


Subject(s)
Drug Delivery Systems , Erythrocytes , Indoles , Isoindoles , Lipids , Trastuzumab , Humans , Erythrocytes/drug effects , Trastuzumab/chemistry , Trastuzumab/administration & dosage , Lipids/chemistry , Indoles/chemistry , Indoles/administration & dosage , Zinc Compounds , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Organometallic Compounds/chemistry , Organometallic Compounds/administration & dosage , Receptor, ErbB-2/immunology , Immunoconjugates/chemistry , Immunoconjugates/administration & dosage , Immunoglobulin G/chemistry , Cell Line, Tumor , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/chemistry , Azides/chemistry
19.
J Control Release ; 369: 531-544, 2024 May.
Article in English | MEDLINE | ID: mdl-38580138

ABSTRACT

Stimulator of the interferon genes (STING) pathway is appealing but challenging to potentiate the innate anti-tumor immunity. In this work, nuclear-targeted chimeric peptide nanorods (designated as PFPD) are constructed to amplify innate immunity through localized DNA damage and STING activation. Among which, the chimeric peptide (PpIX-FFVLKPKKKRKV) is fabricated with photosensitizer and nucleus targeting peptide sequence, which can self-assemble into nanorods and load STING agonist of DMXAA. The uniform nanosize distribution and good stability of PFPD improve the sequential targeting delivery of drugs towards tumor cells and nuclei. Under light irradiation, PFPD produce a large amount of reactive oxygen species (ROS) to destroy nuclear DNA in situ, and the released cytosolic DNA fragment will efficiently activate innate anti-tumor immunity in combination with STING agonist. In vitro and in vivo results indicate the superior ability of PFPD to activate natural killer cells and T cells, thus efficiently eradicating lung metastatic tumor without inducing unwanted side effects. This work provides a sophisticated strategy for localized activation of innate immunity for systemic tumor treatment, which may inspire the rational design of nanomedicine for tumor precision therapy.


Subject(s)
DNA Damage , Immunity, Innate , Membrane Proteins , Animals , Immunity, Innate/drug effects , Humans , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacology , Cell Nucleus/metabolism , Mice, Inbred BALB C , Cell Line, Tumor , Nanotubes, Peptide/chemistry , Reactive Oxygen Species/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lung Neoplasms/drug therapy , Female , Mice , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Peptides/administration & dosage , Peptides/chemistry
20.
Biomed Pharmacother ; 174: 116486, 2024 May.
Article in English | MEDLINE | ID: mdl-38520865

ABSTRACT

Recurrence and metastasis of gastric cancer is a major therapeutic challenge for treatment. The presence of cancer stem cells (CSCs) is a major obstacle to the success of current cancer therapy, often leading to treatment resistance and tumor recurrence and metastasis. Therefore, it is important to develop effective strategies to eradicate CSCs. In this study, we developed a combined therapeutic strategy of photothermal therapy (PTT) and gastric cancer stem cells (GCSCs) inhibition by successfully synthesizing nanoliposomes loaded with IR780 (photosensitizer) and EN4 (c-Myc inhibitor). The nanocomposites are biocompatible and exhibit superior photoacoustic (PA) imaging properties. Under laser irradiation, IR780-mediated PTT effectively and rapidly killed tumor cells, while EN4 synergistically inhibited the self-renewal and stemness of GCSCs by suppressing the expression and activity of the pluripotent transcription factor c-Myc, preventing the tumor progression of gastric cancer. This Nano-EN-IR@Lip is expected to be a novel clinical nanomedicine for the integration of gastric cancer diagnosis, treatment and prevention.


Subject(s)
Liposomes , Neoplastic Stem Cells , Photosensitizing Agents , Photothermal Therapy , Stomach Neoplasms , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Stomach Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Humans , Photothermal Therapy/methods , Animals , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Indoles/pharmacology , Indoles/chemistry , Nanoparticles/chemistry , Mice, Nude , Combined Modality Therapy , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Nanocomposites/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...