Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.344
Filtrar
Más filtros











Intervalo de año de publicación
1.
Methods Mol Biol ; 2833: 51-56, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38949700

RESUMEN

Photodynamic therapy (PDT) is an established therapy used for the treatment of cutaneous skin cancers and other non-infective ailments. There has been recent interest in the opportunity to use aPDT (antimicrobial PDT) to treat skin and soft tissue infections. PDT utilizes photosensitizers that infiltrate all cells and "sensitize" them to a given wavelength of light. The photosensitizer is simply highly absorbent to a given wavelength of light and when excited will produce, in the presence of oxygen, damaging oxygen radicals and singlet oxygen. Bacterial cells are comparatively poor at combatting oxidative stress when compared with human cells therefore a degree of selective toxicity can be achieved with aPDT.In this chapter, we outline methodologies for testing aPDT in vitro using standard lab equipment.


Asunto(s)
Fotoquimioterapia , Fármacos Fotosensibilizantes , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Fotoquimioterapia/métodos , Humanos , Oxígeno Singlete/metabolismo , Antiinfecciosos/farmacología
2.
Int J Nanomedicine ; 19: 6377-6397, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38952677

RESUMEN

Background: How to ingeniously design multi-effect photosensitizers (PSs), including multimodal imaging and multi-channel therapy, is of great significance for highly spatiotemporal controllable precise phototherapy of malignant tumors. Methods: Herein, a novel multifunctional zinc(II) phthalocyanine-based planar micromolecule amphiphile (ZnPc 1) was successfully designed and synthesized, in which N atom with photoinduced electron transfer effect was introduced to enhance the near-infrared absorbance and nonradiative heat generation. After simple self-assembling into nanoparticles (NPs), ZnPc 1 NPs would exhibit enhanced multimodal imaging properties including fluorescence (FL) imaging (FLI) /photoacoustic (PA) imaging (PAI) /infrared (IR) thermal imaging, which was further used to guide the combined photodynamic therapy (PDT) and photothermal therapy (PTT). Results: It was that under the self-guidance of the multimodal imaging, ZnPc 1 NPs could precisely pinpoint the tumor from the vertical and horizontal boundaries achieving highly efficient and accurate treatment of cancer. Conclusion: Accordingly, the integration of FL/PA/IR multimodal imaging and PDT/PTT synergistic therapy pathway into one ZnPc 1 could provide a blueprint for the next generation of phototherapy, which offered a new paradigm for the integration of diagnosis and treatment in tumor and a promising prospect for precise cancer therapy.


Asunto(s)
Indoles , Isoindoles , Imagen Multimodal , Nanopartículas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Imagen Multimodal/métodos , Animales , Humanos , Indoles/química , Indoles/farmacología , Fotoquimioterapia/métodos , Nanopartículas/química , Ratones , Compuestos de Zinc/química , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Línea Celular Tumoral , Técnicas Fotoacústicas/métodos , Terapia Fototérmica/métodos , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Ratones Endogámicos BALB C , Fototerapia/métodos , Femenino
3.
Nat Commun ; 15(1): 5508, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951161

RESUMEN

Keratoconus, a disorder characterized by corneal thinning and weakening, results in vision loss. Corneal crosslinking (CXL) can halt the progression of keratoconus. The development of accelerated corneal crosslinking (A-CXL) protocols to shorten the treatment time has been hampered by the rapid depletion of stromal oxygen when higher UVA intensities are used, resulting in a reduced cross-linking effect. It is therefore imperative to develop better methods to increase the oxygen concentration within the corneal stroma during the A-CXL process. Photocatalytic oxygen-generating nanomaterials are promising candidates to solve the hypoxia problem during A-CXL. Biocompatible graphitic carbon nitride (g-C3N4) quantum dots (QDs)-based oxygen self-sufficient platforms including g-C3N4 QDs and riboflavin/g-C3N4 QDs composites (RF@g-C3N4 QDs) have been developed in this study. Both display excellent photocatalytic oxygen generation ability, high reactive oxygen species (ROS) yield, and excellent biosafety. More importantly, the A-CXL effect of the g-C3N4 QDs or RF@g-C3N4 QDs composite on male New Zealand white rabbits is better than that of the riboflavin 5'-phosphate sodium (RF) A-CXL protocol under the same conditions, indicating excellent strengthening of the cornea after A-CXL treatments. These lead us to suggest the potential application of g-C3N4 QDs in A-CXL for corneal ectasias and other corneal diseases.


Asunto(s)
Reactivos de Enlaces Cruzados , Grafito , Oxígeno , Puntos Cuánticos , Riboflavina , Puntos Cuánticos/química , Animales , Grafito/química , Oxígeno/metabolismo , Riboflavina/farmacología , Conejos , Masculino , Reactivos de Enlaces Cruzados/química , Compuestos de Nitrógeno/química , Especies Reactivas de Oxígeno/metabolismo , Queratocono/tratamiento farmacológico , Queratocono/metabolismo , Rayos Ultravioleta , Córnea/efectos de los fármacos , Córnea/metabolismo , Córnea/patología , Humanos , Fármacos Fotosensibilizantes/farmacología , Sustancia Propia/metabolismo , Sustancia Propia/efectos de los fármacos
4.
Anal Chim Acta ; 1316: 342860, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38969429

RESUMEN

BACKGROUND: Glutathione (GSH), a highly abundant thiol compound within cells, plays a critical role in physiological processes and exhibits close correlation with cancer. Among molecular imaging technologies, most probes have relatively short emission wavelengths and lack photoacoustic imaging (PA) capability, resulting in the inability to obtain tissue images with high penetration depth. The presence of GSH in the tumor microenvironment neutralizes ROS, diminishing the therapeutic effect of PDT, thus resulting in often unsatisfactory therapeutic efficacy. Therefore, it is imperative to develop a dual-modal probe for the detection of GSH and the diagnosis and treatment of cancer. RESULTS: In this study, we synthesized a novel dual-modal probe, Cy-Bio-GSH, utilizing near-infrared fluorescence (NIRF) and photoacoustic (PA) imaging techniques for GSH detection. The probe integrates cyanine dye as the fluorophore, nitroazobenzene as the recognition moiety, and biotin as the tumor-targeting moiety. Upon reacting with GSH, the probe emits NIR fluorescence at 820 nm and generates a PA signal. Significantly, this reaction activates the photodynamic and photothermal properties of the probe. By depleting GSH and employing a synergistic photothermal therapy (PTT) treatment, the therapeutic efficacy of photodynamic therapy (PDT) is remarkably enhanced. In-vivo experiments confirm the capability of the probe to detect GSH via NIRF and PA imaging. Notably, the combined tumor-targeting ability and PDT/PTT synergistic therapy enhance therapeutic outcomes for tumors and facilitate their ablation. SIGNIFICANCE: A novel tumor-targeting and dual-modal imaging probe (Cy-Bio-GSH) is synthesized, exhibiting remarkable sensitivity and selectivity to GSH, enabling the visualization of GSH in cells and the differentiation between normal and cancer cells. Cy-Bio-GSH enhances PDT/PTT with effective killing of cancer cells and makes the ablation of tumors in mice. This work represents the first tumor-targeting probe for GSH detection, and provides crucial tool for cancer diagnosis and treatment by dual-modal imaging with improved PDT/PTT synergistic therapy.


Asunto(s)
Biotina , Glutatión , Técnicas Fotoacústicas , Fotoquimioterapia , Glutatión/química , Glutatión/metabolismo , Animales , Humanos , Ratones , Biotina/química , Colorantes Fluorescentes/química , Colorantes Fluorescentes/síntesis química , Imagen Óptica , Femenino , Terapia Fototérmica , Ratones Desnudos , Ratones Endogámicos BALB C , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/uso terapéutico
5.
Nanotheranostics ; 8(4): 442-457, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38961886

RESUMEN

The global incidence of cancer continues to rise, posing a significant public health concern. Although numerous cancer therapies exist, each has limitations and complications. The present study explores alternative cancer treatment approaches, combining hyperthermia and photodynamic therapy (PDT). Magnetic nanoparticles (MNPs) and amine-functionalized carbon quantum dots (A-CQDs) were synthesized separately and then covalently conjugated to form a single nanosystem for combinational therapy (M-CQDs). The successful conjugation was confirmed using zeta potential, Fourier transform infrared spectroscopy (FT-IR), and UV-visible spectroscopy. Morphological examination in transmission electron microscopy (TEM) further verified the conjugation of CQDs with MNPs. Energy dispersive X-ray spectroscopy (EDX) revealed that M-CQDs contain approximately 12 weight percentages of carbon. Hyperthermia studies showed that both MNP and M-CQDs maintain a constant therapeutic temperature at lower frequencies (260.84 kHz) with high specific absorption rates (SAR) of 118.11 and 95.04 W/g, respectively. In vitro studies demonstrated that MNPs, A-CQDs, and M-CQDs are non-toxic, and combinational therapy (PDT + hyperthermia) resulted in significantly lower cell viability (~4%) compared to individual therapies. Similar results were obtained with Hoechst and propidium iodide (PI) staining assays. Hence, the combination therapy of PDT and hyperthermia shows promise as a potential alternative to conventional therapies, and it could be further explored in combination with existing conventional treatments.


Asunto(s)
Carbono , Hipertermia Inducida , Nanopartículas de Magnetita , Neoplasias , Fotoquimioterapia , Puntos Cuánticos , Puntos Cuánticos/química , Fotoquimioterapia/métodos , Humanos , Carbono/química , Hipertermia Inducida/métodos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Línea Celular Tumoral , Terapia Combinada , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología
6.
Lasers Med Sci ; 39(1): 172, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38965092

RESUMEN

The study utilized 5-ALA-PDT to treat patients with CIN or VaIN and assessed their clinical response, HPV clearance, and influencing factors after photodynamic therapy (PDT). This study involved 56 patients who received 5-ALA-PDT in a single center from May 2020 to March 2022, including 12 patients with CIN, 30 patients with VaIN, and 14 patients with both CIN and VaIN. Follow-up were conducted within 6 and 12 months after treatment to evaluate the clinical effectiveness of PDT. The assessment criteria included histological response (ER, elimination rate, RR, regression rate) and HPV clearance. Additionally, factors that could potentially influence the outcomes were analyzed. After PDT, the histological response showed an ER of 48.2% (27/56) and a RR of 80.4% (45/56) within 6 months of follow-up. The elimination rate increased to 69.6% (39/56) within 12 months, along with a regression rate of 82.1% (46/56). The rates of HPV clearance were observed to be 37.5% (21/56) and 44.6% (25/56) within 6 and 12 months, respectively. The study also revealed that HPV clearance significantly influenced histologic elimination within 6 months (p < 0.001) and histologic regression within 12 months (p < 0.01). Furthermore, premenopausal women exhibited a higher HPV clearance rate compared to postmenopausal women (61.5% vs. 30.0%, p = 0.036). 5-ALA PDT can be considered as an available option for the treatment of lower genital squamous intraepithelial lesions. The efficacy of its histologic response depends on HPV clearance. Additionally, it has been found that premenopausal women may benefit more from this treatment.


Asunto(s)
Ácido Aminolevulínico , Fotoquimioterapia , Fármacos Fotosensibilizantes , Humanos , Femenino , Fotoquimioterapia/métodos , Ácido Aminolevulínico/uso terapéutico , Ácido Aminolevulínico/administración & dosificación , Adulto , Fármacos Fotosensibilizantes/uso terapéutico , Fármacos Fotosensibilizantes/administración & dosificación , Persona de Mediana Edad , Resultado del Tratamiento , Displasia del Cuello del Útero/tratamiento farmacológico , Displasia del Cuello del Útero/virología , Displasia del Cuello del Útero/patología , Infecciones por Papillomavirus/tratamiento farmacológico , Infecciones por Papillomavirus/virología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/virología , Neoplasias del Cuello Uterino/patología , Adulto Joven , Anciano
7.
BMC Microbiol ; 24(1): 246, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970013

RESUMEN

Previous studies have shown that antimicrobial photodynamic inactivation (aPDI) can be strongly potentiated by the addition of the non-toxic inorganic salt, potassium iodide (KI). This approach was shown to apply to many different photosensitizers, including the xanthene dye Rose Bengal (RB) excited by green light (540 nm). Rose Bengal diacetate (RBDA) is a lipophilic RB derivative that is easily taken up by cells and hydrolyzed to produce an active photosensitizer. Because KI is not taken up by microbial cells, it was of interest to see if aPDI mediated by RBDA could also be potentiated by KI. The addition of 100 mM KI strongly potentiated the killing of Gram-positive methicillin-resistant Staphylocccus aureus, Gram-negative Eschericia coli, and fungal yeast Candida albicans when treated with RBDA (up to 15 µM) for 2 hours followed by green light (540 nm, 10 J/cm2). Both RBDA aPDI regimens (400 µM RBDA with or without 400 mM KI followed by 20 J/cm2 green light) accelerated the healing of MRSA-infected excisional wounds in diabetic mice, without damaging the host tissue.


Asunto(s)
Candida albicans , Staphylococcus aureus Resistente a Meticilina , Fármacos Fotosensibilizantes , Yoduro de Potasio , Rosa Bengala , Infecciones Estafilocócicas , Cicatrización de Heridas , Animales , Rosa Bengala/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Yoduro de Potasio/farmacología , Ratones , Candida albicans/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Escherichia coli/efectos de los fármacos , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Experimental/tratamiento farmacológico , Fotoquimioterapia/métodos , Sinergismo Farmacológico , Luz , Masculino
8.
ACS Nano ; 18(26): 17086-17099, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38952327

RESUMEN

Traditional external field-assisted therapies, e.g., microwave (MW) therapy and phototherapy, cannot effectively and minimally damage eliminate deep-seated infection, owing to the poor penetrability of light and low reactive oxygen species (ROS) stimulation capability of MW. Herein, an implantable and wireless-powered therapeutic platform (CNT-FeTHQ-TS), in which external MW can be converted into internal light via MW wireless-powered light-emitting chips, is designed to eradicate deep-seated tissue infections by MW-induced deep-seated photodynamic therapy. In application, CNT-FeTHQ-TS is implanted at internal lesions, and the chip emits light under external MW irradiation. Subsequently, CNT-FeTHQ coating in the platform can respond to both MW and light simultaneously to generate ROS and MW-hyperthermia for rapid and precise sterilization at focus. Importantly, MW also improves the photodynamic performance of CNT-FeTHQ by introducing vacancies in FeTHQ to facilitate the photoexcitation process and changing the spin state of electrons to inhibit the complexation of photogenerated electron-hole pairs, which were confirmed by simulation calculations and in situ MW-irradiated photoluminescence experiments. In vivo, CNT-FeTHQ-TS can effectively cure mice with Staphylococcus aureus infection in dorsal subcutaneous tissue. This work overcomes the key clinical limitations of safe energy transmission and conversion for treating deep-seated infections.


Asunto(s)
Microondas , Fotoquimioterapia , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Tecnología Inalámbrica , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Luz , Staphylococcus aureus/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Ratones Endogámicos BALB C , Antibacterianos/farmacología , Antibacterianos/química
9.
J Nanobiotechnology ; 22(1): 375, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926721

RESUMEN

As an emerging cancer treatment strategy, reactive oxygen species-based tumor catalytic therapies face enormous challenges due to hypoxia and the overexpression of glutathione (GSH) in the tumor microenvironment. Herein, a self-assembled copper-based nanoplatform, TCCHA, was designed for enzyme-like catalysis-enhanced chemodynamic/photodynamic/antiangiogenic tritherapy against hepatocellular carcinoma. TCCHA was fabricated from Cu2+, 3,3'-dithiobis (propionohydrazide), and photosensitizer chlorine e6 via a facile one-pot self-assembly strategy, after which an aldehyde hyaluronic acid was coated, followed by loading of the antivascular drug AL3818. The obtained TCCHA nanoparticles exhibited pH/GSH dual-responsive drug release behaviors and multienzymatic activities, including Fenton, glutathione peroxidase-, and catalase-like activities. TCCHA, a redox homeostasis disruptor, promotes ⋅OH generation and GSH depletion, thus increasing the efficacy of chemodynamic therapy. TCCHA, which has catalase-like activity, can also reinforce the efficacy of photodynamic therapy by amplifying O2 production. In vivo, TCCHA efficiently inhibited tumor angiogenesis and suppressed tumor growth without apparent systemic toxicity. Overall, this study presents a facile strategy for the preparation of multienzyme-like nanoparticles, and TCCHA nanoparticles display great potential for enzyme catalysis-enhanced chemodynamic/photodynamic/antiangiogenic triple therapy against cancer.


Asunto(s)
Carcinoma Hepatocelular , Cobre , Neoplasias Hepáticas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Cobre/química , Cobre/farmacología , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Fotoquimioterapia/métodos , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Humanos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Ratones Endogámicos BALB C , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/química , Porfirinas/química , Porfirinas/farmacología , Clorofilidas , Glutatión/metabolismo , Nanopartículas/química , Catálisis , Nanopartículas del Metal/química , Liberación de Fármacos , Ratones Desnudos , Antineoplásicos/farmacología , Antineoplásicos/química
10.
Int J Mol Sci ; 25(12)2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38928126

RESUMEN

Photodynamic Therapy (PDT) is recognized for its exceptional effectiveness as a promising cancer treatment method. However, it is noted that overexposure to the dosage and sunlight in traditional PDT can result in damage to healthy tissues, due to the low tumor selectivity of currently available photosensitizers (PSs). To address this challenge, we introduce herein a new strategy where the small molecule-targeted agent, erlotinib, is integrated into a boron dipyrromethene (BODIPY)-based PS to form conjugate 6 to enhance the precision of PDT. This conjugate demonstrates optical absorption, fluorescence emission, and singlet oxygen generation efficiency comparable to the reference compound 7, which lacks erlotinib. In vitro studies reveal that, after internalization, conjugate 6 predominantly accumulates in the lysosomes of HepG2 cells, exhibiting significant photocytotoxicity with an IC50 value of 3.01 µM. A distinct preference for HepG2 cells over HELF cells is observed with conjugate 6 but not with compound 7. In vivo experiments further confirm that conjugate 6 has a specific affinity for tumor tissues, and the combination treatment of conjugate 6 with laser illumination can effectively eradicate H22 tumors in mice with outstanding biosafety. This study presents a novel and potential PS for achieving precise PDT against cancer.


Asunto(s)
Clorhidrato de Erlotinib , Neoplasias Hepáticas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Porfobilinógeno , Humanos , Fotoquimioterapia/métodos , Animales , Ratones , Porfobilinógeno/análogos & derivados , Porfobilinógeno/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Células Hep G2 , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Clorhidrato de Erlotinib/farmacología , Clorhidrato de Erlotinib/química , Compuestos de Boro/química , Compuestos de Boro/farmacología
11.
J Inorg Biochem ; 258: 112623, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38823065

RESUMEN

A strategy for cancer treatment was implemented, based on chemo-photodynamic therapy, utilizing a novel formulation, low-cost system called Cas-ZnONPs. This system consisted of the incorporation of Casiopeina III-ia (CasIII-ia), a hydrophilic copper coordination compound with well-documented anti-neoplastic activity, on Zinc oxide nanoparticles (ZnONPs) with apoptotic activity and lipophilicity, allowing them to permeate biological barriers. Additionally, ZnONPs exhibited fluorescence, with emission at different wavelengths depending on their agglomeration and enabling real-time tracking biodistribution. Also, ZnONPs served as a sensitizer, generating reactive oxygen species (ROS) in situ. In in vitro studies on HeLa and MDA-MB-231 cell lines, a synergistic effect was observed with the impregnated CasIII-ia on ZnONPs. The anticancer activity had an increase in cellular inhibition, depending on the dose of exposure to UV-vis irradiation. In in vivo studies utilized zebrafish models for xenotransplanting stained MDA-MB-231 cells and testing the effectiveness of Cas-ZnONPs treatment. The treatment successfully eliminated cancer cells, both when combined with Photodynamic Therapy (PDT) and when used alone. However, a significantly higher concentration (50 times) of Cas-ZnONPs was required in the absence of PDT. This demonstrates the potential of Cas-ZnONPs in cancer treatment, especially when combined with PDT.


Asunto(s)
Antineoplásicos , Fotoquimioterapia , Pez Cebra , Humanos , Fotoquimioterapia/métodos , Animales , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Óxido de Zinc/química , Óxido de Zinc/farmacología , Células HeLa , Especies Reactivas de Oxígeno/metabolismo , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Línea Celular Tumoral , Nanopartículas/química , Apoptosis/efectos de los fármacos , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Cobre/química
12.
J Mater Chem B ; 12(25): 6175-6189, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38831689

RESUMEN

In this study, the heavy-atom-free BODIPY dendrimer TM4-BDP was synthesized for near-infrared photodynamic therapy, and was composed of a triphenylamine-BODIPY dimer and four 1-(2-morpholinoethyl)-1H-indole-3-ethenyl groups. The TM4-BDP could achieve near-infrared photodynamic therapy through two different photosensitive pathways, which include one-photon excitation at 660 nm and two-photon excitation at 1000 nm. In the one-photon excitation pathway, the TM4-BDP could generate singlet oxygen and superoxide radicals under 660 nm illumination. In addition, the one-photon PDT experiment in human nasopharyngeal carcinoma (CNE-2) cells also indicated that the TM4-BDP could specifically accumulate in lysosomes and show great cell phototoxicity with an IC50 of 22.1 µM. In the two-photon excitation pathway, the two-photon absorption cross-section at 1030 nm of TM4-BDP was determined to be 383 GM, which means that it could generate reactive oxygen species (ROS) under 1000 nm femtosecond laser excitation. Moreover, the two-photon PDT experiment in zebrafish also indicated the TM4-BDP could be used for two-photon fluorescence imaging and two-photon induced ROS generation in biological environments. Furthermore, in terms of the ROS generation mechanism, the TM4-BDP employed a novel spin-vibronic coupling intersystem crossing (SV-ISC) process for the mechanism of ROS generation and the femtosecond transient absorption spectra indicated that this novel SV-ISC mechanism was closely related to its charge transfer state lifetime. These above experiments of TM4-BDP demonstrate that the dendrimer design is an effective strategy for constructing heavy-atom-free BODIPY photosensitizers in the near-infrared region and lay the foundation for two-photon photodynamic therapy in future clinical trials.


Asunto(s)
Compuestos de Boro , Dendrímeros , Fotoquimioterapia , Fotones , Fármacos Fotosensibilizantes , Pez Cebra , Animales , Compuestos de Boro/química , Compuestos de Boro/farmacología , Compuestos de Boro/síntesis química , Dendrímeros/química , Dendrímeros/farmacología , Dendrímeros/síntesis química , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/síntesis química , Humanos , Estructura Molecular , Especies Reactivas de Oxígeno/metabolismo , Supervivencia Celular/efectos de los fármacos , Línea Celular Tumoral
13.
J Control Release ; 371: 406-428, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38849093

RESUMEN

Preventing the recurrence of melanoma after surgery and accelerating wound healing are among the most challenging aspects of melanoma management. Photothermal therapy has been widely used to treat tumors and bacterial infections and promote wound healing. Owing to its efficacy and specificity, it may be used for postoperative management of tumors. However, its use is limited by the uncontrollable distribution of photosensitizers and the likelihood of damage to the surrounding normal tissue. Hydrogels provide a moist environment with strong biocompatibility and adhesion for wound healing owing to their highly hydrophilic three-dimensional network structure. In addition, these materials serve as excellent drug carriers for tumor treatment and wound healing. It is possible to combine the advantages of both of these agents through different loading modalities to provide a powerful platform for the prevention of tumor recurrence and wound healing. This review summarizes the design strategies, research progress and mechanism of action of hydrogels used in photothermal therapy and discusses their role in preventing tumor recurrence and accelerating wound healing. These findings provide valuable insights into the postoperative management of melanoma and may guide the development of promising multifunctional hydrogels for photothermal therapy.


Asunto(s)
Hidrogeles , Melanoma , Terapia Fototérmica , Cicatrización de Heridas , Hidrogeles/química , Hidrogeles/administración & dosificación , Humanos , Melanoma/terapia , Terapia Fototérmica/métodos , Animales , Cicatrización de Heridas/efectos de los fármacos , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/uso terapéutico , Portadores de Fármacos/química , Recurrencia Local de Neoplasia/prevención & control
14.
J Control Release ; 371: 470-483, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38849094

RESUMEN

Hypoimmunogenicity and the immunosuppressive microenvironment of ovarian cancer severely restrict the capability of immune-mediated tumor killing. Immunogenic cell death (ICD) introduces a theoretical principle for antitumor immunity by increasing antigen exposure and presentation. Despite recent research progress, the currently available ICD inducers are still very limited, and many of them can hardly induce sufficient ICD based on traditional endoplasmic reticulum (ER) stress. Accumulating evidence indicates that inducing mitochondrial stress usually shows a higher efficiency in evoking large-scale ICD than that via ER stress. Inspired by this, herein, a mitochondria-targeted polyprodrug nanoparticle (named Mito-CMPN) serves as a much superior ICD inducer, effectively inducing chemo-photodynamic therapy-caused mitochondrial stress in tumor cells. The rationally designed stimuli-responsive polyprodrugs, which can self-assemble into nanoparticles, were functionalized with rhodamine B for mitochondrial targeting, cisplatin and mitoxantrone (MTO) for synergistic chemo-immunotherapy, and MTO also serves as a photosensitizer for photodynamic immunotherapy. The effectiveness and robustness of Mito-CMPNs in reversing the immunosuppressive microenvironment is verified in both an ovarian cancer subcutaneous model and a high-grade serous ovarian cancer model. Our results support that the induction of abundant ICD by focused mitochondrial stress is a highly effective strategy to improve the therapeutic efficacy of immunosuppressive ovarian cancer.


Asunto(s)
Antineoplásicos , Mitocondrias , Nanopartículas , Neoplasias Ováricas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Mitocondrias/efectos de los fármacos , Fotoquimioterapia/métodos , Animales , Humanos , Línea Celular Tumoral , Fármacos Fotosensibilizantes/administración & dosificación , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Profármacos/administración & dosificación , Profármacos/uso terapéutico , Profármacos/farmacología , Muerte Celular Inmunogénica/efectos de los fármacos , Ratones Endogámicos BALB C , Cisplatino/farmacología , Cisplatino/administración & dosificación , Cisplatino/uso terapéutico , Inmunoterapia/métodos , Microambiente Tumoral/efectos de los fármacos
15.
Turk J Ophthalmol ; 54(3): 120-126, 2024 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-38853628

RESUMEN

Objectives: To investigate the clinical efficacy and safety of the modified Cretan protocol in patients with post-laser in situ keratomileusis ectasia (PLE). Materials and Methods: In this retrospective study, 26 eyes of 16 patients with PLE were treated with the modified Cretan protocol (combined transepithelial phototherapeutic keratectomy and accelerated corneal collagen cross-linking). Visual, refractive, tomographic, and aberrometric outcomes and point spread function (PSF) were recorded preoperatively and at 6, 12, and 24 months after treatment. Results: Both uncorrected and best corrected visual acuity were stable at 24 months postoperatively compared to baseline (from 0.89±0.36 to 0.79±0.33 logarithm of the minimum angle of resolution [LogMAR] and 0.31±0.25 to 0.24±0.19 LogMAR, respectively, p>0.05 for all values). The mean K1, K2, Kmean, thinnest corneal thickness, and spherical aberration at baseline were 45.76±5.75 diopters (D), 48.62±6.17 D, 47.13±5.89 D, 433.16±56.86 µm, and -0.21±0.63 µm respectively. These values were reduced to 42.86±6.34 D, 45.92±6.74 D, 44.21±6.4 D, 391.07±54.76 µm, and -0.51±0.58 µm at 24 months postoperatively (p<0.001, p=0.002, p<0.001, p=0.001, and p=0.02, respectively). The mean spherical equivalent, manifest cylinder, Kmax, central corneal thickness, other corneal aberrations (root mean square, trefoil, coma, quatrefoil, astigmatism), and PSF remained stable (p>0.05 for all variables), while anterior and posterior elevation were significantly improved at 24 months postoperatively (p<0.001 and p=0.02, respectively). No surgical complications occurred during the 24-month follow-up. Conclusion: The modified Cretan protocol is a safe and effective treatment option for PLE patients that provides visual stabilization and significant improvement in topographic parameters during the 24-month follow-up. Further studies are needed to support our results.


Asunto(s)
Topografía de la Córnea , Reactivos de Enlaces Cruzados , Queratomileusis por Láser In Situ , Fármacos Fotosensibilizantes , Refracción Ocular , Agudeza Visual , Humanos , Estudios Retrospectivos , Queratomileusis por Láser In Situ/métodos , Queratomileusis por Láser In Situ/efectos adversos , Masculino , Femenino , Adulto , Dilatación Patológica/etiología , Refracción Ocular/fisiología , Reactivos de Enlaces Cruzados/uso terapéutico , Resultado del Tratamiento , Fármacos Fotosensibilizantes/uso terapéutico , Adulto Joven , Colágeno , Láseres de Excímeros/uso terapéutico , Estudios de Seguimiento , Riboflavina/uso terapéutico , Fotoquimioterapia/métodos , Enfermedades de la Córnea/cirugía , Enfermedades de la Córnea/etiología , Enfermedades de la Córnea/diagnóstico , Enfermedades de la Córnea/fisiopatología , Córnea/patología , Córnea/cirugía , Complicaciones Posoperatorias/diagnóstico , Miopía/cirugía , Miopía/fisiopatología , Rayos Ultravioleta
16.
Mol Pharm ; 21(7): 3296-3309, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38861020

RESUMEN

Cetuximab (Cet)-IRDye800CW, among other antibody-IRDye800CW conjugates, is a potentially effective tool for delineating tumor margins during fluorescence image-guided surgery (IGS). However, residual disease often leads to recurrence. Photodynamic therapy (PDT) following IGS is proposed as an approach to eliminate residual disease but suffers from a lack of molecular specificity for cancer cells. Antibody-targeted PDT offers a potential solution for this specificity problem. In this study, we show, for the first time, that Cet-IRDye800CW is capable of antibody-targeted PDT in vitro when the payload of dye molecules is increased from 2 (clinical version) to 11 per antibody. Cet-IRDye800CW (1:11) produces singlet oxygen, hydroxyl radicals, and peroxynitrite upon activation with 810 nm light. In vitro assays on FaDu head and neck cancer cells confirm that Cet-IRDye800CW (1:11) maintains cancer cell binding specificity and is capable of inducing up to ∼90% phototoxicity in FaDu cancer cells. The phototoxicity of Cet-IRDye800CW conjugates using 810 nm light follows a dye payload-dependent trend. Cet-IRDye800CW (1:11) is also found to be more phototoxic to FaDu cancer cells and less toxic in the dark than the approved chromophore indocyanine green, which can also act as a PDT agent. We propose that antibody-targeted PDT using high-payload Cet-IRDye800CW (1:11) could hold potential for eliminating residual disease postoperatively when using sustained illumination devices, such as fiber optic patches and implantable surgical bed balloon applicators. This approach could also potentially be applicable to a wide variety of resectable cancers that are amenable to IGS-PDT, using their respective approved full-length antibodies as a template for high-payload IRDye800CW conjugation.


Asunto(s)
Cetuximab , Indoles , Fotoquimioterapia , Humanos , Fotoquimioterapia/métodos , Indoles/química , Cetuximab/química , Cetuximab/farmacología , Línea Celular Tumoral , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Fármacos Fotosensibilizantes/química , Bencenosulfonatos
17.
J Mater Chem B ; 12(26): 6384-6393, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38845563

RESUMEN

Type I photodynamic therapy is considered to be a more promising cancer treatment than type II photodynamic therapy due to its non-oxygen-dependent characteristics. In this work, three D-A structure N,N'-dihydrophenazine (DHP)-based photosensitizers DP-CNPY, SMP-CNPY and DMP-CNPY were designed and synthesized by introducing different numbers of methyl groups in the backbone neighbor of DHP as the donor and combined with the typical strong electron acceptor 2-(pyridin-4-yl)acetonitrile. Among the three photosensitizers, SMP-CNPY with one methyl modification showed the best type I ROS (O2-˙, ˙OH) generation capacity and AIE performance. By encapsulation, SMP-CNPY was fabricated into nanoparticles, and SMP-CNPY NPs exhibited lipid droplet targeting ability with near-infrared (NIR) emission. Cell experiments have proved that SMP-CNPY NPs can effectively kill different kinds of cancer cells under normal oxygen conditions. Even under hypoxic and extreme hypoxic conditions, SMP-CNPY NPs can still produce ROS and kill cancer cells. This work holds significant potential in the field of type I AIE-active photosensitizers and provides a new strategy for overcoming the hypoxic dilemma in the malignant tumor microenvironment.


Asunto(s)
Gotas Lipídicas , Fotoquimioterapia , Fármacos Fotosensibilizantes , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/síntesis química , Humanos , Gotas Lipídicas/química , Gotas Lipídicas/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Especies Reactivas de Oxígeno/metabolismo , Nanopartículas/química , Tamaño de la Partícula , Imagen Óptica , Supervivencia Celular/efectos de los fármacos , Estructura Molecular , Ensayos de Selección de Medicamentos Antitumorales , Línea Celular Tumoral
18.
Photochem Photobiol Sci ; 23(7): 1361-1372, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38865066

RESUMEN

Colorectal cancer (CRC) is significantly contributed to global cancer mortality rates. Treating CRC is particularly challenging due to metastasis and drug resistance. There is a pressing need for new treatment strategies against metastatic CRC. Photodynamic therapy (PDT) offers a well-established, minimally invasive treatment option for cancer with limited side effects. Hypericin (HYP), a potent photosensitizer for PDT, has been documented to induce cytotoxicity and apoptosis in various types of cancers. However, there are few reports on the inhibitory effects of HYP-mediated PDT on the metastatic ability of CRC cells. Here, we evaluate the inhibitory effects of HYP-mediated PDT against metastatic CRC cells and define its underlying mechanisms. Wound-healing and Transwell assays show that HYP-mediated PDT suppresses migration and invasion of CRC cells. F-actin visualization assays indicate HYP-mediated PDT decreases F-actin formation in CRC cells. TEM assays reveal HYP-mediated PDT disrupts pseudopodia formation of CRC cells. Mechanistically, immunofluorescence and western blotting results show that HYP-mediated PDT upregulates E-cadherin and downregulates N-cadherin and Vimentin. HYP-mediated PDT also suppresses key EMT regulators, including Snail, MMP9, ZEB1 and α-SMA. Additionally, the expressions of RhoA and ROCK1 are downregulated by HYP-mediated PDT. Together, these findings suggest that HYP-mediated PDT inhibits the migration and invasion of HCT116 and SW620 cells by modulating EMT and RhoA-ROCK1 signaling pathway. Thus, HYP-mediated PDT presents a potential therapeutic option for CRC.


Asunto(s)
Antracenos , Neoplasias Colorrectales , Transición Epitelial-Mesenquimal , Perileno , Fotoquimioterapia , Fármacos Fotosensibilizantes , Transducción de Señal , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA , Humanos , Perileno/análogos & derivados , Perileno/farmacología , Perileno/química , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Antracenos/farmacología , Transducción de Señal/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Transición Epitelial-Mesenquimal/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Metástasis de la Neoplasia , Ensayos de Selección de Medicamentos Antitumorales
19.
J Mater Chem B ; 12(26): 6285-6304, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38895829

RESUMEN

Phototherapy, including photothermal therapy (PTT) and photodynamic therapy (PDT), has the advantages of spatiotemporal selectivity, non-invasiveness, and negligible drug resistance. Phototherapy has been approved for treating superficial epidermal tumors. However, its therapeutic efficacy is limited by the hypoxic tumor microenvironment and the highly expressed heat shock protein. Moreover, poor tissue penetration and focused irradiation laser region in phototherapy make treating deep tissues and metastatic tumors challenging. Combination therapy strategies, which integrate the advantages of each treatment and overcome their disadvantages, can significantly improve the therapeutic efficacy. Recently, many combination therapy strategies have been reported. Our study summarizes the strategies used for combining phototherapy with other cancer treatments such as chemotherapy, immunotherapy, sonodynamic therapy, gas therapy, starvation therapy, and chemodynamic therapy. Some research cases were selected to analyze the combination therapy effect, delivery platform feature, and synergetic anticancer mechanisms. Moreover, additional research cases are summarized in the tables. This review provides strong evidence that phototherapy-based combination strategies can enhance the anticancer effect compared with phototherapy alone. Additionally, the challenges and future perspectives associated with these combinational therapies are discussed.


Asunto(s)
Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Fototerapia/métodos , Terapia Combinada , Animales , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico
20.
Cancer Genomics Proteomics ; 21(4): 361-367, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38944426

RESUMEN

BACKGROUND/AIM: Breast cancer (BC) is the most common malignant disease worldwide. Localized stages of BC can be successfully treated by surgery. However, local recurrence occurs in about 4-10% of patients, requiring systemic treatments that impair the patients' quality of life and shortens life expectancy. Therefore, new therapeutic options are needed, which can be used intraoperatively and contribute to the complete removal of residual tumor cells in the surgical area. In the present study, we describe a cysteine-modified variant of the anti-HER2 antibody trastuzumab, that was coupled to the silicon phthalocyanine photosensitizer dye WB692-CB1 for the photoimmunotherapy (PIT) of BC. MATERIALS AND METHODS: The cysteine modified trastuzumab variant was cloned and expressed in Expi293F cells. After purification via immobilized affinity chromatography, the antibody was coupled to the dye. Cell binding of the antibody and the antibody dye conjugate was measured by flow cytometry. After incubation of BC cells with the conjugate and activation of the dye by irradiation with red light, cell viability was determined. RESULTS: The antibody and the conjugate showed specific binding to HER2-expressing BC cells. Treatment of the HER2high BC cell line SK-BR-3 with the conjugate followed by irradiation with a red light dose of 32 J/cm2 led to complete cell killing within 24 h. CONCLUSION: Our novel antibody dye conjugate represents a promising candidate for intraoperative treatment of localized BC, aiming to eliminate residual tumor cells in the surgical area and potentially reduce local recurrence, thereby improving recovery prospects for BC patients.


Asunto(s)
Neoplasias de la Mama , Inmunoterapia , Receptor ErbB-2 , Trastuzumab , Humanos , Neoplasias de la Mama/terapia , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Femenino , Receptor ErbB-2/metabolismo , Inmunoterapia/métodos , Trastuzumab/farmacología , Trastuzumab/uso terapéutico , Fototerapia/métodos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Línea Celular Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA