Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Photodiagnosis Photodyn Ther ; 31: 101852, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32585403

RESUMEN

Photodynamic Therapy (PDT), is a treatment option for cancer.It involves the photochemical interaction of light, photosensitizer (PS) and molecular oxygen to produce radical species as well as singlet oxygen which induce cell death. Anthraquinones (AQs) have been extensively studied with respect to their UV/Vis absorption characteristics and their photosensitizing properties in photodynamic reactions. We study the photoactivity of different natural AQs (Parietin, Soranjidiol and Rubiadin) in treating monolayers and multicellular tumor spheroids (MCTSs). Rubiadin and soranjidiol were isolated and purified from the stem and leaves of Heterophyllae pustulata, and PTN was from the liquen Teloschistes flavicans by using repeated combination of several chromatographic techniques. Monolayer and spheroids of human colorectal adenocarcinoma SW480 cells were incubated with different concentrations of the AQs and then irradiated at room temperature. 24 h post-PDT cell viability, nuclear morphology and type of cell death were analyzed. We observed that Soranjidiol and Rubiadin showed no significant difference in the photosensitizing ability on monoculture of colon cancer cells (LD80 at 50 µM and 10 J/cm2, for both AQs). Nevertheless, for Parietin (PTN) LD80 was achieved at (20 µM using the same light dose (10 J/cm2). The death mechanism induced post-PDT was necrosis by use of Soranjidol and Rubiadin and apoptosis by use of PTN. Furthermore, in MCTSs of 300 and 900 µm, the treatment PTN- PDT produces the greatest cytotoxic effect. The three AQs analyzed could be promising chemotherapeutic candidates as anticancer PDT agents.


Asunto(s)
Adenocarcinoma , Neoplasias Colorrectales , Fotoquimioterapia , Adenocarcinoma/tratamiento farmacológico , Ascomicetos , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico
2.
Front Immunol ; 10: 2614, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31781113

RESUMEN

The immune response against cancer generated by type-I-interferons (IFN-1) has recently been described. Exogenous and endogenous IFN-α/ß have an important role in immune surveillance and control of tumor development. In addition, IFN-1s have recently emerged as novel DAMPs for the consecutive events connecting innate and adaptive immunity, and they also have been postulated as an essential requirement for induction of immunogenic cell death (ICD). In this context, photodynamic therapy (PDT) has been previously linked to the ICD. PDT consists in the administration of a photosensitizer (PS) and its activation by irradiation of the affected area with visible light producing excitation of the PS. This leads to the local generation of harmful reactive oxygen species (ROS) with limited or no systemic defects. In the current work, Me-ALA inducing PpIX (endogenous PS) was administrated to B16-OVA melanoma cells. PpIX preferentially localized in the endoplasmic reticulum (ER). Subsequent PpIX activation with visible light significantly induced oxidative ER-stress mediated-apoptotic cell death. Under these conditions, the present study was the first to report the in vitro upregulation of IFN-1 expression in response to photodynamic treatment in melanoma. This IFN-α/ß transcripts upregulation was concurrent with IRF-3 phosphorylation at levels that efficiently activated STAT1 and increased ligand receptor (cGAS) and ISG (CXCL10, MX1, ISG15) expression. The IFN-1 pathway has been identified as a critical molecular pathway for the antitumor host immune response, more specifically for the dendritic cells (DCs) functions. In this sense, PDT-treated melanoma cells induced IFN-1-dependent phenotypic maturation of monocyte-derived dendritic cells (DCs) by enhancing co-stimulatory signals (CD80, MHC-II) and tumor-directed chemotaxis. Collectively, our findings showed a new effect of PDT-treated cancer cells by modulating the IFN-1 pathway and its impact on the activation of DCs, emphasizing the potential relevance of PDT in adoptive immunotherapy protocols.


Asunto(s)
Células Dendríticas/inmunología , Interferón Tipo I/inmunología , Melanoma Experimental/tratamiento farmacológico , Fotoquimioterapia , Animales , Apoptosis , Línea Celular Tumoral , Luz , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Fotosensibilizantes/uso terapéutico , Protoporfirinas/uso terapéutico
3.
Cell Oncol (Dordr) ; 42(2): 173-196, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30756254

RESUMEN

PURPOSE: Previous analyses of the tumor microenvironment (TME) have resulted in a concept that tumor progression may depend on interactions between cancer cells and its surrounding stroma. An important aspect of these interactions is the ability of cancer cells to modulate stroma behavior, and vice versa, through the action of a variety of soluble mediators. Here, we aimed to identify soluble factors present in the TME of colorectal cancer cells that may affect relevant pathways through secretome profiling. METHODS: To partially recapitulate the TME and its architecture, we co-cultured colorectal cancer cells (SW480, TC) with stromal fibroblasts (MRC-5, F) as 3D-spheroids. Subsequent characterization of both homotypic (TC) and heterotypic (TC + F) spheroid secretomes was performed using label-free liquid chromatography-mass spectrometry (LC-MS). RESULTS: Through bioinformatic analysis using the NCI-Pathway Interaction Database (NCI-PID) we found that the HIF-1 signaling pathway was most highly enriched among the proteins whose secretion was enhanced in the heterotypic spheroids. Previously, we found that HIF-1 may be associated with resistance of colorectal cancer cells to photodynamic therapy (PDT), an antitumor therapy that combines photosensitizing agents, O2 and light to create a harmful photochemical reaction. Here, we found that the presence of fibroblasts considerably diminished the sensitivity of colorectal cancer cells to photodynamic activity. Although the biological significance of the HIF-1 pathway of secretomes was decreased after photosensitization, this decrease was partially reversed in heterotypic 3D-spheroids. HIF-1 pathway modulation by both PDT and stromal fibroblasts was confirmed through expression assessment of the HIF-target VEGF, as well as through HIF transcriptional activity assessment. CONCLUSION: Collectively, our results delineate a potential mechanism by which stromal fibroblasts may enhance colorectal cancer cell survival and photodynamic treatment resistance via HIF-1 pathway modulation.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos , Fibroblastos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fotoquimioterapia , Proteoma/metabolismo , Proteómica/métodos , Esferoides Celulares/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Transducción de Señal , Microambiente Tumoral
4.
Photochem Photobiol Sci ; 18(2): 546-554, 2019 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-30681107

RESUMEN

Photodynamic therapy (PDT) is a clinically approved treatment that exerts a selective cytotoxic activity toward cancer cells. The procedure involves the administration of a photosensitizer drug followed by its activation by visible light. In the presence of oxygen, a series of events lead to tumor cell death. PDT releases different cell signals, some of these lead to death while others can lead to survival. The surviving or resistant cells contribute to the recurrence of tumors after treatment, from which the necessity to understand this molecular response induced by PDT arises. It has been shown that both Heat Shock Proteins (HSPs) and autophagy promote PDT resistance. Moreover, both of them can be stimulated by PDT treatment. However, the molecular interplay between HSPs and autophagy in the photodynamic therapy context is poorly understood. We studied whether PDT induces autophagic activity through HSPs. We demonstrated that PDT promoted HSP27 expression, which in turn triggered autophagic cell survival as well as inhibited apoptosis in colon cancer cells. In addition, an overexpression of the HSP27/autophagy axis was observed in skin carcinoma cells resistant to PDT.


Asunto(s)
Autofagia/efectos de los fármacos , Autofagia/efectos de la radiación , Proteínas de Choque Térmico HSP27/metabolismo , Fotoquimioterapia , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Técnicas de Silenciamiento del Gen , Proteínas de Choque Térmico HSP27/deficiencia , Proteínas de Choque Térmico HSP27/genética , Humanos , Fármacos Fotosensibilizantes/farmacología , Protoporfirinas/farmacología
5.
Biomed Pharmacother ; 108: 1553-1564, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30372857

RESUMEN

The elevated expression of NQO1 in many human solid tumors along with its ability to activate quinone-based anticancer agents makes it an excellent target for enzyme-directed drug development. NQO1 plays an important role in melanogenesis and given its correlation with a poor patient outcome we propose this enzyme as an intriguing target for molecular-based therapeutic regimen against melanoma. Unfortunately, the natural product ß-Lapachone (ß-Lap), whose antitumor activity is based on NQO1, reported dose-limiting toxicity which hampered its pre-clinical and clinical use. Therefore, new effective and safe therapeutic NQO1-bioactivatable agents for melanoma treatment are desirable. Regarding NQO1, we demonstrated that halogenated ß-Lap derivative named PFB is an excellent substrate and effective tumor-selective anticancer compound. In addition, PFB resulted more attractive than the parent ß-Lap for treating metastatic-derived melanoma cells. In this context, it would be interesting to design strategies to induce NQO1 activity in cancer cells as a promising combinatorial approach with bioreductive drugs. In this sense, we had reported that photodynamic therapy (PDT) significantly upregulated NQO1 expression. Based on this event, here we demonstrated that the cytotoxic regimen consisting of PFB plus PDT improved synergistic therapeutic combination on melanoma cells. In conclusion, our contribution provides a strong rationale for using therapies that associate photo- and chemotherapy to effectively treat melanoma with modular NQO1 status.


Asunto(s)
Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/radioterapia , NAD(P)H Deshidrogenasa (Quinona)/biosíntesis , Naftoquinonas/uso terapéutico , Fotoquimioterapia/métodos , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Naftoquinonas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología
6.
Nanomedicine (Lond) ; 13(6): 605-624, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29376764

RESUMEN

AIM: Assess biocompatibility, uptake and photodynamic therapy (PDT) mechanism of metallated porphyrin doped conjugated polymer nanoparticles (CPNs) in human brain and colorectal tumor cells and macrophages. MATERIALS & METHODS: CPNs were developed employing 9,9-dioctylfluorene-alt-benzothiadiazole, an amphiphilic polymer (PS-PEG-COOH),  and platinum octaethylporphyrin. T98G, SW480 and RAW 264.7 cell lines were exposed to CPNs to assess uptake and intracellular localization. Additionally, a PDT protocol using CPNs was employed for the in vitro killing of cancer and macrophage cell lines. RESULTS & CONCLUSION: CPNs were well incorporated into glioblastoma and macrophage cells with localization in lysosomes. SW480 cells were less efficient incorporating CPNs with localization in the plasma membrane. In all cell lines PDT treatment was efficient inducing oxidative stress that triggered apoptosis.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Porfirinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Humanos , Macrófagos/efectos de los fármacos , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/química , Fotoquimioterapia , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacología , Polietilenglicoles/química , Polietilenglicoles/farmacología , Polímeros/química , Poliestirenos/química , Poliestirenos/farmacología , Porfirinas/química , Células RAW 264.7
7.
PLoS One ; 12(5): e0177801, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28545088

RESUMEN

Photodynamic therapy (PDT), a promising treatment option for cancer, involves the activation of a photosensitizer (PS) by local irradiation with visible light. Excitation of the PS leads to a series of photochemical reactions and consequently the local generation of harmful reactive oxygen species (ROS) causing limited or none systemic defects. However, the development of resistance to this promising therapy has slowed down its translation into the clinical practice. Thus, there is an increase need in understanding of the molecular mechanism underlying resistance to PDT. Here, we aimed to examine whether a relationship exists between PDT outcome and ROS-involvement in the resistance mechanism in photosensitized cancer cells. In order to recapitulate tumor architecture of the respective original tumor, we developed a multicellular three-dimensional spheroid system comprising a normoxic periphery, surrounding a hypoxic core. Using Me-ALA, a prodrug of the PS PpIX, in human colorectal spheroids we demonstrate that HIF-1 transcriptional activity was strongly up-regulated and mediates PDT resistant phenotype. RNAi knockdown of HIF-1 impairs resistance to PDT. Oxidative stress-mediated activation of ERK1/2 followed PDT was involved on positive modulation of HIF-1 transcriptional activity after photodynamic treatment. ROS scavenging and MEK/ERK pathway inhibition abrogated the PDT-mediated HIF-1 upregulation. Together our data demonstrate that resistance to PDT is in part mediated by the activation of a ROS-ERK1/2-HIF-1 axis, thus, identifying novel therapeutic targets that could be used in combination with PDT.


Asunto(s)
Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos , Factor 1 Inducible por Hipoxia/genética , Fotoquimioterapia/métodos , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas/citología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Biológicos , Esferoides Celulares , Células Tumorales Cultivadas/efectos de los fármacos , Regulación hacia Arriba
8.
Tumour Biol ; 37(1): 541-52, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26232323

RESUMEN

The study of cellular interactions in the tumor microenvironment has become one of the main areas of research in the fight against cancer. Tumor-associated macrophages (TAMs) influence tumor progression and therapy response due to its functional plasticity. Regarding cancer treatment, photodynamic therapy (PDT) is a minimally invasive and clinically approved procedure that involves the administration of a photosensitizer (PS), a nontoxic photosensitizing drug which is selectively retained in neoplastic tissue. Here, we investigated the role of resident and nonresident macrophages in the context of a PDT-treated colorectal tumor by developing a combination of 2-D and three-dimensional (3-D) experimental platform, recreating tumor-stroma interactions in vitro. Enhancement of cytotoxicity of PDT was achieved in the presence of nonresident macrophages which had a strong anti-tumor phenotype mediated by the production of nitric oxide, IL-6, and tumor necrosis factor alpha (TNF-α). On the contrary, tumor resident macrophages induced a pro-tumor phenotype promoting tumor cell migration and endothelial stimulation. Due to their plasticity, tumor-resident or tumor-recruited macrophages can differentially influence the response of tumors to PDT, so their multifactorial roles should be considered in the overall design of anti-tumor therapeutic.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Macrófagos/citología , Fotoquimioterapia/métodos , Microambiente Tumoral/efectos de los fármacos , Animales , Anexina A5/química , Antineoplásicos/química , Apoptosis , Arginasa/química , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Medios de Cultivo Condicionados/química , Células Endoteliales/citología , Ensayo de Inmunoadsorción Enzimática , Humanos , Imagenología Tridimensional , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Óxido Nítrico/química , Fármacos Fotosensibilizantes/química , Esferoides Celulares/química , Factor de Necrosis Tumoral alfa/metabolismo
9.
Tumour Biol ; 36(12): 9127-36, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26419592

RESUMEN

Melanoma is among the most aggressive and treatment-resistant human skin cancer. Photodynamic therapy (PDT), a minimally invasive therapeutic modality, is a promising approach to treating melanoma. It combines a non-toxic photoactivatable drug called photosensitizer with harmless visible light to generate reactive oxygen species which mediate the antitumor effects. The aim of this review was to compile the available data about PDT on melanoma. Our comparative analysis revealed a disconnection between several hypotheses generated by in vitro therapeutic studies and in vivo and clinical assays. This fact led us to highlight new preclinical experimental platforms that mimic the complexity of tumor biology. The tumor and its stromal microenvironment have a dynamic and reciprocal interaction that plays a critical role in tumor resistance, and these interactions can be exploited for novel therapeutic targets. In this sense, we review two strategies used by photodynamic researchers: (a) developing 3D culture systems which mimic tumor architecture and (b) heterotypic cultures that resemble tumor microenvironment to favor therapeutic regimen design. After this comprehensive review of the literature, we suggest that new complementary preclinical models are required to better optimize the clinical outcome of PDT on skin melanoma.


Asunto(s)
Melanoma/terapia , Fotoquimioterapia , Microambiente Tumoral/genética , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Humanos , Melanoma/patología , Fármacos Fotosensibilizantes/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Esferoides Celulares , Resultado del Tratamiento
10.
World J Clin Oncol ; 5(5): 901-7, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25493228

RESUMEN

Photodynamic therapy is a minimally invasive and clinically approved procedure for eliminating selected malignant cells with specific light activation of a photosensitizer agent. Whereas interstitial and intra-operative approaches have been investigated for the ablation of a broad range of superficial or bulky solid tumors such as breast cancer, the majority of approved photodynamic therapy protocols are for the treatment of superficial lesions of skin and luminal organs. This review article will discuss recent progress in research focused mainly on assessing the efficacies of various photosensitizers used in photodynamic therapy, as well as the combinatory strategies of various therapeutic modalities for improving treatments of parenchymal and/or stromal tissues of breast cancer solid tumors. Cytotoxic agents are used in cancer treatments for their effect on rapidly proliferating cancer cells. However, such therapeutics often lack specificity, which can lead to toxicity and undesirable side effects. Many approaches are designed to target tumors. Selective therapies can be established by focusing on distinctive intracellular (receptors, apoptotic pathways, multidrug resistance system, nitric oxide-mediated stress) and environmental (glucose, pH) differences between tumor and healthy tissue. A rational design of effective combination regimens for breast cancer treatment involves a better understanding of the mechanisms and molecular interactions of cytotoxic agents that underlie drug resistance and sensitivity.

11.
Photodiagnosis Photodyn Ther ; 11(2): 182-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24561303

RESUMEN

Over the past decade the science has studied synthetic photosensitizers used in photodynamic therapy (PDT) or photochemotherapy as anticancer candidates. In this context, compounds extracted from vegetable species present interesting potential in the cancer field. In our laboratory, we studied Heterophyllaea pustulata a phototoxic shrub that habit the northwest of Argentina. From this vegetal, by in vitro germination, we obtained Rubiadin and Soranjidiol, two anthraquinones that exhibited significant photocytotoxicity on human cancer cells. In addition, the fraction obtained from callus cultures allowed us to get a satisfactory content of these compounds compared to those found from the original plant. Under PDT regimen, we found that cell destruction resulted in a dose-dependent manner and occasioned apoptosis on photosensitized cells. Biochemical analysis revealed the involvement of caspase-3, PARP cleavage and DNA fragmentation in Rubiadin induced apoptosis. Moreover, Soranjidiol-PDT led to µ-calpain-induced apoptosis involving caspases-3-independent DNA fragmentation. We also showed that both anthraquinones are cytoplasmatically distributed and out of nucleus. In addition, we demonstrated a synergic cytotoxic effect when we combined them. Our data demonstrated that Rubiadin and Soranjidiol could be further considered as natural photocytotoxic compounds against cancer cells and callus cultures are a plausible source of these anthraquinonic compounds.


Asunto(s)
Antraquinonas/administración & dosificación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Fotoquimioterapia/métodos , Extractos Vegetales/administración & dosificación , Apoptosis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Células MCF-7 , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Resultado del Tratamiento
12.
Phytomedicine ; 20(11): 1007-12, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23746950

RESUMEN

ß-Lapachone is a phytochemotherapeutic originally isolated from Lapacho tree whose extract has been used medicinally for centuries. It is well known that NAD(P)H:quinone oxidoreductase (NQO1) activity is the principal determinant of ß-Lapachone cytotoxicity. As NQO1 is overexpressed in most common carcinomas, recent investigations suggest its potential application against cancer. Photodynamic therapy (PDT) is a clinically approved and rapidly developing cancer treatment. PDT involves the administration of photosensitizer (PS) followed by local illumination with visible light of specific wavelength. In the presence of oxygen molecules, the light illumination of PS can lead to a series of photochemical reactions and consequently the generation of cytotoxic reactive oxygen species (ROS). It has been reported that ß-Lapachone synergistically interacts with ionizing radiation, hyperthermia and cisplatin and that the sensitivity of cells to ß-Lapachone is closely related to the activity of NQO1. So, the present study aimed to investigate the feasibility of PDT to increase the anticancer effect of ß-Lapachone by up-regulating NQO1 expression on breast cancer MCF-7c3 cells. NQO1 expression was evaluated by Western blot analysis at different times after PDT using ME-ALA as PS. The cytotoxicity of the photodynamic treatment and ß-Lapachone alone or in combination was determined by MTT assay and the combination index (CI)-isobologram method and the dose reduction index (DRI) analysis were used to assess the effect of drug combinations. Our studies for the first time demonstrated that the expression of NQO1 is induced 24h after photodynamic treatment. The sensitivity of cancer cells to ß-Lapachone treatment increased 24h after PDT and a synergistic inhibitory effect on MCF-7c3 cells was showed. Taken together, these results lead us to conclude that the synergistic interaction between ß-Lapachone and PDT in killing cells was consistent with the up-regulation of NQO1. The combination of ß-Lapachone and PDT is a potentially promising modality for the treatment of cancer.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/uso terapéutico , Fotoquimioterapia , Fitoterapia , Tabebuia/química , Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Luz , Células MCF-7 , Naftoquinonas/farmacología , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
13.
Biochim Biophys Acta ; 1835(1): 36-45, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23046998

RESUMEN

Photodynamic therapy (PDT) is a novel cancer treatment. It involves the activation of a photosensitizer (PS) with light of specific wavelength, which interacts with molecular oxygen to generate singlet oxygen and other reactive oxygen species (ROS) that lead to tumor cell death. When a tumor is treated with PDT, in addition to affect cancer cells, the extracellular matrix and the other cellular components of the microenvironment are altered and finally this had effects on the tumor cells survival. Furthermore, the heterogeneity in the availability of nutrients and oxygen in the different regions of a tridimensional tumor has a strong impact on the sensitivity of cells to PDT. In this review, we summarize how PDT affects indirectly to the tumor cells, by the alterations on the extracellular matrix, the cell adhesion and the effects over the immune response. Also, we describe direct PDT effects on cancer cells, considering the intratumoral role that autophagy mediated by hypoxia-inducible factor 1 (HIF-1) has on the efficiency of the treatment.


Asunto(s)
Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Microambiente Tumoral/efectos de los fármacos , Animales , Autofagia/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología
14.
Exp Ther Med ; 1(4): 713-718, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22993594

RESUMEN

In the present study, a comparative photodynamic therapy (PDT) study was performed using the phthalocyanine derivatives, ZnPc(OCH(3))(4) and ZnPc(CF(3))(4), in a mouse tumor model, under identical experimental procedures. We studied the ablation of tumors induced by PDT. The end-point was to compare the photodynamic efficacy of ZnPc(OCH(3))(4) and ZnPc(CF(3))(4). ZnPc(OCH(3))(4) and ZnPc(CF(3))(4) were administered intraperitoneally at a dose of 0.2 mg/kg body weight. The injections of drugs were carried out in Balb/c mice bearing subcutaneously inoculated LM2 mouse mammary adenocarcinoma. Histological examination and serum biochemical parameters were used to evaluate hepatic and renal toxicity and function. Phototherapeutic studies were achieved employing a light intensity of 210 J/cm(2). After PDT, tumoral regression analyses were carried out, and the degree of tumor cell death was measured utilizing the vital stain Evan's blue. In this pilot study, we revealed that the cytotoxic effect of ZnPc(OCH(3))(4) after PDT led to a higher success rate compared to ZnPc(CF(3))(4)-PDT when both were intraperitoneally injectioned. Both phthalocynanine derivatives were able to induce ablation in the tumors. In summary, these results demonstrate the feasibility of ZnPc(OCH(3))(4)- or ZnPc(CF(3))(4)-PDT and its potential as a treatment for small tumors.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA