Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Photochem Photobiol B ; 178: 521-529, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29245122

RESUMO

The inhibitor of DNA binding and cell differentiation (Id) proteins are dominant negative regulators of the helix-loop-helix transcription factor family and play a key role during development as well as in vascular disorders and cancer. In fact, impairing the Id-protein activity in cancer cells reduces cell growth and even chemoresistance. Recently, we have shown that a synthetic Id-protein ligand (1Y) consisting of a cyclic nonapeptide can reduce the viability of the two breast cancer cell lines MCF-7 and T47D and of the bladder cancer cells T24 to about 50% at concentrations ≥100µM. Moreover, the cyclopeptide displays both proapoptotic and antiproliferative effects on MCF-7 cells. Herein, we show that the cyclopeptide does not induce cell death at the dose of 5µΜ, but it still inhibits MCF-7 and T24 cell proliferation, which correlates with an increased protein level of the cell-cycle regulator p27Kip1. Furthermore, 1Y-pretreated MCF-7, T47D, and T24 cells are more susceptible than untreated cells to the phototoxic effects of the three photosensitizers meta-tetra(hydroxyphenyl)chlorin, porfimer sodium, and hypericin, which are applied in photodynamic therapy (PDT). The combination of the Id-protein ligand with each of the light-activated photosensitizers shows synergistic effects on the reduction of cell viability. In conclusion, an Id-protein ligand with moderate cancer cell killing activity at concentrations ≥100µM can be applied at a 20-fold lower and barely toxic dose to raise the sensitivity of cancer cells towards phototoxicity associated with photodynamic treatment. This suggests the potential benefit of targeting the Id proteins in combined drug approaches for cancer therapy.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteína 1 Inibidora de Diferenciação/antagonistas & inibidores , Peptídeos Cíclicos/toxicidade , Fármacos Fotossensibilizantes/toxicidade , Antracenos , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Éter de Diematoporfirina/toxicidade , Sinergismo Farmacológico , Humanos , Proteína 1 Inibidora de Diferenciação/metabolismo , Luz , Células MCF-7 , Nanoestruturas/química , Nanoestruturas/toxicidade , Peptídeos Cíclicos/química , Perileno/análogos & derivados , Perileno/toxicidade , Fármacos Fotossensibilizantes/química , Porfirinas/toxicidade
2.
Cell Physiol Biochem ; 35(4): 1527-36, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25791936

RESUMO

BACKGROUND/AIMS: Photodynamic therapy (PDT) is a promising noninvasive technique, which has been successfully applied to the treatment of human cancers. Studies have shown that the Bcl-2 family proteins play important roles in PDT-induced apoptosis. However, whether Bcl-2-interacting mediator of cell death (Bim) is involved in photodynamic treatment remains unknown. In this study, we attempt to determine the effect of Bim on Photofrin photodynamic treatment (PPT)-induced apoptosis in human lung adenocarcinoma ASTC-a-1 cells. METHODS: The translocation of Bim/Bax of the cells were monitored by laser confocal scanning microscope. The levels of Bim protein and activated caspase-3 in cells were detected by western blot assay. Caspase-3 activities were measured by Caspase-3 Fluorogenic Substrate (Ac-DEVD-AFC) analysis. The induction of apoptosis was detected by Hoechst 33258 and PI staining as well as flow cytometry analysis. The effect of Bim on PPT-induced apoptosis was determined by RNAi. RESULTS: BimL translocated to mitochondria in response to PPT, similar to the downstream pro-apoptotic protein Bax activation. PPT increased the level of Bim and activated caspase-3 in cells and that knockdown of Bim by RNAi significantly protected against caspase-3 activity. PPT-induced apoptosis were suppressed in cells transfected with shRNA-Bim. CONCLUSION: We demonstrated the involvement of Bim in PPT-induced apoptosis in human ASTC-a-1 lung adenocarcinoma cells and suggested that enhancing Bim activity might be a potential strategy for treating human cancers.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/efeitos dos fármacos , Éter de Diematoporfirina/toxicidade , Proteínas de Membrana/metabolismo , Fármacos Fotossensibilizantes/toxicidade , Proteínas Proto-Oncogênicas/metabolismo , Apoptose/efeitos da radiação , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Western Blotting , Caspase 3/metabolismo , Linhagem Celular Tumoral , Humanos , Lasers , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Microscopia Eletrônica de Varredura , Mitocôndrias/metabolismo , Fotoquimioterapia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína X Associada a bcl-2/metabolismo
3.
Int J Oncol ; 39(5): 1133-41, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21850369

RESUMO

Clinical studies have indicated that photodynamic therapy (PDT) significantly prolonged the median survival of patients with gliomas. Experimental studies demonstrate that increasing optical energy and photosensitizer dose leads to increased volume of tumor necrosis. However, increasing the light dose delivered to the tumor may increase the risks of inducing permanent neurological deficits. In the current study, we sought to test the behavioral deficits induced in normal rats by brain PDT and the neurorestorative effects of atorvastatin on PDT-induced behavioral deficits. Considering its potential as a combination treatment of brain tumors, we investigated both in vitro and in vivo whether atorvastatin treatment promotes brain tumor growth. Non-tumored Fischer rats received PDT (n=18). Nine of the PDT-treated animals were treated with atorvastatin. Control animals underwent the same surgical procedure, but did not receive Photofrin and laser light. PDT-treated animals had significant behavioral deficits on days 2, 5, 7, 9 and 14 after PDT, compared with surgery controls. PDT-treated animals receiving atorvastatin displayed significantly ameliorated behavioral deficits on days 7, 9 and 14 after PDT, compared to PDT-treated rats. In vitro tumor cell viability and growth were evaluated. Atorvastatin did not affect the growth of glioma cells. Fischer rats with intracranial 7-day-old 9L glioma tumor cell implantation were randomly subjected to no treatment, PDT alone, atorvastatin alone, or combined treatment with atorvastatin and PDT (6 rats/group). Our data indicate that atorvastatin did not promote tumor growth in either PDT treated and non-treated rats. However, atorvastatin significantly reduced the cell damage caused by PDT. To further test the mechanisms underlying the atorvastatin-mediated reduction of functional deficits, we investigated the effects of atorvastatin on angiogenesis and synaptogenesis. Our data demonstrate that atorvastatin significantly induced angiogenesis and synaptogenesis in the PDT-damaged brain tissue. Our data indicate that PDT induces functional deficits. Atorvastatin treatment promotes functional restoration after PDT, but does not promote glioma growth in vitro and in vivo. Atorvastatin reduces astrocyte and endothelial cell damage caused by PDT and induces angiogenesis and synaptogenesis after PDT. Thus consideration and further testing of the combination of atorvastatin and PDT for the treatment of glioma is warranted.


Assuntos
Ácidos Heptanoicos/uso terapêutico , Transtornos Mentais/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Fotoquimioterapia/efeitos adversos , Pirróis/uso terapêutico , Animais , Astrócitos/efeitos dos fármacos , Atorvastatina , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Proliferação de Células , Éter de Diematoporfirina/toxicidade , Células Endoteliais/efeitos dos fármacos , Glioma/patologia , Glioma/terapia , Ácidos Heptanoicos/farmacologia , Humanos , Transtornos Mentais/induzido quimicamente , Transtornos Mentais/prevenção & controle , Neovascularização Fisiológica/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Pirróis/farmacologia , Ratos , Ratos Endogâmicos F344 , Sinaptofisina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator de von Willebrand/metabolismo
4.
Photochem Photobiol Sci ; 10(6): 1056-65, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21409208

RESUMO

Biological oxygen measurements by phosphorescence quenching make use of exogenous phosphorescent probes, which are introduced directly into the medium of interest (e.g. blood or interstitial fluid) where they serve as molecular sensors for oxygen. The byproduct of the quenching reaction is singlet oxygen, a highly reactive species capable of damaging biological tissue. Consequently, potential probe phototoxicity is a concern for biological applications. Herein, we compared the ability of polyethyleneglycol (PEG)-coated Pd tetrabenzoporphyrin (PdTBP)-based dendritic nanoprobes of three successive generations to sensitize singlet oxygen. It was found that the size of the dendrimer has practically no effect on the singlet oxygen sensitization efficiency in spite of the strong attenuation of the triplet quenching rate with an increase in the dendrimer generation. This unexpected result is due to the fact that the lifetime of the PdTBP triplet state in the absence of oxygen increases with dendritic generation, thus compensating for the concomitant decrease in the rate of quenching. Nevertheless, in spite of their ability to sensitize singlet oxygen, the phosphorescent probes were found to be non-phototoxic when compared with the commonly used photodynamic drug Photofrin in a standard cell-survival assay. The lack of phototoxicity is presumably due to the inability of PEGylated probes to associate with cell surfaces and/or penetrate cellular membranes. In contrast, conventional photosensitizers bind to cell components and act by generating singlet oxygen inside or in the immediate vicinity of cellular organelles. Therefore, PEGylated dendritic probes are safe to use for tissue oxygen measurements as long as the light doses are less than or equal to those commonly employed in photodynamic therapy.


Assuntos
Dendrímeros/química , Substâncias Luminescentes/química , Oxigênio/química , Porfirinas/química , Animais , Linhagem Celular Tumoral , Éter de Diematoporfirina/toxicidade , Luz , Substâncias Luminescentes/toxicidade , Camundongos , Paládio/química , Polietilenoglicóis/química , Porfirinas/toxicidade , Oxigênio Singlete/química , Oxigênio Singlete/metabolismo , Espectrometria de Fluorescência
5.
J Photochem Photobiol B ; 100(3): 173-80, 2010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20599390

RESUMO

At present a wide range of photosensitizers are employed in photodynamic therapy (PDT) that have very different characteristics. Although, countless in vitro studies on the attributes of photosensitizers do exist, a direct comparison of these substances on one cell line are rare and may contribute to the choice of the optimal photoactive substance for a specific application. We therefore evaluated the properties of six widespread photosensitizers, namely Foscan, Fospeg, hypericin, aluminum (III) phthalocyanine tetrasulfonate chloride (AlPcS(4)), 5-aminolevulinic acid (ALA), and Photofrin in terms of: (i) cytotoxicity without illumination, (ii) phototoxicity, (iii) cellular uptake and release, and (iv) apoptosis induction in A431 human epidermoid carcinoma cells using comparable illumination regimens. We clearly show that meso-tetrahydroxyphenylchlorin (mTHPC, Foscan) is a very effective photosensitizer inducing high phototoxicity at very low concentrations. Similar in vitro characteristics and phototoxicity were observed for Fospeg, the water-soluble formulation of mTHPC. Hypericin, a photosensitizer extracted from plants of the Hypericum genus, is very effective in inducing apoptosis over a wide range of light fluences. AlPcS(4) absorbs light of 674 nm wavelength providing a higher penetration depth in tissue. Its hydrophilic character allows for application as aqueous solution. ALA can be administered at very high concentrations without producing cytotoxic effects in the dark. The intracellular concentration of protoporphyrin IX rapidly decreases after withdrawal of ALA, thus minimizing the period of light sensitivity post PDT. Among all photosensitizers Photofrin has most clinical approvals and serves as standard.


Assuntos
Fármacos Fotossensibilizantes/toxicidade , Ácido Aminolevulínico/toxicidade , Antracenos , Antineoplásicos/toxicidade , Apoptose , Linhagem Celular Tumoral , Éter de Diematoporfirina/toxicidade , Humanos , Indóis/toxicidade , Mesoporfirinas/toxicidade , Compostos Organometálicos/toxicidade , Perileno/análogos & derivados , Perileno/toxicidade , Fotoquimioterapia
6.
Photochem Photobiol Sci ; 8(12): 1683-93, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20024165

RESUMO

Photodynamic therapy (PDT) can lead to the creation of heterogeneous, response-limiting hypoxia during illumination, which may be controlled in part through illumination fluence rate. In the present report we consider (1) regional differences in hypoxia, vascular response, and cell kill as a function of tumor depth and (2) the role of fluence rate as a mediator of depth-dependent regional intratumor heterogeneity. Intradermal RIF murine tumors were treated with Photofrin PDT using surface illumination at an irradiance of 75 or 38 mW cm(-2). Regional heterogeneity in tumor response was examined through comparison of effects in the surface vs. base of tumors, i.e. along a plane parallel to the skin surface and perpendicular to the incident illumination. 75 mW cm(-2) PDT created significantly greater hypoxia in tumor bases relative to their surfaces. Increased hypoxia in the tumor base could not be attributed to regional differences in Photofrin concentration nor effects of fluence rate distribution on photochemical oxygen consumption, but significant depth-dependent heterogeneity in vascular responses and cytotoxic response were detected. At a lower fluence rate of 38 mW cm(-2), no detectable regional differences in hypoxia or cytotoxic responses were apparent, and heterogeneity in vascular response was significantly less than that during 75 mW cm(-2) PDT. This research suggests that the benefits of low-fluence-rate PDT are mediated in part by a reduction in intratumor heterogeneity in hypoxic, vascular and cytotoxic responses.


Assuntos
Éter de Diematoporfirina/uso terapêutico , Fibrossarcoma/tratamento farmacológico , Neoplasias Induzidas por Radiação/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Animais , Hipóxia Celular , Éter de Diematoporfirina/toxicidade , Fibrossarcoma/metabolismo , Luz , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Induzidas por Radiação/metabolismo , Nitroimidazóis/química , Nitroimidazóis/metabolismo , Fotoquimioterapia , Fármacos Fotossensibilizantes/toxicidade
7.
J Photochem Photobiol B ; 85(2): 92-101, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16829117

RESUMO

The invasive nature of malignant gliomas makes treatment by surgery alone extremely difficult. However, the preferential accumulation of photosensitisers in neoplastic tissues suggests photodynamic therapy (PDT) may be useful as an adjuvant therapy following tumour resection. In this study, the potential use of three different photosensitisers, namely Photofrin, 5-aminolevulinic acid (5-ALA) and calphostin C in the treatment of glioma was investigated. The uptake, cytotoxicity on U87 and GBM6840 glioma cell lines were determined by flow cytometry and MTT assay respectively. Their effect on glioma cell invasiveness was evaluated by (1) measuring the levels of matrix degradation enzymes matrix metalloproteinase (MMP)-2 and -9 using gelatin zymography, and (2) Matrigel invasion assay. The results showed that uptake of calphostin C reached saturation within 2 h, while Photofrin and 5-ALA induced protoporphyrin IX (PpIX) levels elevated steadily up to 24 h. Photocytotoxic effect on the two glioma cell lines was similar with LD50 at optimal uptake: 1 microg/mL Photofrin at 1.5 J/cm(2); 1 mM 5-ALA at 2 J/cm(2) and 100 nM calphostin C at 2 J/cm(2). The inhibition in cell proliferation after Photofrin treatment was similar for both cell lines, which correlated to more cells being arrested in the G0/G1 phase of the cell cycle (P<0.01). By contrast, U87 was more sensitive to calphostin C whereas GBM6840 was more susceptible to 5-ALA treatment. The ability of both cell lines to migrate through the Matrigel artificial basement membrane was significantly reduced after PDT (P<0.001). This might be due to a decreased production in MMP-2 and MMP-9, together with the reduction of adhesion molecule expression. Photofrin was most superior in inhibiting cell invasion and calphostin C was least effective in reducing adhesion molecule expression. Taken together, PDT could be useful in the treatment of gliomas but the choice of photosensitisers must be taken into consideration.


Assuntos
Ácido Aminolevulínico/farmacologia , Éter de Diematoporfirina/farmacologia , Glioma/patologia , Naftalenos/farmacologia , Fármacos Fotossensibilizantes/farmacologia , Ácido Aminolevulínico/toxicidade , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Éter de Diematoporfirina/toxicidade , Glioma/metabolismo , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Naftalenos/toxicidade , Invasividade Neoplásica/patologia , Fármacos Fotossensibilizantes/toxicidade
8.
Cancer Lett ; 241(1): 42-8, 2006 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-16303246

RESUMO

In recent years, photodynamic therapy (PDT) with a photosensitizer and laser has been given attention, especially for the treatment of superficial cancers, such as lung, gastric, bladder and cervical cancer. In this study, in order to enhance the efficacy of PDT, photofrin liposome (PF-Lip) was prepared with dimyristoylphosphatidylcholine, dimyristoylphosphatidylglycerol and cholesterol. Polyethyleneglycol modified photofrin liposome (PF-PEG-Lip) was prepared by modification of PF-Lip with monomethoxypolyethyleneglycol-2.3-dimyristoylglycerol. PF-Lip and PF-PEG-Lip entrapped with photofrin with 81.0+/-5.9 and 81.2+/-9.2%, respectively. The particle size of each liposome was 114.3+/-5.7nm (PF-Lip) and 118+/-3.5nm (PF-PEG-Lip), respectively. It was suggested that PEGylated liposomes has no effect on the trapping ratio of PF and particle size. Phototoxicity was enhanced by liposomalization, especially PEG-modification. However, PF-PEG-Lip inhibited the uptake of photofrin into tumor cells. The amount of singlet oxygen from photofrin solution (PF-sol) and each liposome was PF-PEG-Lip=PF-Lip>PF-sol. The photofrin release revel of PF-PEG-Lip was lower than that of PF-Lip. In conclusion, the phototoxicity of PF-PEG-Lip was significantly higher than that of PF-sol or PF-Lip. It is expected that formation of a fixed aqueous layer on the liposome membrane by PEGylation physically changed it into the stable state of PF-PEG-Lip.


Assuntos
Éter de Diematoporfirina/toxicidade , Fármacos Fotossensibilizantes/química , Polietilenoglicóis/química , Linhagem Celular Tumoral , Éter de Diematoporfirina/química , Feminino , Humanos , Lipossomos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Tamanho da Partícula , Fotoquimioterapia , Sarcoma/metabolismo , Sarcoma/patologia , Oxigênio Singlete/metabolismo
9.
J Cell Physiol ; 194(3): 363-75, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12548556

RESUMO

Photodynamic therapy (PDT) is a kind of photochemo-therapeutic treatment that exerts its effect mainly through the induction of cell death. Distinct types of cell death may be elicited by different PDT regimes. In this study, the mechanisms involved in the death of human epidermoid carcinoma A431 cells triggered by PDT with Photofrin (a clinically approved photosensitizer) were characterized. Photofrin distributes dynamically in A431 cells; the plasma membranes and Golgi complex are the main target sites of Photofrin after a brief (3 h) and prolonged (24 h) incubation, respectively. Cells with differentially localized Photofrin displayed distinct death phenotypes in response to PDT. The effects of PDT on cells with plasma membrane-localized Photofrin were further studied in details. Cells stopped proliferating post PDT at Photofrin dose >7 micro g/ml, and at higher dose (28 micro g/ml) plasma membrane disruption and cell swelling were observed immediately after PDT. Dramatic alterations of several important signaling events were detected in A431 cells post Photofrin-PDT, including (i) immediate formation of reactive oxygen species (ROS), (ii) rapid activation of c-Jun N-terminal kinase, (iii) delayed activation of caspase-3 and cleavage of polyADP-ribose polymerase and p21-activated kinase 2, and (iv) loss of mitochondrial membrane potential. Intriguingly, the characteristics of typical apoptosis such as phosphatidylserine externalization and DNA fragmentation were not detected in the cell death process caused by this PDT regime. In conclusion, our results show that when plasma membranes are the main targets, Photofrin-PDT can lead to instant ROS formation and subsequent activation of downstream signaling events similar to those elicited by many apoptotic stimuli, but the damage of plasma membranes renders the death phenotype more necrosis like.


Assuntos
Antineoplásicos/farmacocinética , Carcinoma de Células Escamosas , Morte Celular/efeitos dos fármacos , Éter de Diematoporfirina/farmacocinética , Neoplasias Cutâneas , Antineoplásicos/toxicidade , Caspase 3 , Caspases/metabolismo , Membrana Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Éter de Diematoporfirina/toxicidade , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Lasers , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fenótipo , Fosfatidilserinas/metabolismo , Fotoquimioterapia , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
10.
Cancer Lett ; 184(2): 171-8, 2002 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-12127689

RESUMO

We investigated the cytotoxic and apoptotic effects of a combination of photodynamic therapy (PDT) and cisplatin (CDDP) on L5178Y (LY) cells. Treatment with PDT by photofrin((R)) (5 microg/ml) alone or with CDDP (20 microg/ml) alone killed 41.5+/-8.5% or 42.9+/-6.5% of LY cells, respectively, while a combination of these two treatments killed 99.7+/-0.6%. Apoptotic cell death after combination treatment was also revealed to be 49.6+/-7.8% compared to 12.4+/-3.4% after PDT alone, and 18.8+/-2.6% after CDDP. This study demonstrated that combined treatment of PDT and CDDP results in enhanced apoptotic cell death as well as a cytotoxic effect on LY cells.


Assuntos
Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/toxicidade , Éter de Diematoporfirina/toxicidade , Fotoquimioterapia , Humanos , Células Tumorais Cultivadas
11.
J Photochem Photobiol B ; 64(1): 55-61, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11705730

RESUMO

Photodynamic therapy (PDT) invokes a number of cellular responses. Other studies have shown that PDT induces transcription and translation of heat shock proteins (Hsps). The expression of mitochondrial heat shock protein, Hsp60, was measured following in vitro Photofrin-mediated PDT in the colon cancer cell line HT29 and its PDT-induced resistant variant HT29-P14 as well as the radiation-induced fibrosarcoma cells RIF-1 and its PDT-induced resistant variant, RIF-8A. Basal levels of Hsp60 were found to be similar in the two murine cell lines. In the human model, the resistant HT29-P14 cell line showed a small increase in basal levels relative to its parental population. Incubation with Photofrin (PII) alone or photosensitization caused a significant increase in Hsp60 levels in all cell lines as determined by flow cytometry. A dose-dependent and temporal relationship for PDT response was observed, maximum levels were detected 6-8 h post PDT, at which time, Hsp60 induction was found to be significantly greater in the two resistant variants. Induction in the RIF cells was also found to be greater after incubation with PII alone at the highest doses tested. These results indicate that the presence of PII and the subsequent oxidative stress of PDT can induce Hsp60 and implicated it as a common factor that may contribute to the resistance observed in the induced resistant cells.


Assuntos
Chaperonina 60/biossíntese , Éter de Diematoporfirina/toxicidade , Fotoquimioterapia , Fármacos Fotossensibilizantes/toxicidade , Animais , Carcinoma , Chaperonina 60/metabolismo , Neoplasias do Colo , Ensaio de Unidades Formadoras de Colônias , Resistencia a Medicamentos Antineoplásicos , Fibrossarcoma , Citometria de Fluxo , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias Induzidas por Radiação , Células Tumorais Cultivadas
12.
Photochem Photobiol ; 73(4): 378-87, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11332033

RESUMO

Confluent layers of MDCK II cells were treated with four different photosensitizers (a purified version of hematoporphyrin derivative [Photofrin], tetra(3-hydroxyphenyl)porphine [3-THPP], meso-tetra(4-sulphonatophenyl)porphine [TPPS4] and ALA-induced Protoporphyrin IX) and irradiated with blue light, with UVA without exogenous photosensitizers, or incubated with the metabolic inhibitors carbonyl cyanide m-chlorophenylhydrazone and 2-deoxy-D-glucose. Necrotic and apoptotic cells were detected about 4 h later by fluorescence microscopy. Dead cells appeared in distinct clusters in the confluent layers. The number of dead cells in these clusters was determined by manual counting and image analysis. Forty-one of the 43 experimental distributions of dead cells in clusters were found to be significantly different from a Monte Carlo simulation of the distribution of independently inactivated cells. However, a Monte Carlo simulation model, assuming that each dead cell increased the probability of inactivation of adjacent cells, fitted 34 of the 43 observed distributions of dead cells in clusters, indicating a significant bystander effect for all the investigated treatments. The bystander-effect model parameter, defined as a cell's increase in probability of dying when it has dead neighbors, was significantly lower for 3-THPP-PDT and TPPS4-PDT than for Photofrin-PDT, ALA-PDT and treatment with metabolic inhibitors.


Assuntos
Trifosfato de Adenosina/metabolismo , Morte Celular/fisiologia , Éter de Diematoporfirina/toxicidade , Fármacos Fotossensibilizantes/toxicidade , Raios Ultravioleta/efeitos adversos , Algoritmos , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/toxicidade , Comunicação Celular , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Células Cultivadas , Desoxiglucose/toxicidade , Modelos Animais de Doenças , Cães , Rim/anatomia & histologia , Rim/efeitos dos fármacos , Rim/efeitos da radiação , Fotoquímica
13.
Photochem Photobiol ; 73(5): 518-24, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11367574

RESUMO

Relatively little is known about the immunosuppression induced in mice which have received cutaneous photodynamic therapy (PDT). Consequently, experiments were undertaken using mice which received dorsal PDT using Photofrin as the photosensitizer in an attempt to characterize the overall nature of the immunosuppression. Photoirradiation of mice at various times after injection indicated there was no correlation between photosensitivity and immunosuppression. The suppression was found to be adoptively transferable and antigen specific suggesting the generation of suppressor cells. Selective cell depletions prior to adoptive transfer indicated a CD4+ T cell to be responsible for the immunosuppression. Interestingly, using allogeneic spleen cells, no effect on the delayed type hypersensitivity (DTH) response was found. The results indicate that the suppression induced by cutaneous PDT, with the exception of the lack of DTH suppression, is similar to that induced by UVB irradiation but unlike that reported using laser PDT of the peritoneal cavity. This suggests that not only the type of photoirradiation but also the site of photoirradiation might determine the character of the induced immunosuppression.


Assuntos
Epitopos/imunologia , Fotoquimioterapia/efeitos adversos , Pele/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Éter de Diematoporfirina/farmacologia , Éter de Diematoporfirina/toxicidade , Feminino , Hipersensibilidade Tardia/induzido quimicamente , Hipersensibilidade Tardia/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/toxicidade , Pele/efeitos dos fármacos , Pele/efeitos da radiação , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/efeitos da radiação , Linfócitos T/efeitos dos fármacos , Linfócitos T/efeitos da radiação
14.
Res Exp Med (Berl) ; 199(6): 341-57, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10945652

RESUMO

Photodynamic therapy (PDT) induces cell-membrane damage and alterations in cancer-cell adhesiveness, an important parameter in cancer metastasis. These alterations result from cell sensitivity to photosensitizers and the distribution of photosensitizers in cells. The efficacy of photosensitizers depends on their close proximity to targets and thus on their pharmacokinetics at the cellular level. We studied the cellular distribution of photosensitizers with a confocal microspectrofluorimeter by analysing the fluorescence emitted by benzoporphyrin derivative-monoacid ring A (BPD-MA) and Photofrin relative to their cell sensitivity. Two cancer cell lines of colonic origin, but with different metastatic properties, were used: PROb (progressive) and REGb (regressive). For BPD-MA (1.75 microg/ml), maximal fluorescence intensity (8,300 cts) was reached after 2 h for PROb and after 1 h (4,900 cts) for REGb. For Photofrin (10 microg/ml), maximal fluorescence intensity (467 cts) was reached after 5 h for PROb and after 3 h (404 cts) for REGb. Intracellular studies revealed stronger cytoplasmic than nuclear fluorescence for both BPD and Photofrin. Both of the sensitizers induced a dose-dependent phototoxicity; LD50 with BPD-MA was 93.3 ng/ml for PROb and 71.1 ng/ml for REGb, under an irradiation of 10 J/cm2. With Photofrin, LD50 was 1,270 ng/ml for PROb and 1,200 ng/ml for REGb under an irradiation of 25 J/cm2. The photosensitizer effect within PROb and REGb cancer cells was assessed by incorporation kinetics and toxicity-phototoxicity tests. The intracellular concentration of the photosensitive agent was one important factor in the effectiveness of PDT, but not the only one contributing to the photodynamic effect. In conclusion, this study showed that there was a clear difference between sensitizer uptake and phototoxicity, even in cancer cells of the same origin. This could induce cell-killing heterogeneity in clinics.


Assuntos
Adenocarcinoma , Antineoplásicos/toxicidade , Neoplasias do Colo , Éter de Diematoporfirina/toxicidade , Fármacos Fotossensibilizantes/toxicidade , Porfirinas/toxicidade , Animais , Antineoplásicos/farmacocinética , Núcleo Celular/metabolismo , Éter de Diematoporfirina/farmacocinética , Processamento de Imagem Assistida por Computador , Microscopia Confocal , Microscopia de Fluorescência , Fármacos Fotossensibilizantes/farmacocinética , Fototerapia/efeitos adversos , Porfirinas/farmacocinética , Ratos , Ratos Endogâmicos , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
15.
Photochem Photobiol ; 71(2): 201-10, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10687395

RESUMO

Although there is evidence that the p53 tumor suppressor plays a role in the response of some human cells to chemotherapy and radiation therapy, its role in the response of human cells to photodynamic therapy (PDT) is less clear. In order to examine the role of p53 in cellular sensitivity to PDT, we have examined the clonogenic survival of normal human fibroblasts that express wild-type p53 and immortalized Li-Fraumeni syndrome (LFS) cells that express only mutant p53, following Photofrin-mediated PDT. The LFS cells were found to be more resistant to PDT compared to normal human fibroblasts. The D37 (LFS cells)/D37 (normal human fibroblasts) was 2.8 +/- 0.3 for seven independent experiments. Although the uptake of Photofrin per cell was 1.6 +/- 0.1-fold greater in normal human fibroblast cells compared to that in LFS cells over the range of Photofrin concentrations employed, PDT treatment at equivalent cellular Photofrin levels also demonstrated an increased resistance for LFS cells compared to normal human fibroblasts. Furthermore, adenovirus-mediated transfer and expression of wild-type p53 in LFS cells resulted in an increased sensitivity to PDT but no change in the uptake of Photofrin per cell. These results suggest a role for p53 in the response of human cells to PDT. Although normal human fibroblasts displayed increased levels of p53 following PDT, we did not detect apoptosis or any marked alteration in the cell cycle of GM38 cells, despite a marked loss of cell viability. In contrast, LFS cells exhibited a prolonged accumulation of cells in G2 phase and underwent apoptosis following PDT at equivalent Photofrin levels. The number of apoptotic LFS cells increased with time after PDT and correlated with the loss of cell viability. A p53-independent induction of apoptosis appears to be an important mechanism contributing to loss of clonogenic survival after PDT in LFS cells, whereas the induction of apoptosis does not appear to be an important mechanism leading to loss of cell survival in the more sensitive normal human fibroblasts following PDT at equivalent cellular Photofrin levels.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Éter de Diematoporfirina/toxicidade , Genes p53 , Fotoquimioterapia , Linhagem Celular , Linhagem Celular Transformada , Sobrevivência Celular/efeitos da radiação , Fibroblastos , Humanos , Síndrome de Li-Fraumeni , Luz , Proteína Supressora de Tumor p53/metabolismo
16.
Br J Cancer ; 80(5-6): 744-55, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10360652

RESUMO

The damage to normal pig bronchial mucosa caused by photodynamic therapy (PDT) using mTHPC and Photofrin as photosensitizers was evaluated. An endobronchial applicator was used to deliver the light with a linear diffuser and to measure the light fluence in situ. The applied fluences were varied, based on existing clinical protocols. A fluence finding experiment with short-term (1-2 days) response as an end point showed considerable damage to the mucosa with the use of Photofrin (fluences 50-275 J cm(-2), drug dose 2 mg kg(-1)) with oedema and blood vessel damage as most important features. In the short-term mTHPC experiment the damage found was slight (fluences 12.5-50 J cm(-2), drug dose 0.15 mg kg(-1)). For both sensitizers, atrophy and acute inflammation of the epithelium and the submucosal glands was observed. The damage was confined to the mucosa and submucosa leaving the cartilage intact. A long-term response experiment showed that fluences of 50 J cm(-2) for mTHPC and 65 J cm(-2) for Photofrin-treated animals caused damage that recovered within 14 days, with sporadic slight fibrosis and occasional inflammation of the submucosal glands. Limited data on the pharmacokinetics of mTHPC show that drug levels in the trachea are similar at 6 and 20 days post injection, indicating a broad time window for treatment. The importance of in situ light dosimetry was stressed by the inter-animal variations in fluence rate for comparable illumination conditions.


Assuntos
Éter de Diematoporfirina/toxicidade , Mesoporfirinas/toxicidade , Fotoquimioterapia/efeitos adversos , Fármacos Fotossensibilizantes/toxicidade , Traqueia/efeitos dos fármacos , Animais , Antineoplásicos/toxicidade , Relação Dose-Resposta à Radiação , Esôfago/metabolismo , Feminino , Seguimentos , Luz , Mesoporfirinas/farmacocinética , Mucosa/efeitos dos fármacos , Mucosa/patologia , Mucosa/efeitos da radiação , Projetos Piloto , Pele/metabolismo , Suínos , Fatores de Tempo , Distribuição Tecidual , Traqueia/patologia , Traqueia/efeitos da radiação
17.
Lasers Surg Med ; 24(4): 276-84, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10327046

RESUMO

BACKGROUND AND OBJECTIVE: New photosensitizers proposed for photodynamic therapy (PDT) treatment of tumors need to be evaluated in animal models to determine the parameters needed for treatment. They also need to be compared with existing photosensitizers for efficacy. We examined the PDT response to lutetium-texaphyrin (PCI-0123) in a mouse mammary adenocarcinoma model and compared it with the PDT response seen when using Photofrin. STUDY DESIGN/MATERIALS AND METHODS: DBA/2 mice with SMT-F tumors were used to explore PCI-0123 toxicity, laser light dose, and drug dose effects on PDT response and to determine the most effective time for light application. The PDT response of PCI-0123-treated tumors was compared with that of Photofrin-treated tumors. RESULTS: Treatment of tumors with 150 J/cm2 of 740 nm laser light 5-6 hr after PCI-0123 administration (40 mg/kg) resulted in a 100% response rate and a 55% cure rate. Tumors treated with 150 J/cm2 of 630 nm laser light 24 hr after Photofrin administration (10 mg/kg) resulted in a 67% response rate and a 16% cure rate. CONCLUSION: PCI-0123 was found to be a more effective photosensitizer than Photofrin.


Assuntos
Terapia a Laser , Neoplasias Mamárias Animais/terapia , Metaloporfirinas/uso terapêutico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Adenocarcinoma , Animais , Éter de Diematoporfirina/uso terapêutico , Éter de Diematoporfirina/toxicidade , Feminino , Neoplasias Mamárias Animais/patologia , Metaloporfirinas/toxicidade , Camundongos , Camundongos Endogâmicos DBA , Células Tumorais Cultivadas/transplante
18.
J Photochem Photobiol B ; 52(1-3): 65-73, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10643074

RESUMO

Photodynamic therapy (PDT) induces among numerous cell targets membrane damage and alteration in cancer cell adhesiveness, an important parameter in cancer metastasis. We have previously shown that hematoporphyrin derivative (HPD)-PDT decreases cancer cell adhesiveness to endothelial cells in vitro and that it reduces the metastatic potential of cells injected into rats. The present study analyzes the influence of PDT in vivo on the metastatic potential of cancers cells and in vitro on the expression of molecules involved in adhesion and in the metastatic process. Photofrin and benzoporphyrin derivative monoacid ring A (BPD) have been evaluated on two colon cancer cell lines obtained from the same cancer [progressive (PROb) and regressive (REGb)] with different metastatic properties. Studies of BPD and Photofrin toxicity and phototoxicity are performed by colorimetric MTT assay on PROb and REGb cells to determine the PDT doses inducing around 25% cell death. Flow cytometry is then used to determine adhesion-molecule expression at the cell surface. ICAM-I, MHC-I, CD44V6 and its lectins (àHt1.3, PNA, SNA and UEA) are studied using cells treated either with BPD (50 ng/ml, 457 nm light, 10 J/cm2) or Photofrin (0.5 microgram/ml, 514 nm light, 25 J/cm2). Changes of metastatic patterns of PROb cells have been assessed by the subcutaneous injection of non-lethally treated BPD or Photofrin cells and counting lung metastases. First, we confirm the metastatic potential reduction induced by PDT with respectively a 71 or 96% decrease of the mean number of metastases (as compared with controls) for PROb cells treated with 50 ng/ml BPD and 10 or 20 J/cm2 irradiation. Concerning Photofrin-PDT-treated cells, we find respectively a 90 or 97% decrease (as compared with controls) of the mean number of metastases for PROb cells treated with 0.5 microgram/ml Photofrin and 25 or 50 J/cm2 irradiation. Then, we observe that CD44V6, its lectins (àHt1.3, PNA, SNA) and MHC-I are significantly decreased (compared with the other molecules tested) in PROb and REGb cells after both BPD and Photofrin PDT treatment. These modifications in adhesion-molecule expression, particularly of CD44V6, can thus account only for part of the decrease in the metastatic potential of PDT-treated cancer cells. Changes in adhesion-molecule expression induced by PDT are only transient, implying that the rate of metastatic reduction is probably not linked simply to these changes.


Assuntos
Neoplasias do Colo/patologia , Éter de Diematoporfirina/uso terapêutico , Glicoproteínas/genética , Receptores de Hialuronatos/genética , Molécula 1 de Adesão Intercelular/genética , Metástase Neoplásica/prevenção & controle , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/uso terapêutico , Animais , Adesão Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Éter de Diematoporfirina/toxicidade , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ratos , Células Tumorais Cultivadas
19.
Photochem Photobiol ; 66(4): 513-7, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9337624

RESUMO

Several studies have reported thrombus formation and/or the release of specific vasoactive eicosanoids, suggesting that platelet activation or damage after photodynamic therapy (PDT) may contribute to blood flow stasis. The role of circulating platelets on blood flow stasis and vascular leakage of macromolecules during and after PDT was assessed in an intravital animal model. Sprague-Dawley rats bearing chondrosarcoma on the right hind limb were injected intravenously (i.v.) with 25 mg/kg Photofrin 24 h before light treatment of 135 J/cm2 at 630 nm. Thrombocytopenia was induced in animals by administration of 3.75 mg/kg of rabbit anti-rat platelet antibody i.v. 30 min before the initiation of the light treatment. This regimen reduced circulating platelet levels from 300,000/mm3 to 20,000/mm3. Reductions in the luminal diameter of the microvasculature in normal muscle and tumor were observed in control animals given Photofrin and light. Venule leakage of macromolecules was noted shortly after the start of light treatment and continued throughout the period of observation. Animals made thrombocytopenic showed none of these changes after PDT in either normal tissues or tumor. The lack of vessel response correlated with the absence of thromboxane release in blood during PDT. These data suggest that platelets and eicosanoid release are necessary for vessel constriction and blood flow stasis after PDT using Photofrin.


Assuntos
Fotoquimioterapia/efeitos adversos , Trombocitopenia/etiologia , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Plaquetas/efeitos da radiação , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/efeitos da radiação , Éter de Diematoporfirina/toxicidade , Microcirculação/efeitos dos fármacos , Microcirculação/fisiopatologia , Microcirculação/efeitos da radiação , Fármacos Fotossensibilizantes/toxicidade , Coelhos , Ratos , Ratos Sprague-Dawley , Trombocitopenia/fisiopatologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/efeitos da radiação
20.
J Clin Laser Med Surg ; 15(2): 83-7, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9612183

RESUMO

Complicated hemangiomas are unique problems in which intervention with the proper laser can be an ideal solution. In this study we evaluated the toxicity of 5-Aminolevulinic acid (5-ALA) and Photofrin using in vitro models. The in vitro toxicity of 5-ALA and Photofrin was examined in a microvascular endothelial cell (MEC) culture system. The measurement of the percentage of MEC killed by various drug concentration using fluorescence viability assay. MEC incubated with 5-ALA at various concentrations for evaluation of dark toxicity showed less than a 50% cell kill. A comparison of different intervals of subcultured MEC showed that the early subculture (3 days after primary culture) is more vulnerable than later subculture (7 days after). Cells treated with Photofrin at various concentrations exhibited less than 50% cell kill (dark toxicity). The comparison of different intervals of subculture (3 days and 7 days after primary culture) showed a result similar to that of 5-ALA. All controls showed 0% cell kill. In conclusion, both 5-ALA and Photofrin are capable of destroying human microvascular endothelial cells in vitro. Drug concentrations and the power density for photodynamic therapy should be considered and will be included in our subsequent studies.


Assuntos
Ácido Aminolevulínico/toxicidade , Éter de Diematoporfirina/toxicidade , Endotélio Vascular/efeitos dos fármacos , Hemangioma/tratamento farmacológico , Sobrevivência Celular , Células Cultivadas/efeitos dos fármacos , Endotélio Vascular/citologia , Hemangioma/patologia , Humanos , Projetos Piloto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...