Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
Methods Mol Biol ; 2451: 559-567, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35505032

RESUMO

Photofrin-based photodynamic therapy (PDT) is approved for clinical use by the US Food and Drug Administration and the European Medicines Agency and is among the most widely used photosensitizer for the treatment of cancer. It was broadly reported that both the innate and the adaptive arms of immune response can be activated by PDT and play a critical role in the anticancer outcome of this treatment. PDT leads to the induction of acute local inflammation that includes leukocyte infiltration as well as increased activation and production of pro-inflammatory factors and cytokines. These events can lead to the development of systemic and specific antitumor immune response. Combining Photofrin-PDT with the epigenetic agent 5-aza-2'-deoxycytidine results in potentiated antitumor effects in vivo. Understanding the molecular mechanisms underlying this phenomenon would be invaluable for clinical development of this therapeutic approach. This chapter describes a detailed protocol allowing evaluation of specific antitumor immune response induced by PDT.


Assuntos
Éter de Diematoporfirina , Fotoquimioterapia , Decitabina/farmacologia , Decitabina/uso terapêutico , Éter de Diematoporfirina/farmacologia , Éter de Diematoporfirina/uso terapêutico , Epigênese Genética , Imunidade , Estados Unidos
2.
J Biomed Opt ; 24(11): 1-11, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31741351

RESUMO

The goal of our study was to determine the susceptibility of different pancreatic cell lines to clinically applicable photodynamic therapy (PDT). The efficacy of PDT of two different commercially available photosensitizers, verteporfin and sodium porfimer, was compared using a panel of four different pancreatic cancer cell lines, PANC-1, BxPC-3, CAPAN-2, and MIA PaCa-2, and an immortalized non-neoplastic pancreatic ductal epithelium cell line, HPNE. The minimum effective concentrations and dose-dependent curves of verteporfin and sodium porfimer on PANC-1 were determined. Since pancreatic cancer is known to have significant stromal components, the effect of PDT on stromal cells was also assessed. To mimic tumor-stroma interaction, a co-culture of primary human fibroblasts or human pancreatic stellate cell (HPSCs) line with PANC-1 was used to test verteporfin-PDT-mediated cell death of PANC-1. Two cytokines (TNF-α and IL-1ß) were used for stimulation of primary fibroblasts (derived from human esophageal biopsies) or HPSCs. The increased expression of smooth muscle actin (α-SMA) confirmed the activation of fibroblasts or HPSC upon treatment with TNF-α and IL-1ß. Cell death assays showed that both sodium porfimer- and verteporfin-mediated PDT-induced cell death in a dose-dependent manner. However, verteporfin-PDT treatment had a greater efficiency with 60 × lower concentration than sodium porfimer-PDT in the PANC-1 incubated with stimulated fibroblasts or HPSC. Moreover, activation of stromal cells did not affect the treatment of the pancreatic cancer cell lines, suggesting that the effects of PDT are independent of the inflammatory microenvironment found in this two-dimensional culture model of cancers.


Assuntos
Éter de Diematoporfirina/farmacologia , Neoplasias Pancreáticas/terapia , Fotoquimioterapia , Células Estromais/citologia , Verteporfina/farmacologia , Biópsia , Morte Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Ensaios de Seleção de Medicamentos Antitumorais , Fibroblastos/citologia , Humanos , Microscopia de Fluorescência , Pâncreas/citologia , Células Estromais/efeitos dos fármacos , Microambiente Tumoral
3.
Lasers Surg Med ; 51(3): 301-308, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30615224

RESUMO

BACKGROUND AND OBJECTIVE: Photodynamic therapy (PDT) has been widely used to treat malignant tumors. Our previous studies indicated that connexin (Cx) 32- and Cx26-composed gap junctional intercellular communication (GJIC) could improve the phototoxicity of PDT. However, the role of heterotypic Cx32/Cx26-formed GJIC in PDT phototoxicity is still unknown. Thus, the present study was aimed to investigate the effect of Cx32/Cx26-formed GJIC on PDT efficacy. METHODS: CCK8 assay was used to detect cell survival after PDT. Western blot assay was utilized to detect Cx32/Cx26 expression. "Parachute" dye-coupling assay was performed to measure the function of GJ channels. The intracellular Ca2+ concentrations were determined using flow cytometer. ELISA assay was performed to detect the intracellular levels of PGE2 and cAMP. RESULTS: The present study demonstrates there is a Cx32/Cx26-formed GJIC-dependent reduction of phototoxicity when cells were exposure to low concentration of Photofrin. Such a protective action is missing at low cell density due to the lack of GJ coupling. Under high-cell density condition, where there is opportunity for the cells to contact each other and form GJ, suppressing Cx32/Cx26-formed GJIC by either inhibiting the expression of Cx32/Cx26 or pretreating with GJ channel inhibitor augments PDT phototoxicity after cells were treated with at 2.5 µg/ml Photofrin. The above results suggest that at low Photofrin concentration, the presence of Cx32/Cx26-formed GJIC may decrease the phototoxicity of PDT, leading to the insensitivity of malignant cells to PDT treatment. The GJIC-mediated PDT insensitivity was associated with Ca2+ and prostaglandin E2 (PGE2 ) signaling pathways. CONCLUSION: The present study provides a cautionary note that for tumors expressing Cx32/Cx26, the presence of Cx32/Cx26-composed GJIC may cause the resistance of tumor cells to PDT. Oppositely, treatment strategies designed to downregulate the expression of Cx32/Cx26 or restrain the function of Cx32/Cx26-mediated GJIC may increase the sensitivity of malignant cell to PDT. Lasers Surg. Med. 51:301-308, 2019. © 2019 Wiley Periodicals, Inc.


Assuntos
Comunicação Celular/efeitos da radiação , Conexina 26/fisiologia , Conexinas/fisiologia , Junções Comunicantes/efeitos da radiação , Células HeLa/efeitos da radiação , Fotoquimioterapia/efeitos adversos , Técnicas de Cultura de Células , Sobrevivência Celular , Éter de Diematoporfirina/farmacologia , Células HeLa/patologia , Humanos , Fármacos Fotossensibilizantes/farmacologia , Proteína beta-1 de Junções Comunicantes
4.
Bioelectrochemistry ; 123: 9-18, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29715586

RESUMO

Electrochemotherapy became one of the therapeutic protocols successfully used in oncology. However, biological effects occurring in cells, especially those which are drug resistant, have not been studied thoroughly. This study presents response of wild and drug resistant breast cancer cells to classical photodynamic therapy with Photofrin or experimental photodynamic therapy with cyanine IR-775, combined with electroporation. Photodynamic reaction or electroporation alone had no cytotoxic effect, but their combination significantly disturbed cellular functions. Applying electroporation allowed the drugs to increase its accumulation, especially for a poorly permeant cyanine in drug resistant cells. FACS analysis showed that even at relatively mild electric field, ca. 90% of cells were permeabilized. High intracellular concentration of drugs triggered the cellular defense system through increased expression of glutathione S-transferase and multidrug resistance proteins (MDR1 and MRP7), particularly in drug resistant cells. Finally, expressively decreased cell metabolism and proliferation, as well as formation of apoptotic bodies and fragmentation of cells were observed after the combined treatment. The results show that electroporation can be used for effective delivery of photosensitizers, even to drug resistant breast cancer cells, which was not tested before. This shows that electro-photodynamic treatment could be a promising approach to overcome a problem of drug resistance in cancer cells.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Carbocianinas/farmacologia , Éter de Diematoporfirina/farmacologia , Eletroquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carbocianinas/administração & dosagem , Carbocianinas/farmacocinética , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Sobrevivência Celular/efeitos dos fármacos , Éter de Diematoporfirina/administração & dosagem , Éter de Diematoporfirina/farmacocinética , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Células MCF-7 , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/farmacocinética
5.
Oncol Rep ; 40(1): 339-345, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29749554

RESUMO

Concurrent low­dose carboplatin/Photofrin® photodynamic therapy (ccPDT) has been shown to promote relapse­free complete tumor regression in cervical or endometrial cancer patients as a fertility­preservation therapy. This study aimed to investigate the molecular mechanism of the enhanced therapeutic efficacy of ccPDT by determining intracellular reactive oxygen species (ROS) and necrotic or apoptotic cell damage in HeLa cells loaded with fluorescent oxidant agents and Photofrin or/and carboplatin under light irradiation. The cytotoxic effects of ccPDT were compared when monitored with a light dose under carboplatin or Photofrin alone. Photofrin­PDT alone did not enhance either hydroxyl radicals (OH•) or superoxide anions (O2•-), but a slight enhancement of hydrogen peroxide (H2O2) production was observed. A larger enhancement of ROS production was obtained in a dose­dependent manner following ccPDT, especially OH• and H2O2, in conjunction with both necrotic and apoptotic cell death, compared with necrotic­prone PDT alone. The carboplatin­mediated Fenton reaction: 2[PtII]2 + H2O2 → [Pt2.25]4 + OH¯+ OH• was proposed to explain the dose­dependent enhancement of OH•. In conclusion, the therapeutic enhancement of ccPDT in vitro was attributable to the carboplatin­mediated synergetic production of OH▪ and apoptotic cellular damage, compared with Photofrin­PDT alone.


Assuntos
Carboplatina/farmacologia , Recidiva Local de Neoplasia/tratamento farmacológico , Fotoquimioterapia , Neoplasias do Colo do Útero/tratamento farmacológico , Apoptose/efeitos dos fármacos , Éter de Diematoporfirina/farmacologia , Feminino , Preservação da Fertilidade/métodos , Células HeLa , Humanos , Peróxido de Hidrogênio/metabolismo , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/fisiopatologia , Fármacos Fotossensibilizantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/fisiopatologia
6.
Photodiagnosis Photodyn Ther ; 22: 241-244, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29702258

RESUMO

Photodynamic therapy (PDT) is an office-based treatment for precancerous and early cancerous skin changes. PDT induces cell death through the production of reactive oxygen species (ROS). Cyclobutane pyrimidine dimers (CPDs) are the most important DNA changes responsible for ultraviolet (UV) carcinogenesis. Recently ROS induced by UVA were shown to generate CPDs via activating melanin. This raised the possibility that PDT induced ROS may also induce CPDs and mutagenesis in melanin containing cells. Previously the effect of PDT on CPDs in melanin containing cells has not been assessed. Our current work aimed to compare the generation of CPDs in melanin containing cells subjected to UVA treatment and porfimer sodium red light PDT. We used ELISA to detect CPDs. After UVA we found a dose dependent increase in CPDs in melanoma cells (B16-F10, MNT-1) with CPD levels peaking hours after discontinuation of UVA treatment. This indicated the generation of UVA induced dark-CPDs in the model. Nevertheless, PDT in biologically relevant doses was unable to induce CPDs. Our work provides evidence for the lack of CPD generation by PDT in melanin containing cells.


Assuntos
Éter de Diematoporfirina/farmacologia , Melaninas/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Dímeros de Pirimidina/biossíntese , Raios Ultravioleta/efeitos adversos , Dano ao DNA/fisiologia , Ensaio de Imunoadsorção Enzimática , Humanos , Melanócitos/efeitos dos fármacos , Melanoma/tratamento farmacológico
7.
BMC Cancer ; 18(1): 210, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29463237

RESUMO

BACKGROUND: Accumulating evidence suggest that autophagy plays a pivotal role in various anticancer therapies, including photodynamic therapy (PDT), acting as a pro-death or pro-survival mechanism in a context-dependent manner. Therefore, we aimed to determine the role of autophagy in Photofrin-based PDT. METHODS: In vitro cytotoxic/cytostatic effects of PDT were evaluated with crystal violet cell viability assay. Autophagy induction was analyzed by immunoblotting and immunofluorescence using anti-LC3 antibody. Autophagy was inhibited by shRNA-mediated ATG5 knockdown or CRISPR/Cas9-mediated ATG5 knockout. Apoptosis was assessed by flow cytometry analysis of propidium iodide and anexin V-positive cells as well as by detection of cleaved PARP and caspase 3 proteins using immunoblotting. Protein carbonylation was evaluated by the 2,4-dinitrophenylhydrazine (DNPH) method. RESULTS: Photofrin-PDT leads to robust autophagy induction in two cancer cell lines, Hela and MCF-7. shRNA-mediated knockdown of ATG5 only partially blocks autophagic response and only marginally affects the sensitivity of Hela and MCF-7 cells to PDT. ATG5 knockout in HeLa cell line utilizing CRISPR/Cas9 genome editing results in increased PDT-mediated cytotoxicity, which is accompanied by an enhanced apoptotic response and increased accumulation of carbonylated proteins. CONCLUSIONS: Altogether, these observations imply that autophagy contributes to Photofrin-PDT resistance by enabling clearance of carbonylated and other damaged proteins. Therefore, autophagy inhibition may serve as a strategy to improve PDT efficacy.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Autofagia/efeitos da radiação , Éter de Diematoporfirina/farmacologia , Fármacos Fotossensibilizantes/farmacologia , Autofagia/genética , Proteína 5 Relacionada à Autofagia/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Expressão Gênica , Técnicas de Silenciamento de Genes , Marcação de Genes , Humanos , Luz , Fotoquimioterapia , Interferência de RNA , RNA Interferente Pequeno/genética
8.
J Photochem Photobiol B ; 179: 46-53, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29331658

RESUMO

Sulforaphene (SFE), a natural isothiocyanate from cruciferous vegetables has shown a potential anticancer effect against cervical and lung cancer. Palliative treatments like photodynamic therapy (PDT) are being implemented for a long time however, the results are still not promising in case of aggressive cancers like anaplastic thyroid cancer. The objective of this work is to establish an alternative method with the combination of photofrin-PDT and sulforaphene, a natural isothiocyanate from cruciferous vegetables, against human anaplastic thyroid cancer to enhance the efficacy of PDT. In this study, cell viability of FRO cells due to combination treatment was analyzed by MTT assay, Cell cycle arrest, MMP depolarization and ROS generation, analyzed by flow cytometry. Western blot analysis of various proliferative proteins was performed to assess the activity of combination treatment against FRO cells. From the results, sulforaphene alone showed no cytotoxicity against normal cells, however, combination of sulforaphene and photofrin mediated PDT showed a noticeable decrease in cell proliferation against FRO cells. Combination treatment synergistically caused cell cycle arrest via ROS generation and MMP depolarization. The expressions of Ras, MEK, ERK, B-Raf proteins significantly modulated due to combination treatment. PDT and SFE can induce apoptosis in anaplastic thyroid cancer cells individually but while treated in combination, it enhanced the apoptotic and anti-proliferative effect, much higher than the individual doses. In summary, our work designates sulforaphene as a unique natural enhancer of efficacy with PDT against anaplastic thyroid cancer.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Isotiocianatos/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Quinases raf/metabolismo , Proteínas ras/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Éter de Diematoporfirina/farmacologia , Éter de Diematoporfirina/uso terapêutico , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Humanos , Isotiocianatos/uso terapêutico , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Carcinoma Anaplásico da Tireoide/tratamento farmacológico , Carcinoma Anaplásico da Tireoide/metabolismo , Carcinoma Anaplásico da Tireoide/patologia , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia
9.
J Biophotonics ; 10(12): 1586-1596, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28417552

RESUMO

In spite of the promising initial treatment responses presented by photodynamic therapy (PDT), 5-year recurrence rates remain high level. Therefore, improvement in the efficacy of PDT is needed. There are reports showing that connexin(Cx) 26-composed gap junctional intercellular communication (GJIC) enhances the intercellular propagation of "death signal", thereby increasing chemotherapeutic cytotoxicity. However, it is unclear whether Cx26-formed GJIC has an effect on PDT phototoxicity. The results in the present study showed that Cx26-composed GJ formation at high density enhances the phototoxicity of Photofrin-PDT. When the Cx26 is not expressed or Cx26 channels are blocked, the phototoxicity in high-density cultures substantially reduces, indicating that the enhanced PDT phototoxicity at high density is mediated by Cx26-composed GJIC. The GJIC-mediated increase in PDT phototoxicity was associated with ROS, calcium and lipid peroxide-mediated stress signaling pathways. The work presents the ability of Cx26-composed GJIC to enhance the sensitivity of malignant cells to PDT, and indicates that maintenance or increase of Cx26-formed GJIC may be a profitable strategy towards the enhancement of PDT therapeutic efficiency. Picture: The survival response of Photofrin-PDT in Dox-treated (Cx26 expressing, GJ-formed) and Dox-untreated cells (Cx26 non-expressing, GJ-unformed) at high-cell density condition.


Assuntos
Cálcio/metabolismo , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/efeitos da radiação , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Peróxidos Lipídicos/metabolismo , Fotoquimioterapia , Aldeídos/metabolismo , Ceramidas/biossíntese , Conexina 26 , Éter de Diematoporfirina/farmacologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Espaço Extracelular/efeitos da radiação , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/efeitos da radiação , Células HeLa , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Espaço Intracelular/efeitos da radiação
10.
ACS Infect Dis ; 3(4): 320-328, 2017 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-28207234

RESUMO

It is known that noncationic porphyrins such as Photofrin (PF) are effective in mediating antimicrobial photodynamic inactivation (aPDI) of Gram-positive bacteria or fungi. However, the aPDI activity of PF against Gram-negative bacteria is accepted to be extremely low. Here we report that the nontoxic inorganic salt potassium iodide (KI) at a concentration of 100 mM when added to microbial cells (108/mL) + PF (10 µM hematoporphyrin equivalent) + 415 nm light (10 J/cm2) can eradicate (>6 log killing) five different Gram-negative species (Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumoniae, Proteus mirabilis, and Acinetobacter baumannii), whereas no killing was obtained without KI. The mechanism of action appears to be the generation of microbicidal molecular iodine (I2/I3-) as shown by comparable bacterial killing when cells were added to the mixture after completion of illumination and light-dependent generation of iodine as detected by the formation of the starch complex. Gram-positive methicillin-resistant Staphylococcus aureus is much more sensitive to aPDI (200-500 nM PF), and in this case potentiation by KI may be mediated mainly by short-lived iodine reactive species. The fungal yeast Candida albicans displayed intermediate sensitivity to PF-aPDI, and killing was also potentiated by KI. The reaction mechanism occurs via singlet oxygen (1O2). KI quenched 1O2 luminescence (1270 nm) at a rate constant of 9.2 × 105 M-1 s-1. Oxygen consumption was increased when PF was illuminated in the presence of KI. Hydrogen peroxide but not superoxide was generated from illuminated PF in the presence of KI. Sodium azide completely inhibited the killing of E. coli with PF/blue light + KI.


Assuntos
Antibacterianos/farmacologia , Éter de Diematoporfirina/farmacologia , Fármacos Fotossensibilizantes/farmacologia , Iodeto de Potássio/farmacologia , Antibacterianos/química , Candida albicans/efeitos dos fármacos , Éter de Diematoporfirina/química , Sinergismo Farmacológico , Bactérias Gram-Negativas/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Fármacos Fotossensibilizantes/química , Iodeto de Potássio/química
11.
Photochem Photobiol ; 93(2): 609-612, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27935055

RESUMO

In prior studies, we have identified the ability of low-level lysosomal photodamage to potentiate the phototoxic effect of subsequent photodamage to mitochondria. The mechanism involves calpain-mediated cleavage of the autophagy-associated protein ATG5 to form a proapoptotic fragment (tATG5). In this report, we explore the permissible time lag between the two targeting procedures along with the effect of simultaneously targeting both lysosomes and mitochondria. This was found to be as effective as the sequential protocol with no gap between the irradiation steps. Inhibition of calpain reversed the enhanced efficacy of the "simultaneous" protocol. It appears that even a minor level of lysosomal photodamage can have a significant effect on the efficacy of subsequent mitochondrial photodamage. We propose that these results may explain the efficacy of Photofrin, a photosensitizing product that also targets both lysosomes and mitochondria for photodamage.


Assuntos
Éter de Diematoporfirina/farmacologia , Lisossomos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Animais , Proteína 5 Relacionada à Autofagia/metabolismo , Linhagem Celular Tumoral , Luz , Lisossomos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Análise Espectral/métodos
12.
Oncol Rep ; 35(6): 3151-8, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27035643

RESUMO

Photodynamic therapy (PDT) exerts direct cytotoxic effects on tumor cells, destroys tumor blood and lymphatic vessels and induces local inflammation. Although PDT triggers the release of immunogenic antigens from tumor cells, the degree of immune stimulation is regimen-dependent. The highest immunogenicity is achieved at sub-lethal doses, which at the same time trigger cytoprotective responses, that include increased expression of glucose-regulated protein 78 (GRP78). To mitigate the cytoprotective effects of GRP78 and preserve the immunoregulatory activity of PDT, we investigated the in vivo efficacy of PDT in combination with EGF-SubA cytotoxin that was shown to potentiate in vitro PDT cytotoxicity by inactivating GRP78. Treatment of immunocompetent BALB/c mice with EGF-SubA improved the efficacy of PDT but only when mice were treated with a dose of EGF-SubA that exerted less pronounced effects on the number of T and B lymphocytes as well as dendritic cells in mouse spleens. The observed antitumor effects were critically dependent on CD8+ T cells and were completely abrogated in immunodeficient SCID mice. All these results suggest that GRP78 targeting improves in vivo PDT efficacy provided intact T-cell immune system.


Assuntos
Antineoplásicos/administração & dosagem , Fator de Crescimento Epidérmico/administração & dosagem , Proteínas de Escherichia coli/administração & dosagem , Proteínas de Choque Térmico/metabolismo , Subtilisinas/administração & dosagem , Animais , Linhagem Celular Tumoral , Terapia Combinada , Éter de Diematoporfirina/farmacologia , Chaperona BiP do Retículo Endoplasmático , Feminino , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Proteínas Recombinantes de Fusão/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Lasers Med Sci ; 30(9): 2353-61, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26450615

RESUMO

Esophageal squamous cell carcinoma (ESCC), the most prevalent cell type of esophageal cancer, remains a dismal disease with poor prognosis. Photodynamic therapy (PDT) is a minimally invasive treatment option for early esophageal cancer. To explore possible factors involved in resistance to PDT in esophageal cancer cells, we selected PDT-resistant subcell lines by repeated treatment of CE48T/VGH (CE48T) ESCC cells with Photofrin-PDT and then analyzed the global gene modulations in the PDT-resistant cells by whole-genome microarray. More than 700 genes reached a fold change greater than 1.5 in each of the PDT-resistant cells compared to parental cells. Among these genes, both tumor necrosis factor (TNF) and EFNA1 genes were significantly upregulated in resistant cell lines. However, they were significantly downregulated in Photofrin-PDT-treated cells compared to untreated cells. The observations made in the microarray analysis were further confirmed by quantitative PCR. We observed that recombinant tumor necrosis factor alpha (TNF-α) activated the gene expression of EFNA1 at both the messenger RNA (mRNA) level and the protein level in CE48T cells. Functional analysis showed that when incubated with oligomeric and monomeric ephrin-A1 simultaneously, ESCC cells became significantly resistant to Photofrin-PDT. Functional analysis further suggested that transmembrane and soluble ephrin-A1 may cooperate to enhance resistance to Photofrin-PDT in ESCC cells.


Assuntos
Carcinoma de Células Escamosas/patologia , Éter de Diematoporfirina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Efrina-A1/farmacologia , Neoplasias Esofágicas/patologia , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Éter de Diematoporfirina/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/efeitos da radiação , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago , Humanos , Fármacos Fotossensibilizantes/uso terapêutico , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação
14.
Redox Biol ; 6: 157-168, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26225731

RESUMO

Intracellular singlet oxygen generation in photofrin-loaded cells caused cell death without discrimination between nonmalignant and malignant cells. In contrast, extracellular singlet oxygen generation caused apoptosis induction selectively in tumor cells through singlet oxygen-mediated inactivation of tumor cell protective catalase and subsequent reactivation of intercellular ROS-mediated apoptosis signaling through the HOCl and the NO/peroxynitrite signaling pathway. Singlet oxygen generation by extracellular photofrin alone was, however, not sufficient for optimal direct inactivation of catalase, but needed to trigger the generation of cell-derived extracellular singlet oxygen through the interaction between H2O2 and peroxynitrite. Thereby, formation of peroxynitrous acid, generation of hydroxyl radicals and formation of perhydroxyl radicals (HO2(.)) through hydroxyl radical/H2O2 interaction seemed to be required as intermediate steps. This amplificatory mechanism led to the formation of singlet oxygen at a sufficiently high concentration for optimal inactivation of membrane-associated catalase. At low initial concentrations of singlet oxygen, an additional amplification step needed to be activated. It depended on singlet oxygen-dependent activation of the FAS receptor and caspase-8, followed by caspase-8-mediated enhancement of NOX activity. The biochemical mechanisms described here might be considered as promising principle for the development of novel approaches in tumor therapy that specifically direct membrane-associated catalase of tumor cells and thus utilize tumor cell-specific apoptosis-inducing ROS signaling.


Assuntos
Catalase/antagonistas & inibidores , Éter de Diematoporfirina/farmacologia , Regulação Neoplásica da Expressão Gênica , Fármacos Fotossensibilizantes/farmacologia , Transdução de Sinais , Oxigênio Singlete/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Caspase 8/genética , Caspase 8/metabolismo , Catalase/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Ácido Hipocloroso/metabolismo , Luz , Metaloporfirinas/farmacologia , Camundongos , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Oxigênio Singlete/metabolismo , Sulfonas/farmacologia , Taurina/farmacologia , Receptor fas/genética , Receptor fas/metabolismo
15.
Biomed Pharmacother ; 71: 30-6, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25960212

RESUMO

Ovarian cancer is among the most lethal cancers in women. The successful anticancer treatment depends on the effectiveness of cytotoxic effect of applied therapeutic procedures either alone or in combination with other treatments. Photodynamic therapy (PDT) is a relatively new method of anticancer therapy. Its dominant mechanism of action is the over-production of reactive oxygen species induced by oxidative stress in malignant cells, which attack lipid membranes, proteins and nucleic acids. One of the important mechanisms is induction of unfolded protein response, ubiquitin-proteasome pathway of protein degradation. The aim of this study was to evaluate the cytotoxic effect of various protective enzymes in ovarian carcinoma clear cell line in comparison to the model breast cell line after photodynamic reaction and photodynamic reaction with 2-methoxyestradiol (2-Me). Human malignant ovarian cell line (OvBH-1) was used and human breast adenocarcinoma cells (MCF-7) were used as a control. Photodynamic reaction (PDR) with Photofrin(®)II and Ph(®)II with 2-Me was performed. The expression of protective proteins by immunocytochemistry (HSP70 and iNOS) and western blot (Hsp27 and Hsp70) methods was evaluated directly, 3 and 6 h after PDR. The changes in cells' cytoskeleton were evaluated using immunofluorescence by confocal microscopy. The expression of iNOS was observed for both experiments with differential intensity and quantity. A higher expression of Hsp70 in MCF-7 cells was observed than in OvBh-1 cells. The reorganization of cytoskeleton and nucleus was observed after 3 and 6 h after exposition to light.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Éter de Diematoporfirina/uso terapêutico , Estradiol/análogos & derivados , Neoplasias Ovarianas/tratamento farmacológico , Fotoquimioterapia , 2-Metoxiestradiol , Adenocarcinoma/patologia , Western Blotting , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Éter de Diematoporfirina/farmacologia , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Imuno-Histoquímica , Óxido Nítrico Sintase Tipo II/metabolismo , Neoplasias Ovarianas/patologia , Oxirredução/efeitos dos fármacos
16.
Melanoma Res ; 25(3): 210-24, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25756554

RESUMO

Photodynamic therapy has been considered ineffective for melanomas because of the competition between the absorbance of melanin from the melanoma and the absorbance of photosensitizers at the photosensitizer excitation light wavelength. Melanomas show considerable heterogeneity and resistance to phototherapy. The effectiveness of photodynamic therapy could be intensified by electroporation for enhanced transport of a photosensitizer by transient pores in the membrane. In this study, photodynamic therapy combined with electroporation was tested in vitro on the human melanoma cell lines melanotic melanoma (MeWo) and amelanotic melanoma (C32). Control experiments were conducted on human keratinocytes (HaCaT). Photofrin was used as a photosensitizer. Photosensitizer distribution, cloning efficacy test, comet assay, and assessment of apoptotic proteins were performed. Melanin levels were determined before and after photodynamic therapy. The experiments indicated that electroporation effectively supports the photodynamic method. It was found that photodynamic therapy with electroporation efficiently induces apoptosis in melanotic and amelanotic melanoma cells.


Assuntos
Antineoplásicos/farmacologia , Éter de Diematoporfirina/farmacologia , Eletroquimioterapia , Melanoma Amelanótico/tratamento farmacológico , Melanoma/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Absorção Fisiológica/efeitos da radiação , Antineoplásicos/efeitos adversos , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caspases/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Ensaio Cometa , Dano ao DNA , Éter de Diematoporfirina/efeitos adversos , Éter de Diematoporfirina/metabolismo , Eletroquimioterapia/efeitos adversos , Eletroporação , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , Melaninas/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Melanoma Amelanótico/metabolismo , Melanoma Amelanótico/patologia , Proteínas de Neoplasias/metabolismo , Fotoquimioterapia/efeitos adversos , Fármacos Fotossensibilizantes/efeitos adversos , Fármacos Fotossensibilizantes/metabolismo , Fatores de Tempo
17.
J Biophotonics ; 8(9): 764-74, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25597481

RESUMO

Despite initially positive responses, recurrences after Photodynamic treatment (PDT) can occur and there is need for improvement in the effectiveness of PDT. Our study uniquely showed that there was a significantly gap junctional intercellular communication (GJIC)-dependent PDT cytotoxicity. The presence of GJIC composed of Connexin 32 increased the PDT phototoxicity in transfected HeLa cells and in the xenograft tumors, and the enhanced phototoxicity of Photofrin-mediated PDT by GJIC was related with ROS and calcium pathways. Our study indicates the possibility that up-regulation or maintenance of gap junction functionality may be used to increase the efficacy of PDT. The phototoxicity effect of Photofrin was substantially greater in Dox-treated cells, which expressed the Cx32 and formed the GJ, than Dox-untreated.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Éter de Diematoporfirina/farmacologia , Junções Comunicantes/patologia , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transporte Biológico , Cálcio/metabolismo , Sinalização do Cálcio/efeitos da radiação , Comunicação Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Éter de Diematoporfirina/metabolismo , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Junções Comunicantes/efeitos da radiação , Células HeLa , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Espaço Intracelular/efeitos da radiação , Fármacos Fotossensibilizantes/metabolismo
18.
Cancer Immunol Immunother ; 64(3): 287-97, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25384911

RESUMO

Effective therapy for advanced cancer often requires treatment of both primary tumors and systemic disease that may not be apparent at initial diagnosis. Numerous studies have shown that stimulation of the host immune system can result in the generation of anti-tumor immune responses capable of controlling metastatic tumor growth. Thus, there is interest in the development of combination therapies that both control primary tumor growth and stimulate anti-tumor immunity for control of metastatic disease and subsequent tumor growth. Photodynamic therapy (PDT) is an FDA-approved anticancer modality that has been shown to enhance anti-tumor immunity. Augmentation of anti-tumor immunity by PDT is regimen dependent, and PDT regimens that enhance anti-tumor immunity have been defined. Unfortunately, these regimens have limited ability to control primary tumor growth. Therefore, a two-step combination therapy was devised in which a tumor-controlling PDT regimen was combined with an immune-enhancing PDT regimen. To determine whether the two-step combination therapy enhanced anti-tumor immunity, resistance to subsequent tumor challenge and T cell activation and function was measured. The ability to control distant disease was also determined. The results showed that the novel combination therapy stimulated anti-tumor immunity while retaining the ability to inhibit primary tumor growth of both murine colon (Colon26-HA) and mammary (4T1) carcinomas. The combination therapy resulted in enhanced tumor-specific T cell activation and controlled metastatic tumor growth. These results suggest that PDT may be an effective adjuvant for therapies that fail to stimulate the host anti-tumor immune response.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Fotoquimioterapia/métodos , Animais , Antineoplásicos/farmacologia , Clorofila/análogos & derivados , Clorofila/farmacologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Éter de Diematoporfirina/farmacologia , Feminino , Ativação Linfocitária , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Fármacos Fotossensibilizantes/farmacologia , Distribuição Aleatória , Linfócitos T/imunologia , Transfecção
19.
Lasers Surg Med ; 46(9): 712-7, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25154610

RESUMO

BACKGROUND AND OBJECTIVE: Moraxella catarrhalis is a significant cause of pediatric otitis media (OM), which is the most prevalent bacterial infection in children and primary reason for antibiotic administration in this population. Moreover, biofilm formation has been implicated as a primary mechanism of chronic or recurrent OM disease. As bacterial biofilms are inherently resistant to most antibiotics and these complex structures also present a significant challenge to the immune system, there is a clear need to identify novel antimicrobial approaches to treat OM infections. In this study, we evaluated the potential efficacy of antibacterial photodynamic therapy (aPDT) with porfimer sodium (Photofrin (PF)) against planktonic as well as biofilm-associated M. catarrhalis. MATERIALS AND METHODS: The bactericidal activity of aPDT with PF was assessed against multiple recent clinical isolates of M. catarrhalis grown planktonically as well as in biofilms. The bactericidal activity of PF-aPDT was quantified by enumeration of colony forming units post-treatment. The effect of aPDT on M. catarrhalis biofilms was further investigated with scanning electron microscopy (SEM) imaging. RESULTS: aPDT with PF significantly reduced M. catarrhalis viability. Although PF-aPDT caused higher killing in planktonic grown organisms (5-6 log kill), biofilm grown bacteria also demonstrated a statistically significant reduction in viable organisms (3-4 log decrease in recoverable bacteria) following treatment as compared to saline only controls (P < 0.01). SEM studies indicated the PF-aPDT treated bacteria exhibited prominent morphological changes with visibly distorted cell membranes. CONCLUSIONS: aPDT with PF elicits significant bactericidal activity against both planktonic and biofilm-associated M. catarrhalis, suggesting this technology warrants further analysis as a potential novel antimicrobial treatment for acute or recurrent OM.


Assuntos
Biofilmes/efeitos dos fármacos , Éter de Diematoporfirina/farmacologia , Moraxella catarrhalis/efeitos dos fármacos , Moraxella catarrhalis/crescimento & desenvolvimento , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Biofilmes/crescimento & desenvolvimento , Biofilmes/efeitos da radiação , Lasers de Corante , Lasers de Estado Sólido , Viabilidade Microbiana/efeitos dos fármacos , Viabilidade Microbiana/efeitos da radiação , Moraxella catarrhalis/efeitos da radiação
20.
Cell Death Dis ; 4: e741, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23887632

RESUMO

Glucose-regulated protein 78 (GRP78) is an endoplasmic reticulum (ER)-resident chaperone and a major regulator of the unfolded protein response (UPR). Accumulating evidence indicate that GRP78 is overexpressed in many cancer cell lines, and contributes to the invasion and metastasis in many human tumors. Besides, GRP78 upregulation is detected in response to different ER stress-inducing anticancer therapies, including photodynamic therapy (PDT). This study demonstrates that GRP78 mRNA and protein levels are elevated in response to PDT in various cancer cell lines. Stable overexpression of GRP78 confers resistance to PDT substantiating its cytoprotective role. Moreover, GRP78-targeting subtilase cytotoxin catalytic subunit fused with epidermal growth factor (EGF-SubA) sensitizes various cancer cells to Photofrin-mediated PDT. The combination treatment is cytotoxic to apoptosis-competent SW-900 lung cancer cells, as well as to Bax-deficient and apoptosis-resistant DU-145 prostate cancer cells. In these cells, PDT and EGF-SubA cytotoxin induce protein kinase R-like ER kinase and inositol-requiring enzyme 1 branches of UPR and also increase the level of C/EBP (CCAAT/enhancer-binding protein) homologous protein, an ER stress-associated apoptosis-promoting transcription factor. Although some apoptotic events such as disruption of mitochondrial membrane and caspase activation are detected after PDT, there is no phosphatidylserine plasma membrane externalization or DNA fragmentation, suggesting that in DU-145 cells the late apoptotic events are missing. Moreover, in SW-900 cells, EGF-SubA cytotoxin potentiates PDT-mediated cell death but attenuates PDT-induced apoptosis. In addition, the cell death cannot be reversed by caspase inhibitor z-VAD, confirming that apoptosis is not a major cell death mode triggered by the combination therapy. Moreover, no typical features of necrotic or autophagic cell death are recognized. Instead, an extensive cellular vacuolation of ER origin is observed. Altogether, these findings indicate that PDT and GRP78-targeting cytotoxin treatment can efficiently kill cancer cells independent on their apoptotic competence and triggers an atypical, non-apoptotic cell death.


Assuntos
Éter de Diematoporfirina/farmacologia , Proteínas de Choque Térmico/metabolismo , Neoplasias/tratamento farmacológico , Fotoquimioterapia/métodos , Subtilisinas/farmacologia , Chaperona BiP do Retículo Endoplasmático , Humanos , Terapia de Alvo Molecular , Neoplasias/metabolismo , Neoplasias/patologia , Microambiente Tumoral , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...