Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Cancer Res ; 61(16): 6185-93, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11507071

ABSTRACT

Neuroblastomas can acquire a sustained high-level drug resistance during chemotherapy and especially myeloablative chemoradiotherapy. p53 mutations are rare in primary neuroblastomas, but a loss of p53 function could play a role in multidrug resistance. We determined p53 function by measuring induction of p21 and/or MDM2 proteins in response to melphalan (L-PAM) in seven L-PAM-sensitive and 11 L-PAM-resistant neuroblastoma cell lines. p53 was functional in seven/seven drug-sensitive but in only 4/11 drug-resistant cell lines (P = 0.01). In four of the seven cell lines lacking p53 function, mutations of p53 were detected by the microarray GeneChip p53 Assay and automated sequencing, whereas six cell lines with functional p53 had no evidence of p53 mutations. All of the cell lines with wild-type (wt) p53 showed a strong transactivation of the p53-HBS/CAT reporter gene, whereas the four cell lines with mutant p53 failed to transactivate p53 HBS/CAT. Overexpression of MDM2 protein (relative to p53 functional lines) was seen in two p53-nonfunctional cell lines with wt p53; one showed genomic amplification of MDM2. Nonfunctional and mutated p53 was detected in a resistant cell line, whereas a sensitive cell line derived from the same patient before treatment had functional and wt p53. Loss of p53 function was selectively achieved by transduction of human papillomavirus 16 E6 (which degrades p53) into two drug-sensitive neuroblastoma cell lines with intact p53, causing high-level drug resistance to L-PAM, carboplatin, and etoposide. These data obtained with neuroblastoma cell lines suggest that the high-level drug resistance observed in some recurrent neuroblastomas is attributable to p53 mutations and/or a loss of p53 function acquired during chemotherapy. If confirmed in patient tumor samples, these data support development of p53-independent therapies for consolidation and/or salvage of recurrent neuroblastomas.


Subject(s)
Drug Resistance, Multiple/physiology , Neuroblastoma/drug therapy , Nuclear Proteins , Repressor Proteins , Tumor Suppressor Protein p53/physiology , Antineoplastic Agents/pharmacology , Carboplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Etoposide/pharmacology , Gene Amplification , Humans , Melphalan/pharmacology , Mutation , Neuroblastoma/genetics , Neuroblastoma/metabolism , Oncogene Proteins, Viral/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Transcriptional Activation , Transduction, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
2.
Cancer Res ; 60(15): 4066-9, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10945611

ABSTRACT

Photodynamic therapy (PDT) is a promising cancer treatment that induces localized tumor destruction via the photochemical generation of cytotoxic singlet oxygen. PDT-mediated oxidative stress elicits direct tumor cell damage as well as microvascular injury within exposed tumors. Reduction in vascular perfusion associated with PDT-mediated microvascular injury produces tumor tissue hypoxia. Using a transplantable BA mouse mammary carcinoma, we show that Photofrin-mediated PDT induced expression of the hypoxia-inducible factor-1alpha (HIF-1alpha) subunit of the heterodimeric HIF-1 transcription factor and also increased protein levels of the HIF-1 target gene, vascular endothelial growth factor (VEGF), within treated tumors. HIF-1alpha and VEGF expression were also observed following tumor clamping, which was used as a positive control for inducing tissue hypoxia. PDT treatment of BA tumor cells grown in culture resulted in a small increase in VEGF expression above basal levels, indicating that PDT-mediated hypoxia and oxidative stress could both be involved in the overexpression of VEGF. Tumor-bearing mice treated with combined antiangiogenic therapy (IM862 or EMAP-II) and PDT had improved tumoricidal responses compared with individual treatments. We also demonstrated that PDT-induced VEGF expression in tumors decreased when either IM862 or EMAP-II was included in the PDT treatment protocol. Our results indicate that combination procedures using antiangiogenic treatments can improve the therapeutic effectiveness of PDT.


Subject(s)
Cytokines , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Photochemotherapy/methods , Transcription Factors , Angiogenesis Inhibitors/pharmacology , Animals , Cell Hypoxia , Combined Modality Therapy , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Dihematoporphyrin Ether/pharmacology , Dipeptides/pharmacology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Female , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphokines/biosynthesis , Lymphokines/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred C3H , Neoplasm Proteins/pharmacology , Neoplasm Transplantation , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Photosensitizing Agents/pharmacology , RNA-Binding Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
3.
Cancer Res ; 60(6): 1637-44, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10749134

ABSTRACT

Oxidative stress associated with photodynamic therapy (PDT) is a transcriptional inducer of genes encoding stress proteins, including those belonging to the heat shock protein (hsp) family. The efficiency of PDT to function as a molecular switch by initiating expression of heterologous genes ligated to the human hsp promoter was examined in the present study. Selective and temporal reporter gene expression was documented after PDT in mouse radiation-induced fibrosarcoma cells stably transfected with recombinant vectors containing an hsp promoter ligated to either the lac-z or CAT reporter genes and in transfected radiation-induced fibrosarcoma tumors grown in C3H mice. Hyperthermia treatments were included as a positive control for all experiments. Expression vectors containing either human p53 or tumor necrosis factor (TNF)-alpha cDNA under the control of an hsp promoter were also constructed and evaluated. A p53 null and TNF-alpha-resistant human ovarian carcinoma (SKOV-3) cell line was stably transfected with either the p53 or TNF-alpha constructs. Inducible expression and function of p53 as well as inducible expression, secretion, and biological activity of TNF-alpha were documented after PDT or hyperthermia in transfected SKOV cells. These results demonstrate that PDT-mediated oxidative stress can function as a molecular switch for the selective and temporal expression of heterologous genes in tumor cells containing expression vectors under the control of an hsp promoter.


Subject(s)
Heat-Shock Proteins/genetics , Oxidative Stress/physiology , Photochemotherapy , Photosensitizing Agents/therapeutic use , Promoter Regions, Genetic/genetics , Animals , Cell Survival/drug effects , Chloramphenicol O-Acetyltransferase/drug effects , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Dihematoporphyrin Ether/therapeutic use , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/metabolism , Humans , Hyperthermia, Induced , Mice , Mice, Inbred C3H , Oxidative Stress/drug effects , Porphyrins/therapeutic use , Protein Binding/drug effects , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements , Temperature , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/drug effects , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
4.
Cancer Res ; 59(2): 331-5, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9927042

ABSTRACT

Photodynamic therapy (PDT) is an effective local cancer treatment that induces cytotoxicity through the intracellular generation of reactive oxygen species. The current study investigated whether abrogation of wild-type p53 expression modified the sensitivity of tumor cells to PDT-mediated oxidative stress. In these experiments, human colon (LS513) and breast (MCF-7) carcinoma cells exhibiting a wild-type p53 phenotype were directly compared to LS513 and MCF-7 cells with abrogated p53 function induced by stable integration of the human papillomavirus type 16 E6 viral oncoprotein. The effectiveness of this viral oncoprotein to target p53 for degradation was confirmed using a p53 transactivation reporter gene assay. Western analysis also confirmed attenuated expression of p53 in E6-transfected cells. Photosensitivity of PDT-treated cells was measured by a clonogenic assay and found to be equivalent for parental and p53-abrogated cells. PDT-mediated oxidative stress resulted in a rapid shift of pRb from a hyperphosphorylated form to a predominantly underphosphorylated form in parental cells that was not preceded by increases in p53 or p21 expression. Hypophosphorylated pRb was also observed in PDT-treated LS513/E6 and MCF-7/E6 cells, further indicating that p53 was not involved in this process. Delayed expression of p53 and p21 proteins was seen in parental cells 24-48 h after photosensitization. Cell cycle analysis showed that the abrogation of p53 had minimal effects on an observed PDT-induced G1 block. Rapid induction of apoptosis was documented in PDT-treated LS513 cells, whereas LS513/E6 treated cells exhibited reduced apoptosis in response to PDT. The MCF-7 cell lines exhibited a minimal apoptotic response to PDT. These results indicate that p53 expression does not directly modulate tumor cell sensitivity to PDT in either apoptosis-responsive (LS513) or nonresponsive (MCF-7) cells.


Subject(s)
Neoplasms/drug therapy , Photochemotherapy , Repressor Proteins , Tumor Suppressor Protein p53/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/physiology , Humans , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins, Viral/physiology , Oxidative Stress , Phosphorylation , Retinoblastoma Protein/metabolism , Tumor Cells, Cultured
5.
J Natl Cancer Inst ; 90(12): 889-905, 1998 Jun 17.
Article in English | MEDLINE | ID: mdl-9637138

ABSTRACT

Photodynamic therapy involves administration of a tumor-localizing photosensitizing agent, which may require metabolic synthesis (i.e., a prodrug), followed by activation of the agent by light of a specific wavelength. This therapy results in a sequence of photochemical and photobiologic processes that cause irreversible photodamage to tumor tissues. Results from preclinical and clinical studies conducted worldwide over a 25-year period have established photodynamic therapy as a useful treatment approach for some cancers. Since 1993, regulatory approval for photodynamic therapy involving use of a partially purified, commercially available hematoporphyrin derivative compound (Photofrin) in patients with early and advanced stage cancer of the lung, digestive tract, and genitourinary tract has been obtained in Canada, The Netherlands, France, Germany, Japan, and the United States. We have attempted to conduct and present a comprehensive review of this rapidly expanding field. Mechanisms of subcellular and tumor localization of photosensitizing agents, as well as of molecular, cellular, and tumor responses associated with photodynamic therapy, are discussed. Technical issues regarding light dosimetry are also considered.


Subject(s)
Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Dihematoporphyrin Ether/therapeutic use , Humans , Inflammation , Neoplasms/immunology
6.
J Photochem Photobiol B ; 42(2): 104-7, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9540217

ABSTRACT

Tumor sensitivity to cancer therapies may be modulated by the p53 status of the malignant cells. Generally, tumors retaining wild-type p53 are more sensitive to radiotherapy and some chemotherapeutic agents than are tumors with either a mutated or deleted p53 phenotype. The role of p53 in the responsiveness to PDT as a cancer treatment is clinically unknown. In the current study, we evaluated the photosensitivity of two human colon carcinoma cell lines, one expressing wild-type p53 protein and the other expressing mutant p53. Wild-type p53 cells were found to be significantly more sensitive to Photofrin-mediated photodynamic treatment measured by clonogenic assay. Uptake of the photosensitizer was equivalent for both cell lines. Interestingly, sensitivity of the colon carcinoma cell lines to ionizing radiation was similar. These two cell lines represent a useful model for examining p53 involvement in the cellular response to PDT-mediated oxidative stress.


Subject(s)
Colonic Neoplasms/drug therapy , Photochemotherapy , Radiation Tolerance , Tumor Suppressor Protein p53/genetics , Colonic Neoplasms/genetics , Humans , Mutation , Tumor Cells, Cultured
7.
Photochem Photobiol ; 66(2): 265-70, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9277147

ABSTRACT

Loss of p53 function has been correlated with decreased sensitivity to chemotherapy and radiation therapy in a variety of human tumors. Comparable analysis of p53 status with sensitivity to oxidative stress induced by photodynamic therapy has not been reported. In the current study we examined photosensitivity in human promyelocytic leukemia HL60 cells exhibiting either wild-type p53, mutated p53 or deleted p53 expression. Experiments were performed using a purpurin, tin ethyl etiopurpurin (SnET2)-, or a porphyrin, Photofrin (PH)-based photosensitizer. Total SnET2 accumulation was comparable in all three cell lines. Uptake of PH was highest in cells expressing wild-type p53 but incubation conditions could be adjusted to achieve equivalent cellular PH levels during experiments that analyzed photosensitivity. Survival measurements demonstrated that HL60 cells expressing wild-type p53 were more sensitive to PH- and SnET2-mediated photosensitization, as well as to UVC irradiation, when compared to HL60 cells exhibiting deleted or mutated p53 phenotypes. A rapid apoptotic response was observed following purpurin- and porphyrin-induced photosensitization in all cell lines. Results of this study indicate that photosensitivity is increased in HL60 cells expressing wild-type p53 and that photosensitizer-mediated oxidative stress can induce apoptosis through a p53-independent mechanism in HL60 cells.


Subject(s)
Radiation Tolerance , Tumor Suppressor Protein p53/metabolism , Cell Survival , HL-60 Cells , Humans , Polymerase Chain Reaction , Porphyrins/metabolism , Radiation-Sensitizing Agents/metabolism
9.
Arch Ophthalmol ; 114(11): 1348-56, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8906025

ABSTRACT

OBJECTIVE: To describe platinum-based chemotherapy combined with local treatment modalities as an alternative to external beam radiotherapy for intraocular retinoblastoma. DESIGN: Platinum levels were measured by atomic absorption analysis in the tumors of 2 patients with retinoblastoma given carboplatin 5 or 2.5 hours before enucleation. Platinum levels in heated vs nonheated Greene melanoma tumors in rabbits were compared. A retrospective review of 172 affected eyes in 136 consecutive patients treated for retinoblastoma between January 1990 and December 1995 was performed. From 1990 to 1992, all treatable eyes initially received systemic carboplatin, 560 mg/m2, followed by 15 to 30 minutes of continuous diode laser hyperthermia (thermochemotherapy). Since 1992, larger tumors were treated initially with 3 monthly cycles of carboplatin, etoposide, and vincristine sulfate to reduce tumor volume (chemoreduction) followed by sequential aggressive local therapy (SALT) during examinations under anesthesia every 2 to 3 weeks. OUTCOME MEASURE: Treatment success was defined as eradication of tumor without enucleation or external beam radiotherapy. RESULTS: Significant therapeutic platinum levels were measured in the human tumors 2.5 and 5 hours after carboplatin administration. Increasing the temperature by 9 degrees C for 15 minutes doubled platinum levels in the rabbit model. Of the 38 eyes with Reese-Ellsworth group 1 through 5b tumors that were treated primarily with thermochemotherapy, all 24 eyes with group 1 and 2 tumors were treated successfully and two of the 4 eyes with group 3 tumors and all 10 eyes with group 5b tumors were treated unsuccessfully. Chemoreduction plus SALT was the primary treatment in 35 eyes and was successful in all 10 eyes with group 1 through 4 tumors and unsuccessful in all 7 eyes with extensive subretinal seeding and all 18 eyes with group 5b tumors with vitreous seeding. Seventy patients received carboplatin or carboplatin, vincristine, and etoposide, with myelosuppression, occasionally associated with bacteremia, being the main side effect. Transfusions were required in 15% of patients. Radiation retinopathy occurred in all 6 eyes treated with iodine 125 plaques. CONCLUSIONS: Thermochemotherapy is successful primary treatment for Reese-Ellsworth group 1 and 2 retinoblastomas. For larger tumors in the absence of vitreous or extensive subretinal seeding, 3 cycles of chemoreduction followed by SALT eradicates residual viable tumor. Chemoreduction plus SALT was not successful in eyes with diffuse vitreous or extensive subretinal seeding. Prior chemotherapy increases the risk for radiation retinopathy following 125I plaque therapy. External beam radiotherapy can safely be avoided in the primary treatment of Reese-Ellsworth groups 1 through 4 nondispersed retinoblastoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Carboplatin/therapeutic use , Eye Neoplasms/therapy , Hyperthermia, Induced , Retinoblastoma/therapy , Animals , Anterior Chamber/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brachytherapy , Carboplatin/analysis , Combined Modality Therapy , Cryotherapy , DNA Adducts/analysis , DNA, Neoplasm/analysis , Etoposide/administration & dosage , Eye Enucleation , Eye Neoplasms/chemistry , Humans , Iodine Radioisotopes/therapeutic use , Laser Coagulation , Melanoma/chemistry , Melanoma/therapy , Rabbits , Retinoblastoma/chemistry , Vincristine/administration & dosage
10.
J Clin Laser Med Surg ; 14(5): 315-21, 1996 Oct.
Article in English | MEDLINE | ID: mdl-9612198

ABSTRACT

The positive clinical results associated with photodynamic therapy (PDT) have led to an expanded need to identify the cellular targets and molecular responses associated with this treatment. Increased knowledge regarding the mechanisms of action associated with PDT-mediated cytotoxicity should contribute to the continued advancement of this therapy. This report focuses on recent studies analyzing PDT resistance and examining stress protein and early response gene activation induced by photosensitizer mediated oxidative stress. Recurring observations from these studies indicate that subcellular targets and cellular responses associated with PDT can vary significantly for different photosensitizers.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/physiology , Fibrosarcoma/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , Heat-Shock Response/drug effects , Photosensitizing Agents/pharmacology , Animals , Cell Division/drug effects , Dihematoporphyrin Ether/pharmacology , Female , Gene Expression Regulation/drug effects , Heat-Shock Response/physiology , Mice , Mice, Inbred C3H , Oxidative Stress/genetics , Oxidative Stress/physiology , Porphyrins/pharmacology , Proto-Oncogenes/physiology , Transcriptional Activation , Tumor Cells, Cultured/drug effects
11.
Cancer Res ; 56(10): 2355-60, 1996 May 15.
Article in English | MEDLINE | ID: mdl-8625311

ABSTRACT

Photodynamic therapy (PDT) is an experimental cancer therapy inducing tumor tissue damage via photosensitizer-mediated oxidative cytotoxicity. A previous report indicates that oxidative stress induced by hydrogen peroxide or menadione activates the heat shock transcription factor in mouse cells but does not result in either increased transcription or translation of heat shock proteins (HSPs). Our study documents that photosensitizer-mediated oxidative stress can activate the heat shock factor as well as increase HSP-70 mRNA and protein levels in mouse RIF-1 cells. The cellular heat shock response after PDT varied for the different photosensitizers being examined. Treatments using either a chlorin (mono-L-aspartyl chlorin-e6)- or purpurin (tin etio-purpurin)-based sensitizer induced HSP-70 expression, whereas identical photosensitization conditions with a porphyrin (Photofrin)-based sensitizer failed to induce a cellular HSP response. These sensitizers, which generate singlet oxygen as the primary oxidant during photosensitization, were used in experiments under isoeffective treatment conditions. HSP-70 expression after photosensitization was associated with the concomitant induction of thermotolerance in PDT-treated cells. Interestingly, reverse transcription-PCR demonstrated that in vivo PDT treatments of RIF-1 tumors induce expression of HSP-70 for all photosensitizers including Photofrin. These results indicate that photosensitizer-generated singlet oxygen exposure can induce in vitro and in vivo HSP-70 expression, and that specific subcellular targets of PDT (which can differ for various sensitizers) are determinants for HSP-70 activation after oxidative stress.


Subject(s)
Anthraquinones , Fibrosarcoma/drug therapy , Heat-Shock Proteins/biosynthesis , Hematoporphyrin Derivative/pharmacology , Lectins/pharmacology , Oxidative Stress/genetics , Photochemotherapy/adverse effects , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Animals , Base Sequence , Female , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , Heat-Shock Proteins/genetics , Mice , Mice, Inbred C3H , Molecular Sequence Data , Oxygen/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Singlet Oxygen , Tumor Cells, Cultured
12.
Cancer Res ; 55(9): 1820-3, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7728745

ABSTRACT

Parental and photodynamic therapy (PDT)-resistant mouse, radiation-induced fibrosarcoma cell lines were evaluated using mRNA differential display in an attempt to identify unique transcripts. We detected one transcript that was consistently present in the parental cells but absent in PDT-resistant cells. The transcript was cloned, sequenced, and identified as alpha-2 macroglobulin receptor/low density lipoprotein receptor-related protein (alpha-2 MR/LRP). Northern and Western immunoblot analysis confirmed that receptor expression was present in the parental cell line but barely detectable in PDT-resistant cells. Functionality of the receptor was evaluated by exposing cells to Pseudomonas exotoxin A. alpha-2 MR/LRP is responsible for Pseudomonas exotoxin A internalization, and only the parental cells exhibited toxin-mediated cytotoxicity. The binding and endocytosis of activated alpha-2 macroglobulin and lipoproteins by alpha-2 MR/LRP are consistent with modulating uptake and localization of photosensitizers. Our results demonstrate that PDT-resistant murine tumor cells exhibit minimal alpha-2 MR/LRP activity and suggest that this receptor plays a role in PDT sensitivity by modulating photosensitizer uptake and/or subcellular localization.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Fibrosarcoma/drug therapy , Fibrosarcoma/ultrastructure , Photochemotherapy , Receptors, Immunologic/drug effects , Receptors, Immunologic/physiology , Receptors, LDL/drug effects , Receptors, LDL/physiology , Virulence Factors , Animals , Base Sequence , Blotting, Western , Cell Survival/drug effects , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Dihematoporphyrin Ether/pharmacokinetics , Dihematoporphyrin Ether/pharmacology , Drug Resistance , Exotoxins/metabolism , Exotoxins/pharmacology , Fibrosarcoma/metabolism , Hematoporphyrin Derivative/pharmacokinetics , Hematoporphyrin Derivative/pharmacology , Hematoporphyrin Photoradiation , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Molecular Sequence Data , RNA, Messenger/analysis , Radiation-Sensitizing Agents/pharmacokinetics , Radiation-Sensitizing Agents/pharmacology , Receptors, Immunologic/metabolism , Receptors, LDL/metabolism , Tumor Cells, Cultured , Pseudomonas aeruginosa Exotoxin A
13.
J Photochem Photobiol B ; 28(1): 13-8, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7791001

ABSTRACT

Biophysical and photobiological properties of three derivatives of chlorin p6 were examined. These agents can be considered as lysyl analogs of the aspartyl chlorin NPe6. Lysyl chlorin p6 diester (LCP) and the triester analog (LCP2) were readily accumulated by murine leukemia L1210 cells, localized in lysosomes, and were relatively inefficient photosensitizing agents in vitro. In contrast, lysyl chlorin e6 imide (LCI) was poorly accumulated, concentrated in mitochondrial and plasma membranes, but was more efficacious. LCI was the most effective agent with regard to photosensitization of a murine tumor in vivo, but all three agents caused substantially more toxicity than was observed with NPe6.


Subject(s)
Leukemia L1210/metabolism , Mammary Neoplasms, Experimental/drug therapy , Photosensitizing Agents/toxicity , Porphyrins/toxicity , Animals , Cell Division/drug effects , Cell Division/radiation effects , Dose-Response Relationship, Radiation , Fluorescent Dyes , Light , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C3H , Microscopy, Fluorescence , Molecular Structure , Photosensitizing Agents/pharmacokinetics , Porphyrins/pharmacokinetics , Porphyrins/therapeutic use , Structure-Activity Relationship , Time Factors , Tissue Distribution , Tumor Cells, Cultured
14.
Lasers Surg Med ; 17(1): 2-31, 1995.
Article in English | MEDLINE | ID: mdl-7564853

ABSTRACT

Photodynamic therapy (PDT) is a treatment modality that utilizes a photosensitizing drug activated by laser generated light, and is proving effective for oncologic and nononcologic applications. This report provides an overview of photosensitizers, photochemistry, photobiology, and the lasers involved in photodynamic therapy. Clinical and preclinical PDT studies involving Photofrin and various second generation photosensitizers are reviewed.


Subject(s)
Photochemotherapy , Animals , Clinical Trials as Topic , Hematoporphyrin Derivative/therapeutic use , Humans , Laser Therapy , Neoplasms/drug therapy , Photobiology , Photochemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/classification , Photosensitizing Agents/therapeutic use , Virus Diseases/drug therapy
15.
Cancer Res ; 54(5): 1374-80, 1994 Mar 01.
Article in English | MEDLINE | ID: mdl-8118827

ABSTRACT

Photodynamic therapy (PDT) generates reactive oxygen species which initiate the cytotoxic events of this tumor treatment. We demonstrate that PDT mediated oxidative stress induced a transient increase in the early response genes c-fos, c-jun, c-myc, and egr-1 in murine radiation-induced fibrosarcoma cells. Incubation of exponentially growing cells with porphyrin based photosensitizers in the dark also induced an increase in mRNA levels of early response genes. However, the xanthine photosensitizer, rose bengal, produced increased c-fos mRNA levels only following light treatment. Nuclear runoff experiments confirmed that the induction of c-fos mRNA is controlled in part at the level of transcription. Likewise, a chloramphenicol acetyltransferase reporter construct containing the major c-fos transcriptional response elements was inducible by porphyrin and PDT. Signal transduction pathways associated with PDT mediated c-fos activation were examined by treating cells with protein kinase inhibitors. Staurosporine and 1-(5-isoquinolinesulfonyl)-2-methylpiperazine inhibited PDT mediated c-fos activation while N-(2-guanidinoethyl)-5-isoquinoline-sulfonamide had no effect. In addition, quinacrine, which can inhibit phospholipase activity, blocked PDT induced c-fos mRNA expression. These results suggest that photosensitizer mediated oxidative stress acts through protein kinase-mediated signal transduction pathway(s) to activate early response genes.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Genes, Immediate-Early/drug effects , Genes, Immediate-Early/genetics , Photochemotherapy , Animals , Drug Interactions , Fibrosarcoma/drug therapy , Fibrosarcoma/genetics , Gene Expression Regulation, Neoplastic/physiology , Genes, fos/genetics , Mice , Phospholipases/antagonists & inhibitors , Photosensitizing Agents/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/toxicity , Stress, Physiological/chemically induced , Stress, Physiological/physiopathology , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
16.
Photochem Photobiol ; 58(5): 753-6, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8284329

ABSTRACT

Clinical photodynamic therapy (PDT) uses the photosensitizer photofrin II to produce singlet molecular oxygen and other reactive oxygen intermediates for localized tumor tissue cytotoxicity. In this report, we show that PDT enhances the DNA binding activity of nuclear factor kappa B (NF kappa B), a transactivator of cytokine gene expression. Photosensitization following a 16 h incubation of photofrin II induced NF kappa B binding activity in mouse leukemia L1210 cells 10-fold above that observed in exponentially growing cultures. Serum starvation, as well as drug-alone and light-alone controls, elevated basal NF kappa B binding activity two- to three-fold. Upstream stimulatory factor binding activity was not modulated by any of the cell treatments and was used to standardize gel mobility shift data. This study identifies porphyrin-mediated PDT as an inducer of NF kappa B binding activity, extending recent findings that NF kappa B activation is a general response to oxidative stress.


Subject(s)
DNA, Neoplasm/metabolism , Dihematoporphyrin Ether/pharmacology , Leukemia L1210/metabolism , NF-kappa B/metabolism , Animals , Base Sequence , DNA-Binding Proteins/metabolism , Light , Mice , Molecular Sequence Data , Protein Binding , Transcription Factors/metabolism , Tumor Cells, Cultured , Upstream Stimulatory Factors
17.
Photochem Photobiol ; 58(4): 581-8, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8248335

ABSTRACT

Photodynamic therapy (PDT) generates reactive oxygen species that are responsible for the initial cytotoxic events produced by this treatment. An extended (16 h) porphyrin incubation prior to light irradiation increased expression of the 75, 78 and 94 kDa glucose-regulated stress proteins (GRP), as well as the cognate form of the 70 kDa heat shock protein. However, these stress proteins were not induced following isoeffective PDT doses using a short (1 h) porphyrin incubation protocol. In the current study, Chinese hamster fibroblasts were used to examine sensitivity to adjunctive PDT and adriamycin as previous reports indicate a correlation between stress protein synthesis and a decrease in adriamycin cytotoxicity. Treatments that either induced GRP (i.e. PDT with an extended porphyrin incubation or exposure to the calcium ionophore A23187) or did not induce GRP (i.e. PDT with a short porphyrin incubation or UV irradiation) were followed at increasing time intervals with a 1 h adriamycin incubation. A time-dependent decrease in adriamycin cytotoxicity was observed when cells were first exposed to either of the PDT protocols or to A23187. Alterations in intracellular drug levels did not account for the change in adriamycin sensitivity. Likewise, intracellular glutathione concentrations and antioxidant enzyme activities were not significantly altered following PDT or A23187. Parameters associated with altered adriamycin sensitivity included a decrease in the percentage of S phase cells following PDT and A23187 as well as a depletion of intracellular ATP after PDT using the extended porphyrin incubation. These results demonstrate that PDT can be added to the growing list of diverse stresses producing transient resistance to adriamycin and that stress protein induction is not universally associated with all oxidative treatments inducing this resistance.


Subject(s)
Dihematoporphyrin Ether/toxicity , Doxorubicin/toxicity , HSP70 Heat-Shock Proteins , Heat-Shock Proteins/biosynthesis , Membrane Proteins/biosynthesis , Photochemotherapy , Animals , Calcimycin/pharmacology , Cell Line , Cricetinae , Cricetulus , Drug Resistance , Fibroblasts/drug effects , Fibroblasts/metabolism , Kinetics , Light , Lung , Time Factors , Ultraviolet Rays
18.
J Cell Physiol ; 153(3): 575-82, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1332981

ABSTRACT

GRP78, a 78,000 dalton protein residing in the endoplasmic reticulum, is postulated to play important roles in protein folding and cell survival during calcium and other physiological stress. Here we describe the construction of an eukaryotic expression vector for the constitutive expression of grp78 antisense RNA and the creation of a CHO cell line, 78WO, which expresses high levels of the grp78 antisense RNA through amplification of the stably transfected antisense vector. We observed that whereas 78WO maintains a basal level of GRP78 similar to that of control cells, GRP78 is no longer inducible by A23187. The 78WO cells have undergone a compensatory increase in grp78 transcription such that the effects of antisense are cancelled out at the protein level under nonstressed conditions. In these same cells, GRP94, a 94,00 dalton ER protein, is also rendered noninducible by A23187. This provides the first evidence that the regulation of two ER proteins might be coupled such that the failure to induce GRP78 results in the down-regulation of GRP94. The 78WO cell line grows with a doubling time of about 26 hr and exhibits decreased tolerance to A23187, suggesting the GRPs contribute to cell viability under calcium stress. The establishment of this cell line, which can be stably maintained, will provide a useful tool for testing whether the induction of the GRPs is important for protein folding or transport and whether their enhanced synthesis is the cause or consequence of a variety of physiological adaptations.


Subject(s)
CHO Cells/metabolism , Calcimycin/pharmacology , Carrier Proteins/genetics , HSP70 Heat-Shock Proteins , Heat-Shock Proteins , Membrane Proteins/antagonists & inhibitors , Molecular Chaperones , RNA, Antisense/metabolism , Animals , Avian Sarcoma Viruses/genetics , CHO Cells/cytology , Calcimycin/antagonists & inhibitors , Carrier Proteins/antagonists & inhibitors , Cell Division , Cell Line, Transformed , Cricetinae , Endoplasmic Reticulum Chaperone BiP , Protein Biosynthesis , Transfection
19.
Cancer Res ; 52(10): 2890-3, 1992 May 15.
Article in English | MEDLINE | ID: mdl-1581904

ABSTRACT

A mouse mammary tumor model was used to evaluate metabolic properties of the photosensitizer mono-L-aspartyl chlorin e6 (NPe6) and to determine the optimal time interval between drug administration and light treatment for effective photodynamic therapy (PDT). Photosensitizer metabolism was evaluated by comparing tissue distribution patterns of NPe6 having 14C atoms positioned on either the tetrapyrrole ring or on the aspartyl residue. High performance liquid chromatographic analysis of photosensitizer extracted from tumor tissue was also obtained as a function of time after drug administration. NPe6 distribution in tissue samples and pharmacological calculations of area under the curve were similar for both forms of [14]NPe6. Likewise, metabolic contaminants of NPe6 were not detected by high performance liquid chromatographic analysis following extraction of the photosensitizer from tumor tissue. Maximal in vivo PDT effectiveness was achieved when light treatments were started within 2 h of drug injection. PDT effectiveness was decreased by 50% when light treatments were initiated 6 h after drug injection and was abolished with a 12-h interval between NPe6 injection and light exposure. Responsiveness to NPe6-mediated PDT was correlated with photosensitizer levels in the plasma but not in tumor tissue. These results show that NPe6 was not metabolized following in vivo administration and that the responsiveness of NPe6 mediated PDT was associated with vascular clearance of the photosensitizer.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Photochemotherapy , Porphyrins/pharmacokinetics , Animals , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Porphyrins/metabolism , Radiation-Sensitizing Agents/metabolism , Radiation-Sensitizing Agents/pharmacokinetics , Time Factors , Tissue Distribution
20.
Photochem Photobiol ; 54(6): 1093-107, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1775531

ABSTRACT

Numerous photosensitizers with absorption peaks spanning the 600-800 nm "therapeutic window" have been and continue to be synthesized. Structural modifications of the dyes can then be made in order to improve tumor deliverability and retention. Chemical alterations can also enhance the yields of light generated reactive oxygen species. Utilization of lipoproteins, emulsions and antibody conjugates can enhance the selectivity of drug localization. Most cell types and subcellular structures are highly photosensitive and biochemical analysis indicates that cellular target sites associated with PDT correlate with photosensitizer location. In vivo data suggest that vascular and direct tumor cell damage as well as systemic and local immunological reactions are involved in PDT responsiveness. Additional mechanistic, synthetic and developmental studies are required in order to fully appreciate the potentials of PDT. However, continued enthusiasm and support for basic PDT research (as observed during the past 8 years) will depend to a large extent on the outcome of the current clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Photochemotherapy , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Drug Design , Drug Evaluation, Preclinical , Humans , Neoplasms, Experimental/drug therapy , Radiation-Sensitizing Agents/therapeutic use , Radiation-Sensitizing Agents/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...