Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Biomedicines ; 11(2)2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36830797

ABSTRACT

Any gene therapy for cancer will be predicated upon its selectivity against cancer cells and non-toxicity to normal cells. Therefore, safeguards are needed to prevent its activation in normal cells. We designed a minimal p14ARF promoter with upstream Ap1 and E2F enhancer elements and a downstream MDR1 inhibitory element, TATA box, and a transcription initiation site (hereafter p14ARFmin). The modified p14ARFmin promoter was linked to bicistronic P14 and truncated BID (tBID) genes, which led to synergistic apoptosis via the intrinsic and extrinsic pathways of apoptosis when expressed. The promoter was designed to be preferentially activated by mutant Ras and completely inhibited by wild-type p53 so that only cells with both mutant Ras and mutant p53 would activate the construct. In comparison to most p53 gene therapies, this construct has selective advantages: (1) p53-based gene therapies with a constitutive CMV promoter cannot differentiate between normal cells and cancer cells, and can be toxic to normal cells; (2) our construct does not induce p21WAF/CIPI in contrast to other p53-based gene therapies, which can induce cell cycle arrest leading to increased chemotherapy resistance; (3) the modified construct (p14ARFmin-p14-tBID) demonstrates bidirectional control of its promoter, which is completely repressed by wild-type p53 and activated only in cells with both RAS and P53 mutations; and (4) a novel combination of genes (p14 and tBID) can synergistically induce potent intrinsic and extrinsic apoptosis in cancer cells.

2.
Mol Cancer Ther ; 12(12): 2792-803, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24126434

ABSTRACT

Pancreatic cancer cell lines with mutated ras underwent an alternative form of cell death (aponecrosis) when treated concomitantly with clinically achievable concentrations of arsenic trioxide, ascorbic acid, and disulfiram (Antabuse; AAA). AAA's major effects are mediated through generation of intracellular reactive oxygen species (ROS) and more than 50% decline in intracellular ATP. N-acetyl cysteine and a superoxide dismutase mimetic prevented aponecrosis and restored intracellular ATP levels. DIDS (4,4'-diisothiocyanatostilbene-2, 2' disulfonic acid), the pan- Voltage-Dependent Anion Channel (VDAC), -1, 2, 3 inhibitor and short hairpin RNA (shRNA) to VDAC-1 blocked cell death and ROS accumulation. In vivo exposure of AAA led to a 62% reduction in mean tumor size and eliminated tumors in 30% of nude mice with PANC-1 xenografts. We concluded that early caspase-independent apoptosis was shifted to VDAC-mediated "targeted" aponecrosis by the addition of disulfiram to arsenic trioxide and ascorbic acid. Conceptually, this work represents a paradigm shift where switching from apoptosis to aponecrosis death pathways, also known as targeted aponecrosis, could be utilized to selectively kill pancreatic cancer cells resistant to apoptosis.


Subject(s)
Adenosine Triphosphate/metabolism , Apoptosis , Necrosis/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Voltage-Dependent Anion Channels/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenic Trioxide , Arsenicals/administration & dosage , Arsenicals/pharmacology , Ascorbic Acid/administration & dosage , Ascorbic Acid/pharmacology , Cell Line, Tumor , Disease Models, Animal , Disulfiram/administration & dosage , Disulfiram/pharmacology , Dose-Response Relationship, Drug , Heterografts , Humans , Male , Mice , Oxides/administration & dosage , Oxides/pharmacology , Pancreatic Neoplasms/drug therapy , Tumor Stem Cell Assay
3.
Cancer Chemother Pharmacol ; 71(3): 663-70, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23370660

ABSTRACT

PURPOSE: We evaluated the efficacy and safety of capecitabine and temozolomide (CAPTEM) in patients with metastatic neuroendocrine tumors (NETs) to the liver. This regimen was based on our studies with carcinoid cell lines that showed synergistic cytotoxicity with sequence-specific dosing of 5-fluorouracil preceding temozolomide (TMZ). METHODS: A retrospective review was conducted of 18 patients with NETs metastatic to the liver who had failed 60 mg/month of Sandostatin LAR™ (100%), chemotherapy (61%), and hepatic chemoembolization (50%). Patients received capecitabine at 600 mg/m(2) orally twice daily on days 1-14 (maximum 1,000 mg orally twice daily) and TMZ 150-200 mg/m(2) divided into two doses daily on days 10-14 of a 28-day cycle. Imaging was performed every 2 cycles, and serum tumor markers were measured every cycle. RESULTS: Using RECIST parameters, 1 patient (5.5%) with midgut carcinoid achieved a surgically proven complete pathological response (CR), 10 patients (55.5%) achieved a partial response (PR), and 4 patients (22.2%) had stable disease (SD). Total response rate was 61%, and clinical benefit (responders and SD) was 83.2%. Of four carcinoid cases treated with CAPTEM, there was 1 CR, 1 PR, 1 SD, and 1 progressive disease. Median progression-free survival was 14.0 months (11.3-18.0 months). Median overall survival from diagnosis of liver metastases was 83 months (28-140 months). The only grade 3 toxicity was thrombocytopenia (11%). There were no grade 4 toxicities, hospitalizations, opportunistic infections, febrile neutropenias, or deaths. CONCLUSIONS: CAPTEM is highly active, well tolerated and may prolong survival in patients with well-differentiated, metastatic NET who have progressed on previous therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroendocrine Tumors/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Male , Middle Aged , Neuroendocrine Tumors/pathology , Retrospective Studies , Survival Analysis , Temozolomide , Treatment Outcome
4.
Methods Mol Biol ; 980: 321-9, 2013.
Article in English | MEDLINE | ID: mdl-23359163

ABSTRACT

The use of fluorescent probes can be an easy and quick method to analyze whether or not reactive oxygen species (ROS) are involved in a particular cellular process or the result of a particular drug treatment. ROS activate a variety of cell signaling and death pathways including apoptosis and necrosis (Raha and Robinson. Am J Med Gen 106:62-70, 2001). Here we describe a number of different probes, their specificities, advantages and limitations, as well as useful techniques important for their use.


Subject(s)
Fluorescent Dyes , Pancreatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Apoptosis , Cell Line, Tumor , Flow Cytometry , Humans , Necrosis , Staining and Labeling
5.
J Biol Chem ; 282(37): 26675-26686, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17636258

ABSTRACT

Cancer cells escape apoptosis by intrinsic or acquired mechanisms of drug resistance. An alternative strategy to circumvent resistance to apoptosis could be through redirection into other death pathways, such as necrosis. However, necrosis is a nonspecific, nontargeted process resulting in cell lysis and inflammation of both cancer and normal cells and is therefore not a viable alternative. Here, we report that a C-terminal peptide of p53, called p53p-Ant, induced targeted necrosis only in multiple mutant p53 human prostate cancer lines and not normal cells, because the mechanism of cytotoxicity by p53p-Ant is dependent on the presence of high levels of mutant p53. Topotecan- and paclitaxel-resistant prostate cancer lines were as sensitive to p53p-Ant-induced targeted necrosis as parental lines. A massive loss of ATP pools and intracellular generation of reactive oxygen species was involved in the mechanism of targeted necrosis, which was inhibited by O(2)(.) scavengers. We hypothesize that targeted necrosis by p53p-Ant is dependent on mutant p53, is mediated by O(2)(.) loss and ATP, and can circumvent chemotherapy resistance to apoptosis. Targeted necrosis, as an alternative pathway for selective killing of cancer cells, may overcome the problems of nonspecificity in utilizing the necrotic pathway.


Subject(s)
Apoptosis/drug effects , Peptide Fragments/pharmacology , Prostatic Neoplasms/pathology , Tumor Suppressor Protein p53/pharmacology , Caspases/physiology , Cell Line, Tumor , Cell Survival , Drug Resistance, Neoplasm , Humans , Male , Mutation , Necrosis , Reactive Oxygen Species , fas Receptor/physiology
6.
Int J Cancer ; 115(1): 55-64, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15645452

ABSTRACT

A p53 C-terminal peptide (aa 361-382, p53p), fused at its C-terminus to the minimal carrier peptide of antennapedia (17 aa, Ant; p53p-Ant), induced rapid apoptosis in human cancer cells, via activation of the Fas pathway. We examined p53p-Ant mechanism of action, toxicity in various human normal, non-malignant, pre-malignant and malignant cancer cells and investigated its biophysical characteristics. p53p-Ant selectively induced cell death in only pre-malignant or malignant cells in a p53-dependent manner and was not toxic to normal and non-malignant cells. p53p-Ant was more toxic to the mutant p53 than wild-type p53 phenotype in H1299 lung cancer cells stably expressing human temperature-sensitive p53 mutant 143Ala. Surface plasmon resonance (BIACORE) analysis demonstrated that this peptide had higher binding affinity to mutant p53 as compared to wild-type p53. p53p-Ant induced-cell death had the classical morphological characteristics of apoptosis and had no features of necrosis. The mechanism of cell death by p53p-Ant was through the FADD/caspase-8-dependent pathway without the involvement of the TRAIL pathway, Bcl-2 family and cell cycle changes. Blocking Fas with antibody did not alter the peptide's effect, suggesting that Fas itself did not interact with the peptide. Transfection with a dominant-negative FADD with a deleted N-terminus inhibited p53p-Ant-induced apoptosis. Its mechanism of action is related to the FADD-induced pathway without restoration of other p53 functions. p53p-Ant is a novel anticancer agent with unique selectivity for human cancer cells and could be useful as a prototype for the development of new anti-cancer agents.


Subject(s)
Apoptosis , Arabidopsis Proteins/metabolism , Caspases/metabolism , Fatty Acid Desaturases/metabolism , Tumor Suppressor Protein p53/chemistry , Apoptosis Regulatory Proteins , Blotting, Western , Caspase 8 , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flow Cytometry/methods , Genes, Dominant , Humans , L-Lactate Dehydrogenase/metabolism , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Mutation , Necrosis , Peptides/chemistry , Phenotype , Plasmids/metabolism , Precancerous Conditions , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Surface Plasmon Resonance , TNF-Related Apoptosis-Inducing Ligand , Temperature , Time Factors , Transfection , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL