Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Oncogene ; 36(6): 820-828, 2017 02 09.
Article in English | MEDLINE | ID: mdl-27425591

ABSTRACT

Patients with human papillomavirus (HPV)-positive head and neck squamous cell carcinoma (HNSCC) have better responses to radiotherapy and higher overall survival rates than do patients with HPV-negative HNSCC, but the mechanisms underlying this phenomenon are unknown. p16 is used as a surrogate marker for HPV infection. Our goal was to examine the role of p16 in HPV-related favorable treatment outcomes and to investigate the mechanisms by which p16 may regulate radiosensitivity. HNSCC cells and xenografts (HPV/p16-positive and -negative) were used. p16-overexpressing and small hairpin RNA-knockdown cells were generated, and the effect of p16 on radiosensitivity was determined by clonogenic cell survival and tumor growth delay assays. DNA double-strand breaks (DSBs) were assessed by immunofluorescence analysis of 53BP1 foci; DSB levels were determined by neutral comet assay; western blotting was used to evaluate protein changes; changes in protein half-life were tested with a cycloheximide assay; gene expression was examined by real-time polymerase chain reaction; and data from The Cancer Genome Atlas HNSCC project were analyzed. p16 overexpression led to downregulation of TRIP12, which in turn led to increased RNF168 levels, repressed DNA damage repair (DDR), increased 53BP1 foci and enhanced radioresponsiveness. Inhibition of TRIP12 expression further led to radiosensitization, and overexpression of TRIP12 was associated with poor survival in patients with HPV-positive HNSCC. These findings reveal that p16 participates in radiosensitization through influencing DDR and support the rationale of blocking TRIP12 to improve radiotherapy outcomes.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/virology , Carrier Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Head and Neck Neoplasms/radiotherapy , Head and Neck Neoplasms/virology , Papillomaviridae/physiology , Papillomavirus Infections/radiotherapy , Ubiquitin-Protein Ligases/metabolism , Animals , Biomarkers, Tumor , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Mice , Papillomaviridae/genetics , Papillomavirus Infections/metabolism , Radiation Tolerance , Random Allocation , Squamous Cell Carcinoma of Head and Neck , Transfection , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
2.
Cancer Gene Ther ; 14(6): 543-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17431403

ABSTRACT

Expression of the PTEN tumor suppressor gene is abnormal in many human cancers. Loss of PTEN expression leads to the activation of downstream signaling pathways that have been associated with resistance to radiation. In non-small cell lung carcinoma (NSCLC), suppressed expression of PTEN is frequently due to methylation of its promoter region. In this study, we tested whether gene transfer of wild-type PTEN into an NSCLC cell line with a known methylated PTEN promoter, H1299, would increase its sensitivity to ionizing radiation. Pretreating H1299 cells with an adenoviral-mediated PTEN (Ad-PTEN)-expressing vector sensitized H1299 cells to radiation. To determine the mechanism responsible for radiosensitization, we first examined radiation-induced apoptosis, which was enhanced but did not correlate with radiosensitizing effect of Ad-PTEN. Therefore, we next examined the ability of Ad-PTEN to modulate the repair of radiation-induced DNA double-strand breaks (DSBs) using the detection of repair foci positive for gamma-H2AX, a protein that becomes evident at the sites of each DSB and that can be visualized by immunofluorescent staining. Compared with controls, the repair of radiation-induced DSBs was retarded in H1299 cells pretreated with Ad-PTEN, consistent with the radiosensitizing effect of the vector. We conclude that signal transduction pathways residing primarily in the cytoplasm may intersect with DNA damage and repair pathways in the nucleus to modulate cellular responses to radiation. Elucidating the mechanisms responsible for this intersection may lead to novel strategies for improving therapy for cancers with defective PTEN.


Subject(s)
Adenoviridae/physiology , Carcinoma, Non-Small-Cell Lung/pathology , DNA Repair/radiation effects , Lung Neoplasms/radiotherapy , PTEN Phosphohydrolase/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Cell Line , Gene Expression , Gene Expression Regulation, Neoplastic , Genetic Vectors , Humans , Lung Neoplasms/therapy , PTEN Phosphohydrolase/genetics , Radiation Tolerance , Radiation-Sensitizing Agents
3.
Cancer Gene Ther ; 13(5): 490-502, 2006 May.
Article in English | MEDLINE | ID: mdl-16282987

ABSTRACT

Current therapies used in the treatment of breast cancer are limited by systemic toxicity, rapid drug metabolism and intrinsic and acquired drug resistance. We have previously shown that adenoviral-mediated transfer of the melanoma differentiation-associated gene-7 (mda-7) elicits growth inhibition and apoptosis in various tumor types. Here, we evaluate the effects of Ad-mda7, alone and in combination with other therapies, against a panel of nine breast tumor cell lines and their normal counterparts; we report selective Ad-mda7-mediated p53-independent growth inhibition, G2/M cell cycle arrest, and apoptosis. In vivo, Ad-mda7 induced p53-independent tumor growth inhibition (P<0.004) in multiple xenograft models. We then evaluated the combination of Ad-mda7 with agents commonly used to treat breast cancer: radiotherapy (XRT), Tamoxifen, Taxotere, Adriamycin, and Herceptin. These agents exhibit diverse modes of action, including formation of bulky adducts, inhibition of DNA replication (Adriamycin, XRT), damage to microtubules (Taxotere), nonsteroidal estrogen antagonists (Tamoxifen), or Her2/neu receptor blockade (Herceptin). Treated with conventional anticancer drugs or radiation, MDA-7-expressing cells display additive or synergistic cytotoxicity and apoptosis that correlates with decreased BCL-2 expression and BAX upregulation. In vivo, animals that received Ad-mda7 and XRT underwent significant reduction of tumor growth (P<0.002). This is the first report of the synergistic effects of Ad-mda7 combined with chemotherapy or radiotherapy on human breast carcinoma cells.


Subject(s)
Breast Neoplasms/therapy , Carcinoma/therapy , Genetic Therapy , Interleukins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Adenoviridae/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Biological Therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Carcinoma/drug therapy , Carcinoma/radiotherapy , Combined Modality Therapy , Female , Gene Transfer Techniques , Humans , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-bcl-2/analysis
4.
Cancer Chemother Pharmacol ; 50(1): 46-52, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12111111

ABSTRACT

PURPOSE: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the tumor necrosis factor (TNF) family that preferentially kills tumor cells. In this study, we sought to determine whether chemotherapeutic agents augment TRAIL-induced cytotoxicity in human prostate cancer cells, and whether this sensitivity can be blocked by overexpression of bcl-2. METHODS: Prostate cancer cells, PC3 and LNCaP, were treated with TRAIL alone, drug alone or a combination of both for 24 h. Cytotoxicity was determined by DNA fragmentation and clonogenic survival assay. RESULTS: Treatment with the conventional chemotherapeutic agents cisplatin (2 and 5 microg/ml), etoposide (10 microM and 20 microM) and doxorubicin (30 and 60 n M) dramatically augmented TRAIL-induced apoptosis in LNCaP and PC3 cells. TRAIL-induced apoptosis was partially abrogated by overexpression of bcl-2 in these two cell lines when it was used in combination with the above agents. Similar results were obtained using clonogenic survival assays where bcl-2 overexpression was also found to marginally protect against TRAIL- and chemotherapy-induced cell killing. CONCLUSIONS: This study demonstrates that combination treatment of prostate cancer cells with TRAIL and chemotherapeutic agents overcomes their resistance by triggering caspase activation. This greater than additive effect of cotreatment with TRAIL and chemotherapy may provide the basis for a new therapeutic approach to induce apoptosis in otherwise resistant cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Membrane Glycoproteins/pharmacology , Prostatic Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , Blotting, Western , Caspases/metabolism , Cell Survival/drug effects , Cisplatin/pharmacology , Combined Modality Therapy , Doxorubicin/pharmacology , Drug Synergism , Etoposide/pharmacology , Humans , Male , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured/drug effects , Tumor Stem Cell Assay
5.
Int J Cancer ; 96(6): 327-33, 2001 Dec 20.
Article in English | MEDLINE | ID: mdl-11745502

ABSTRACT

The proteasome pathway is important for the turnover of many regulatory proteins. This pathway has recently become a target for antitumor agents and several research groups have demonstrated that inhibitors with specificities for the proteasome are potent apoptosis-inducing agents. Many mechanisms by which proteasome inhibitors exert their effects have been suggested, including inhibition of NF-kappa B activity and stabilization of the p53 tumor suppressor protein. We investigated the ability of inhibitors with specificities for the proteasome and for another protein degradation enzyme, calpain, to sensitize a murine B-cell lymphoma with constitutive NF-kappa B1 homodimer activity and high expression of Bcl-2 protein to radiation-induced apoptosis. Protease inhibitors tested were calpain inhibitor I, calpain inhibitor II, calpeptin, MG132, and Lactacystin. All five inhibitors induced apoptosis and sensitized cells to radiation despite the maintenance of Bcl-2 protein levels throughout the course of treatment. An electrophoretic migration shift assay for NF-kappa B1 activity provided evidence that reversal of NF-kappa B activity was not required for induction of cell death; however, p53 levels were elevated for all inhibitors tested. HL-60 cells, devoid of p53, could not be sensitized to radiation by MG132 treatment, suggesting that p53 was important for cell death induced by combined treatment with protease inhibitors and radiation. We concluded that protease inhibitors are capable of overcoming the protective effects of Bcl-2 to induce apoptosis and suggest that protease inhibitor treatment, when combined with ionizing radiation, leads to p53-mediated apoptosis.


Subject(s)
Apoptosis/radiation effects , Lymphoma/radiotherapy , Multienzyme Complexes/antagonists & inhibitors , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/analysis , Animals , Apoptosis/drug effects , Cysteine Endopeptidases/physiology , Humans , Lymphoma/pathology , Mice , Multienzyme Complexes/physiology , NF-kappa B/metabolism , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-bcl-2/physiology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/analysis
6.
Carcinogenesis ; 22(12): 1939-46, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751423

ABSTRACT

Fanconi anemia (FA) is a human autosomal disorder characterized by cancer susceptibility and cellular sensitivity to DNA crosslinking agents such as mitomycin C and diepoxybutane. Six FA genes have been cloned including a gene designated XRCC9 (for X-ray Repair Cross Complementing), isolated using a mitomycin C-hypersensitive Chinese hamster cell mutant termed UV40, and subsequently found to be identical to FANCG. A nuclear complex containing the FANCA, FANCC, FANCE, FANCF and FANCG proteins is needed for the activation of a sixth FA protein FANCD2. When monoubiquitinated, the FANCD2 protein co-localizes with the breast cancer susceptibility protein BRCA1 in DNA damage induced foci. In this study, we have assigned NM3, a nitrogen mustard-hypersensitive Chinese hamster mutant to the same genetic complementation group as UV40. NM3, like human FA cell lines (but unlike UV40) exhibits a normal spontaneous level of sister chromatid exchange. We show that both NM3 and UV40 are also hypersensitive to other DNA crosslinking agents (including diepoxybutane and chlorambucil) and to non-crosslinking DNA damaging agents (including bleomycin, streptonigrin and EMS), and that all these sensitivities are all corrected upon transfection of the human FANCG/XRCC9 cDNA. Using immunoblotting, NM3 and UV40 were found not to express the active monoubiquitinated isoform of the FANCD2 protein, although expression of the FANCD-L isoform was restored in the FANCG cDNA transformants, correlating with the correction of mutagen-sensitivity. These data indicate that cellular resistance to these DNA damaging agents requires FANCG and that the FA gene pathway, via its activation of FANCD2 and that protein's subsequent interaction with BRCA1, is involved in maintaining genomic stability in response not only to DNA interstrand crosslinks but also a range of other DNA damages including DNA strand breaks. NM3 and other "FA-like" Chinese hamster mutants should provide an important resource for the study of these processes in mammalian cells.


Subject(s)
DNA Damage/drug effects , DNA-Binding Proteins/genetics , Mutagens/pharmacology , Mutation/genetics , Nuclear Proteins/metabolism , Sister Chromatid Exchange/genetics , Ubiquitin/metabolism , Animals , Bleomycin/pharmacology , CHO Cells , Cell Line , Cricetinae , DNA Damage/genetics , Epoxy Compounds/pharmacology , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein , Fanconi Anemia Complementation Group G Protein , Gamma Rays , Genetic Complementation Test , Humans , Hybrid Cells , Mechlorethamine/pharmacology , Mitomycin/pharmacology , Nuclear Proteins/chemistry , Sister Chromatid Exchange/drug effects , Ultraviolet Rays
7.
Semin Oncol ; 28(4 Suppl 14): 50-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11605184

ABSTRACT

Targeting the specific genetic lesions responsible for carcinogenesis and cancer progression is an attractive strategy for developing more effective anticancer therapies and reducing treatment-related toxicity. The restoration of defective tumor suppressor gene pathways by replacement of tumor suppressor genes in cancer cells has been studied in lung cancer. The most extensively studied agent is the wild-type p53 tumor suppressor gene delivered by an adenoviral vector. Clinical trials to date in non-small cell lung cancer and head and neck cancer have consistently shown evidence of gene transduction and expression, mediation of apoptosis, and clinical responses including pathologic complete responses. However, it also is clear that this approach can be improved. Promising avenues for investigation include improved gene delivery systems, induction of bystander effects, and adjuvant use of gene therapy with conventional chemotherapy, radiation therapy, and surgery. However, these strategies will need further refinement to succeed clinically. This review examines several important issues in cancer gene therapy in general and the most recent achievements in gene therapy for non-small cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Genetic Therapy , Lung Neoplasms/therapy , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Clinical Trials as Topic , Evaluation Studies as Topic , Genes, Tumor Suppressor , Genes, p53 , Humans , Lung Neoplasms/genetics
8.
Clin Cancer Res ; 7(9): 2887-97, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11555607

ABSTRACT

Chemotherapy given sequentially or concurrently with external beam radiation therapy has emerged as a standard for the treatment of locally advanced lung cancer. Gene therapy by adenovirus-mediated wild-type p53 gene transfer has been shown to inhibit lung cancer growth in vitro, in animal models, and in human clinical trials. However, no information is available on the combined effects of p53 gene transfer, chemotherapy, and radiation therapy on lung cancer growth in vitro and in vivo. Therefore, we developed two-dimensional and three-dimensional isobologram modeling and statistical methods to evaluate the synergistic, additive, or antagonistic efficacy among these therapeutic agents in human non-small cell lung cancer cell lines A549, H460, H322, and H1299, at the ID50 and ID80 levels. The combination of these three therapeutic agents exhibited synergistic inhibitory effects on tumor cell growth in all four cell lines at both the ID50 and the ID80 levels in vitro. In mouse models with H1299 and A549 xenografts, combined treatment synergistically inhibited tumor growth in the absence of any apparent increase in toxicity, when compared with other treatment and control groups. Together, our findings suggest that a combination of gene therapy, chemotherapy, and radiation therapy may be an effective strategy for human cancer treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Division/drug effects , Lung Neoplasms/therapy , Paclitaxel/pharmacology , Taxoids , Tumor Suppressor Protein p53/physiology , Adenoviridae/genetics , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Division/genetics , Cell Division/radiation effects , Combined Modality Therapy , Docetaxel , Female , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Paclitaxel/analogs & derivatives , Paclitaxel/therapeutic use , Radiotherapy , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
9.
J Biol Chem ; 276(48): 45380-6, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11567031

ABSTRACT

The bcl-2 proto-oncogene is frequently expressed in human cancer. Although bcl-2 was first cloned as the t(14;18) translocation breakpoint from human follicular B-cell lymphoma, it has become apparent that many cell types express bcl-2 because of transcriptional regulation. As such, several transcription factors have been demonstrated to activate expression of bcl-2, including NF-kappaB. We investigated the role of NF-kappaB1 (p50) homodimers in the expression of Bcl-2 in two murine B-cell lymphoma cell lines: LY-as, an apoptosis-proficient line with low Bcl-2 protein expression and no nuclear NF-kappaB activity, and LY-ar, a nonapoptotic line with constitutive p50 homodimer activity and 30 times more Bcl-2 protein expression than LY-as. We found that nuclear p50 homodimer activity correlated with Bcl-2 expression in these cell types and identified several sites within the bcl-2 5'-flanking region that p50 was capable of binding. In vitro transcription revealed that recombinant p50 enhanced the production of run-off transcripts from the bcl-2 P1 promoter. Additional in vitro transcription experiments suggested the sites by which p50 afforded this effect. We conclude that the p50 homodimer is capable of transcriptional activation of the bcl-2 gene and suggest that its nuclear activity contributes to the expression of bcl-2 in LY-ar cells.


Subject(s)
NF-kappa B/chemistry , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription, Genetic , Animals , Apoptosis , Binding Sites , Blotting, Western , Calpain/pharmacology , Cell Line , Cell Nucleus/metabolism , Dimerization , Enzyme Inhibitors/pharmacology , Mice , NF-kappa B p50 Subunit , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
10.
Oncogene ; 20(29): 3757-65, 2001 Jun 28.
Article in English | MEDLINE | ID: mdl-11439339

ABSTRACT

To determine if TRAIL-induced apoptosis in human prostate tumor cells was suppressed by bcl-2, we compared the levels of apoptosis induced by recombinant human TRAIL in pairs of isogenic cell lines that do or do not express bcl-2. Three human prostate tumor cell lines (PC3, DU145 and LNCaP) and their bcl-2-expressing counterparts were tested for their susceptibility to TRAIL. Cells were exposed to TRAIL in the presence of cycloheximide which acted as a sensitizer. Apoptosis was induced rapidly in PC3 and DU145 neo-control transfected cells, whereas induction in LNCaP required 24 h. All three cell line variants expressing bcl-2 were resistant to the apoptotic effects of TRAIL. Caspase 3 and 8 activation was also detected in the neo control cells after treatment with TRAIL, demonstrating the rapid activation of the caspase cascade similar to that seen with other death receptors. Bcl-2 overexpression in these cells blocked activation of these caspases, suggesting that bcl-2 expression of human cancer cells may be a critical factor in the therapeutic efficacy of TRAIL.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/pharmacology , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Antineoplastic Agents/metabolism , Apoptosis Regulatory Proteins , BH3 Interacting Domain Death Agonist Protein , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/biosynthesis , Carrier Proteins/metabolism , Caspases/metabolism , Cycloheximide/pharmacology , Cytochrome c Group/metabolism , Enzyme Activation , Humans , Ligands , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Prostatic Neoplasms/metabolism , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
11.
Int J Radiat Biol ; 77(2): 185-94, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11236925

ABSTRACT

PURPOSE: We compared the ability of adenoviral-mediated wild-type p53 RPR/INGN201(Ad5/CMV/p53) to radiosensitize non-small cell lung carcinoma (NSCLC) and normal lung fibroblast cells. MATERIALS AND METHODS: NSCLC cell lines (A549 and H322) and human lung fibroblast cells (MRC-9 and CCD-16) were used in this study. Radiosensitivity was determined by clonogenic assay and tumor growth delay. Expression of p53, Bax, and p21WAF1 protein were evaluated by immunoblot. A FITC conjugate of annexin V was used for flow cytometric detection of apoptosis. RESULTS: Clonogenic and apoptotic assays indicated that Ad5/CMV/p53 enhanced the radiosensitivity of both NSCLC cell lines. On the other hand, the two normal human fibroblast cell lines appeared to be resistant to the cytotoxic effects of Ad5/CMV/p53 and were not radiosensitized compared to the NSCLC cells. According to immunoblot analysis, Bax expression was increased in the NSCLC cells treated with the combination therapy; Bax expression, however, was unchanged in normal cells. In in vivo studies, tumor growth suppression was enhanced by this combination strategy in xenograft tumors growing in nude mice compared to Ad5/CMV/p53 or radiation therapy when used alone. CONCLUSIONS: Our data indicate that therapy using Ad5/CMV/p53 and irradiation in combination is more effective than either treatment when used alone on NSCLC cells, is not limited to cells with defective endogenous p53, and does not enhance the radiosensitivity of normal cells.


Subject(s)
Adenoviridae/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Fibroblasts/radiation effects , Genes, p53/genetics , Lung/radiation effects , Proto-Oncogene Proteins c-bcl-2 , Tumor Suppressor Protein p53/biosynthesis , Animals , Annexin A5/pharmacology , Apoptosis/genetics , Apoptosis/radiation effects , Blotting, Western , Cell Division , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Dose-Response Relationship, Radiation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Fluorescein-5-isothiocyanate/pharmacology , Fluorescent Dyes/pharmacology , Gene Transfer Techniques , Humans , Lung Neoplasms , Male , Mice , Mice, Nude , Neoplasm Transplantation , Proto-Oncogene Proteins/biosynthesis , Radiation Tolerance/genetics , Time Factors , Transduction, Genetic , Tumor Cells, Cultured , bcl-2-Associated X Protein
12.
Acta Oncol ; 40(6): 739-44, 2001.
Article in English | MEDLINE | ID: mdl-11765069

ABSTRACT

Clinical trials of p53 gene replacement have provided information that will be useful in the design of future gene therapy strategies. Direct intratumor injection has low toxicity and thus can be readily combined with existing treatments. Post-injection gene expression can be documented and occurs in the presence of an anti-adenovirus immune response. Importantly, this treatment can cause tumor regression or prolonged stabilization. Future research directions will include development of more efficient vectors, use of novel genes, and combined modality approaches. Unresectable tumors are a prominent problem in oncology, with proven therapies such as radiotherapy and chemotherapy controlling less than 20% of lung cancers. Based on the preclinical and clinical studies discussed, it now seems that these conventional therapies may provide renewed potential when used in conjunction with transfer of a functional p53 gene.


Subject(s)
DNA Damage , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Genetic Therapy , Lung Neoplasms/genetics , Adenoviridae/genetics , Adenoviridae/immunology , Antineoplastic Agents/therapeutic use , Combined Modality Therapy , Genetic Vectors , Humans , Lung Neoplasms/drug therapy , Radiotherapy
13.
Anticancer Res ; 21(5): 3141-6, 2001.
Article in English | MEDLINE | ID: mdl-11848465

ABSTRACT

BACKGROUND: Cancer cells are characterized by multiple genetic defects which result in altered rates of cell division, cell death and ability to differentiate. These same molecular alterations may also contribute to therapeutic resistance. We examined the potential contribution of the pro-apoptotic gene, bax, to suppressing the growth of prostate cancer cells. MATERIALS AND METHODS: The bax-deficient DU145 prostate cancer cell line was transfected with a hemagluttinin-tagged bax (HA-bax) vector to generate stable expressing bax clones. RESULTS: Ha-bax clones exhibited a significant reduction in tumor growth compared to vector control and parental cells when xenografted into nude mice. HA-bax clones were significantly more sensitive to cell death induction by cis-diamminedichloroplatinum, etoposide, doxorubicin and gamma-radiation than vector control cells. Sensitivity to paclitaxel remained unaltered in the Ha-bax cells. CONCLUSION: These findings suggest that bax may possess a tumor suppressor function in prostatic glandular epithelial cells and be an important determinant of sensitivity to therapeutic cell death induction.


Subject(s)
Cell Death/drug effects , Cell Death/physiology , Cell Death/radiation effects , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Division/physiology , Hemagglutinins/biosynthesis , Hemagglutinins/genetics , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Radiation Tolerance/physiology , Transplantation, Heterologous , Tumor Cells, Cultured , bcl-2-Associated X Protein
14.
Oncogene ; 19(47): 5359-66, 2000 Nov 09.
Article in English | MEDLINE | ID: mdl-11103937

ABSTRACT

We examined the influence of adenovirus-mediated wild-type p16INK4a (Ad/p16) expression on the radiation sensitivity of NSCLC cell lines, all of which lacked constitutive p16INK4a but each of which varied in p53 status: A549 (-p16INK4a/ +pRb/wt-p53), H322 (-p16INK4a/ +pRb/mt-p53), and H1299 (-p16INK4a/ +pRb/deleted-p53). The in vitro clonogenic survival results indicate that Ad/p16 enhanced the radiosensitivity of A549 but not H322 or H1299. Further analysis indicated that the apoptosis induced by combination therapy using Ad/p16 plus irradiation was dependent on the endogenous p53 status of the cancer cells. We performed Western blotting to analyse the p53 protein expression of A549 cells treated with either Ad/p16 or Ad/Luc. Endogenous p53 protein levels were higher in A549 cells transfected with Ad/p16 than in those transfected with Ad/Luc. Furthermore, when wt-p53 protein expression was restored in H1299 using Ad/ p53, Ad/p16 stabilized p53 protein expression and radiosensitized the cells. These results suggest that Ad/ p16-induced stabilization of p53 protein may play an important role in Ad/p16 mediated radiosensitization by enhancing or restoring apoptosis properties. Thus, Ad/ p16 plus radiation in combination may be a useful gene therapy strategy for tumors that have wt-p53 but nonfunctional p16INK4a.


Subject(s)
Apoptosis/radiation effects , Carrier Proteins/physiology , Radiation Tolerance/radiation effects , Tumor Suppressor Protein p53/biosynthesis , Adenoviruses, Human , Blotting, Western/methods , Carcinoma, Non-Small-Cell Lung , Carrier Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p16 , Gene Expression , Genetic Vectors , Humans , Lung Neoplasms , RNA, Messenger , Ribonucleases/metabolism , Tumor Cells, Cultured
15.
Int J Radiat Oncol Biol Phys ; 48(5): 1507-12, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11121656

ABSTRACT

PURPOSE/OBJECTIVE: The effect of adenoviral-mediated p53 transgene expression on the radiation response of two human prostate cancer cell lines, the p53(wild-type) LNCaP and p53(null) PC3 lines, was examined. The objective was to determine if this vector sensitizes cells to radiation independently of their p53 status. METHODS AND MATERIALS: A recombinant adenovirus-5 vector (RPR/INGN 201, Introgen Therapeutics, Houston, TX) containing a CMV promoter and wild-type p53-cDNA (Ad5-p53) was used to facilitate p53 transgene expression. A multiplicity of infection (MOI) of 10-40 viral particles per cell was used, based on Ad5/CMV/lacz infection and staining for the beta-galactosidase reporter gene product. Clonogenic assays were performed to evaluate the degree of sensitization to radiation of viral-transduced cells compared with irradiated nontransduced controls. The relative efficacy of these treatments to induce apoptotic cell death was determined using the TUNEL assay. RESULTS: The delivery of Ad5-p53 (10 MOI) reduced control plating efficiency from 36.5% to 0.86% in the LNCaP cell line and from 75.1% to 4.1% in the PC3 cell line. After correcting for the effect of Ad5-p53 on plating efficiency, the surviving fraction after 2 Gy (SF2) of gamma-irradiation was reduced over 2.5-fold, from 0.187 to 0.072, with transgene p53 expression in the LNCaP cell line. Surviving fraction after 4 Gy (SF4) was reduced over 4.5-fold, from 0.014 to 0.003, after Ad5-p53 treatment. In the PC3 cell line, Ad5-p53 (40 MOI) reduced SF2 over 1.9-fold from 0.708 to 0.367, and SF4 over 6-fold from 0.335 to 0.056. In both the LNCaP and PC3 cell lines, the combination of Ad5-p53 plus radiation (2 Gy) resulted in supra-additive apoptosis (approximately 20% for LNCaP and approximately 15% for PC3 at 50 MOI), above that seen from the addition of the controls; control vector Ad5-pA plus RT (0.15% for LNCaP and 1.44% for PC3), Ad5-p53 alone (28.6% for LNCaP and 21.7% for PC3), RT alone (0% for LNCaP and 0.23% for PC3), or Ad5-pA alone (0.1% for LNCaP and 0.29% for PC3). CONCLUSION: The clonogenic survival and apoptosis data demonstrate that p53 transgene expression sensitizes human prostate adenocarcinoma cells in vitro to irradiation. As this effect was observed in both the p53(wild-type) LNCaP and p53(null) PC3 lines, radiosensitization was independent of p53 status.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, p53/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/radiotherapy , Radiation Tolerance/genetics , Transgenes/genetics , Adenoviridae/genetics , Cell Survival/genetics , Genes, Reporter , Genetic Vectors/physiology , Humans , Male , Prostatic Neoplasms/physiopathology , Radiobiology , Transfection , Tumor Cells, Cultured/radiation effects , beta-Galactosidase/genetics
16.
Semin Radiat Oncol ; 10(4): 333-42, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11040334

ABSTRACT

Fewer than 15% of the 170,000 patients who develop lung cancer each year will survive their disease, which shows the need for novel, more specific, and less toxic therapeutic strategies. Recent advances in molecular biology have made it possible to ascertain which genetic alterations contribute to the etiology of cancer. For example, the tumor-suppressor gene, p53, responsible for directing repair of damaged DNA or committing a cell to apoptosis, is mutated or otherwise altered in more than 50% of cancers, including 40% to 70% of non-small cell lung cancers. Many p53-deficient tumors have proven remarkably resistant to radiotherapy and chemotherapy. The preclinical and clinical studies of gene therapy reviewed in this article show (1) successful transfer and expression of a potentially therapeutic p53 gene construct in tumor cells, (2) observation of antitumor effects in vitro and in vivo, and (3) most critically, a lack of significant toxicity. The results of these studies indicate that gene replacement therapy is a feasible alternative therapy for cancer. In addition, these studies show that transfer of the p53 gene can induce radiation sensitization in previously radiation-resistant tumors, leading to the possibility of new therapeutic protocols combining gene replacement with radiation therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Genetic Therapy , Lung Neoplasms/therapy , Apoptosis/physiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Cycle/physiology , Combined Modality Therapy , Gene Transfer Techniques , Genes, p53/physiology , Genetic Vectors , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy
17.
Clin Cancer Res ; 6(3): 887-90, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741712

ABSTRACT

Overexpression of wild-type p53 in cancer cells by adenovirus-mediated p53 gene transfer can result in the induction of apoptosis. To identify the potential mediators of this p53-induced apoptosis, we examined apoptotic protein levels in human lung cancer cells after Adp53 gene transfer. We observed up-regulation of Bax and Bak protein levels 18-36 h after transduction with Adp53 in H1299, H358, and H322 lung cancer cells. Contrary to expected observations, no changes in Bcl-2 and Bcl-X(L) protein levels were observed. Morphological cell changes and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining showed evidence of apoptosis in all cell lines 48 h after transduction with Adp53. These results indicate that the induction of apoptosis by adenovirus-mediated p53 transfer may be mediated by the induction of proapoptotic mechanisms rather than suppression of antiapoptotic mechanisms.


Subject(s)
Gene Transfer Techniques , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Adenoviridae/genetics , Apoptosis/genetics , Blotting, Western , DNA, Recombinant/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Up-Regulation , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein , bcl-X Protein
18.
Antioxid Redox Signal ; 2(3): 537-50, 2000.
Article in English | MEDLINE | ID: mdl-11229367

ABSTRACT

The oncogene Bcl-2 has attracted recent research attention as recognition of the importance of Bcl-2 control over apoptosis commitment in disease development and clinical response to therapy has been targeted for pharmacological intervention. Much of the basic science research regarding Bcl-2 has focused on the role that Bcl-2 plays in directly regulating mitochondrial function. This has come about because of Bcl-2's localization to mitochondrial membranes and its reported interaction with the mitochondrial megachannel. During the time that the mitochondrial function of Bcl-2 was being investigated, a smaller, yet potentially as important, role for Bcl-2 was being pursued by investigators who were following up the initial study of Bcl-2 knockout mice. These mice expressed a phenotype consistent with that of mice exposed to chronic oxidative stress. This research into the redox aspects of Bcl-2 function has led to a hypothesis that Bcl-2-expressing cells have enhanced antioxidant capacities that suppress oxidative stress signals generated during the initiation phase of many apoptotic pathways. This review will further develop the idea of Bcl-2's role in regulating cellular redox pathways associated with apoptosis, as well as integrate recently reported evidence that ties the antioxidant effects of Bcl-2 to mitochondrial function, thereby unifying both mitochondrial and redox aspects of Bcl-2 function.


Subject(s)
Oxidation-Reduction , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Antioxidants/pharmacology , Apoptosis , Glutathione/metabolism , Humans , Mice , Mitochondria/metabolism , Models, Biological , Oxidative Stress , Signal Transduction
19.
Oncology (Williston Park) ; 13(10 Suppl 5): 148-54, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10550840

ABSTRACT

Gene therapy has the potential to provide cancer treatments based on novel mechanisms of action with potentially low toxicities. This therapy may provide more effective control of locoregional recurrence in diseases like non-small-cell lung cancer (NSCLC) as well as systemic control of micrometastases. Despite current limitations, retroviral and adenoviral vectors can, in certain circumstances, provide an effective means of delivering therapeutic genes to tumor cells. Although multiple genes are involved in carcinogenesis, mutations of the p53 gene are the most frequent abnormality identified in human tumors. Preclinical studies both in vitro and in vivo have shown that restoring p53 function can induce apoptosis in cancer cells. High levels of p53 expression and DNA-damaging agents like cisplatin (Platinol) and ionizing radiation work synergistically to induce apoptosis in cancer cells. Phase I clinical trials now show that p53 gene replacement therapy using both retroviral and adenoviral vectors is feasible and safe. In addition, p53 gene replacement therapy induces tumor regression in patients with advanced NSCLC and in those with recurrent head and neck cancer. This article describes various gene therapy strategies under investigation, reviews preclinical data that provide a rationale for the gene replacement approach, and discusses the clinical trial data available to date.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Genes, p53 , Genetic Therapy/methods , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Cell Cycle/physiology , Clinical Trials as Topic , Combined Modality Therapy , Gene Expression , Genes, Tumor Suppressor/drug effects , Genes, Tumor Suppressor/radiation effects , Genes, p53/drug effects , Genes, p53/radiation effects , Genetic Therapy/trends , Genetic Vectors , Humans , Radiation-Sensitizing Agents/therapeutic use
20.
Carcinogenesis ; 20(9): 1715-20, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10469615

ABSTRACT

We have developed an in vivo model system of mouse mammary preneoplasias in order to examine the cell and molecular changes that occur during tumorigenesis. Most of these preneoplasias are characterized by an alveolar hyperplasia morphologically similar to that present in normal pregnant mammary gland, but have tumor forming capabilities ranging from very low to high. One of these hyperplasias, the TM3 HOG (transformed mammary hyperplastic outgrowth), forms tumors infrequently and has the unusual characteristic of spontaneous regression. We have observed that 7-8 months post-transplantation into the cleared mammary fat pad of a BALB/c mouse, the TM3 hyperplasia will begin to regress, leaving only a sparse ductal tree with remnant alveolar structures by 10-12 months post-transplantation. We have sought to elucidate the mechanism of this regression by determining the apoptotic and proliferative rates of the alveolar cells during TM3 HOG development. Studies show that apoptotic rates in the TM3 HOG are consistently high (4-7%) at all times after transplantation. This apoptotic rate is higher than the rates found in other preneoplasias in our system and approach the rates observed in the normal involuting gland. An unusual p53 mutation, a serine insertion at codon 233, may be causally related to the high spontaneous apoptotic frequencies as well as elevated inducible apoptotic frequencies in TM3. In addition, a sudden decrease ( approximately 63%) in proliferation occurs around 8 months post-transplantation. Furthermore, transplantation experiments indicate that the ability of the 8-month-old host and/or mammary gland to support growth of preneoplastic mammary tissues is markedly diminished compared with 3- or 6-month-old hosts. The results presented here suggest that the persistent high apoptotic rates, concomitant with decreased proliferation rates, may be responsible for TM3's regression and implicate a unique mutant p53 as a causal factor. Additionally, the results suggest that host determinants can interact with specific molecular changes in the preneoplastic cells to influence growth and progression of the preneoplastic populations.


Subject(s)
Apoptosis/genetics , Genes, p53 , Mammary Glands, Animal/pathology , Precancerous Conditions/genetics , Age Factors , Animals , Apoptosis/radiation effects , Cell Division , Codon/genetics , Female , Gamma Rays , Hyperplasia , Mammary Glands, Animal/transplantation , Mice , Mice, Inbred BALB C , Mutation , Precancerous Conditions/pathology , Remission, Spontaneous , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...