Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Br J Cancer ; 91(8): 1425-7, 2004 Oct 18.
Article in English | MEDLINE | ID: mdl-15467765

ABSTRACT

Recently, completed phase III studies demonstrated a survival benefit for a fixed number of cycles of docetaxel-containing chemotherapy treatment of androgen-independent prostate cancer (AIPC). Management of patients who respond well to initial chemotherapy for AIPC remains ill-defined. We previously reported that in a select group of such patients, retreatment with the same regimen was feasible and was associated with quality of life gains. Here, we report that multiple cycles of such intermittent chemotherapy are feasible. We prospectively tested intermittent chemotherapy in eight AIPC patients responding to calcitriol plus docetaxel who reached a serum prostate-specific antigen (PSA) <4 ng ml(-1) (22% of the 37 patients who were initially treated with this regimen). Chemotherapy was suspended until a rise in PSA > or =50% and 1 ng ml(-1). The median duration of the first treatment holiday was 20 weeks (13-74 weeks) and all patients retained sensitivity to retreatment. Four patients were eligible for a second chemotherapy holiday, and the median duration was 21 weeks (17-28 weeks). Two patients elected to take a third chemotherapy holiday, which lasted 10 and 28 weeks. The median time to treatment failure was 26.5 months (95% CI 23.6-29.4 months), and the median survival is 41 months (95% CI 33.7-48.3 months). Multiple cycles of intermittent chemotherapy interrupted by clinically meaningful treatment holidays are feasible in a subset of AIPC patients treated with this docetaxel-containing regimen. Intermittent chemotherapy for AIPC is feasible and deserves further study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Bone Neoplasms/secondary , Calcitriol/administration & dosage , Disease-Free Survival , Docetaxel , Feasibility Studies , Humans , Lymphatic Metastasis , Male , Neoplasms, Hormone-Dependent/pathology , Prospective Studies , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Survival Rate , Taxoids/administration & dosage
2.
Br J Cancer ; 89(6): 968-70, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12966410

ABSTRACT

Intermittent use of chemotherapy for androgen-independent prostate cancer (AIPC) instead of treatment until disease progression may reduce toxicity. We prospectively tested this approach in eight AIPC patients responding to calcitriol plus docetaxel who reached a serum prostate-specific antigen (PSA) <4 ng ml(-1). Chemotherapy was suspended until a rise in PSA>/=50% and 1 ng ml(-1). The median duration of treatment holiday was 20 weeks (13-43+weeks) and all patients retained sensitivity to re-treatment. Chemotherapy holiday was associated with an improvement of fatigue (P=0.05). Intermittent chemotherapy for AIPC is feasible and deserves further study.


Subject(s)
Androgen Antagonists/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Prostatic Neoplasms/drug therapy , Taxoids , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Bone Neoplasms/secondary , Calcitriol/administration & dosage , Dexamethasone/administration & dosage , Disease-Free Survival , Docetaxel , Humans , Lymphatic Metastasis , Male , Middle Aged , Paclitaxel/administration & dosage , Prospective Studies , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Survival Rate
3.
Article in English | MEDLINE | ID: mdl-15195126

ABSTRACT

Glutathione S-transferase P1 (GSTP1) is markedly downregulated in prostate cancer and prostatic intraepithelial neoplasia compared to normal prostate tissue. Downregulation of GSTP1 may, therefore, be an early event in prostate carcinogenesis. An A-->G polymorphism at nucleotide 313 results in an amino acid substitution (Ile105Val) in the substrate binding site of GSTP1 and reduces catalytic activity of GSTP1. In a study of 36 prostate cancer patients, Harries et al. reported that the Ile/Ile genotype is associated with a decreased risk of prostate cancer (odds ratio 0.4 (0.17-0.82)). We sought to confirm this finding and to examine the impact of this polymorphism together with several related polymorphisms implicated as risk factors for carcinogen-associated malignancies. One hundred and seventeen patients with prostate adenocarcinoma and 183 population-based controls were recruited to this case-control study. Genotyping of the GSTP1 (Ile105Val), GSTM1 (null), GSTT1 (null) and CYP1A1 (Ile462Val) genes was performed using polymerase chain reaction (PCR) based techniques on DNA prepared from peripheral blood. A questionnaire was used to collect demographic information from each subject. Cases were significantly older (P<0.0001) and had significantly greater family history of prostate cancer (P<0.0001), confirming known risk factors for this disease. By chi(2) analysis, none of the genotype distributions varied among cases and controls. Using a logistic regression model to control for known risk factors we were also unable to demonstrate a significant association with prostate cancer for any of the polymorphisms tested. This population fails to identify a relationship between the above polymorphisms and prostate adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Glutathione Transferase/genetics , Isoenzymes/genetics , Polymorphism, Genetic , Prostatic Neoplasms/genetics , Adenocarcinoma/enzymology , Aged , Amino Acid Substitution , Case-Control Studies , Cytochrome P-450 CYP1A1/genetics , Genotype , Glutathione S-Transferase pi , Glutathione Transferase/deficiency , Homozygote , Humans , Male , Middle Aged , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/enzymology , Risk Factors
4.
Prostate ; 49(4): 263-6, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11746272

ABSTRACT

BACKGROUND: A common germline polymorphism of p53 produces a protein with an Arg to Pro change at codon 72. This Pro variant has altered biochemical properties suggesting altered cancer susceptibility. METHODS: A case control study with 115 men with prostate cancer and 181 community control male subjects was conducted. Demographics, family history of cancer, and blood were obtained. Codon 72 genotypes were determined using PCR. RESULTS: The Pro/Pro genotype was associated with a markedly lower risk of prostate cancer (OR = 0.23, CI = 0.07-0.79, P = 0.012). Similar reduction in risk was observed when the analysis was limited to Caucasian subjects (86% of total). Reduction in risk remained significant in a logistic regression model after correcting for age and family history of prostate cancer (OR = 0.14, CI = 0.03-0.71, P = 0.017). CONCLUSIONS: Men with the p53 codon 72 Pro/Pro genotype appear to be at reduced risk of prostate cancer.


Subject(s)
Adenocarcinoma/genetics , Genes, p53/genetics , Polymorphism, Genetic , Prostatic Neoplasms/genetics , Aged , Case-Control Studies , Codon , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Electrophoresis, Agar Gel , Genotype , Humans , Logistic Models , Male , Middle Aged , Polymerase Chain Reaction , Surveys and Questionnaires
5.
Ann Oncol ; 12(9): 1273-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11697840

ABSTRACT

BACKGROUND: This study sought to define the activity and toxicity of weekly docetaxel in patients with androgen-independent prostate cancer and cancer-related pain. PATIENTS AND METHODS: Twenty-five patients were treated with docetaxel 36 mg/m2 i.v. administered weekly for six consecutive weeks followed by two weeks without treatment. This eight-week treatment cycle was repeated until progression or unacceptable toxicity. Endpoints included palliative response (a 2-point reduction on the 6-point Present Pain Intensity scale without an increase in analgesic consumption or a 50% decrease in analgesic use without an increase in pain), PSA response (a 50% decrease maintained at least four weeks), measurable disease response, survival, and toxicity. RESULTS: Twelve of 25 patients (48%, 95% confidence interval (95% CI): 28%-68%) had a palliative response. Eleven of the 24 patients who entered with an elevated PSA (46%, 95% CI: 25%-67%) had a PSA response. Two of five patients with measurable disease had a partial response. Toxicity of therapy was modest with no treatment-related mortality. Twenty-five percent of patients experienced a grade 3 or 4 hematologic toxicity and 36% of patients experienced a grade 3 non-hematologic toxicity. CONCLUSIONS: Weekly docetaxel is well tolerated in patients with androgen-independent prostate cancer and has significant activity as measured by relief of pain, reduction in PSA, and reduction in measurable disease.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/analogs & derivatives , Paclitaxel/pharmacology , Prostatic Neoplasms/drug therapy , Taxoids , Aged , Aged, 80 and over , Analgesics/therapeutic use , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/adverse effects , Disease Progression , Docetaxel , Drug Administration Schedule , Humans , Infusions, Intravenous , Male , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Pain/drug therapy , Pain/etiology , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology
6.
Mol Biol Rep ; 28(1): 19-25, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11710560

ABSTRACT

To investigate mechanisms of rat glutathione S-transferase P1 gene (rGSTP1) expression regulation during chemical carcinogenesis. we studied enhancer elements located in the region between -2.5 kb to -2.2 kb. The region was upstream from the start site of transcription and was divided into two major fragments, GPEI and GPEII. The GPEII fragment was further divided into two smaller fragments, GPEII- I and GPEII-2. Using a luciferase reporter system, we identified a strong enhancer of GPEI and a weak enhancer of GPEII in HeLa and a rat hepatoma cell line CBRH79 19 cell. The enhancer of GPEII was located within the GPEII-I region. Chemical stimulation by glycidyl methatylate (GMA) and phorbol 12-o-tetradecanoate 13-acetate (TPA) analysis revealed that induction of rGSTP1 expression was mainly through GPEI. Although H2O2 could enhance GPEII enhancer activity, the enhancement is not mediated by the NF-kappaB factor that bound the NF-kappaB site in GPEII. Using electrophoretic mobility shift assays (EMSA) and the UV cross-linking assays, we found that HeLa and CBRH7919 cells had proteins that specifically bound GPEI core sequence and a 64 kDa protein that interacted with GPEII-1. The cells from normal rat liver did not express the binding proteins. Therefore, the trans-acting factors seem to be closely related to GPEI, GPEII enhancer activities and may play an important role in high expression of rGSTPI gene.


Subject(s)
Carcinogens/pharmacology , Enhancer Elements, Genetic , Glutathione Transferase/drug effects , Glutathione Transferase/genetics , Isoenzymes/drug effects , Isoenzymes/genetics , 5' Flanking Region , Animals , Carcinoma, Hepatocellular/chemically induced , Cells, Cultured , Gene Expression Regulation, Enzymologic , Genes, Reporter , Glutathione S-Transferase pi , Glutathione Transferase/metabolism , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , I-kappa B Proteins/metabolism , Isoenzymes/metabolism , Liver/drug effects , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Rats , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic , Transfection
7.
Semin Oncol ; 28(4 Suppl 15): 49-55, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11685729

ABSTRACT

Novel treatment regimens for androgen-independent prostate cancer (AIPC) are needed because currently available approaches have not been shown to improve survival. Docetaxel provides a good foundation for new therapeutic combinations because of its promising single-agent activity against prostate cancer and its favorable tolerability profile, particularly when administered weekly. In both tissue culture and animal models of prostate cancer, calcitriol (the biologically active form of vitamin D) enhanced the activity of docetaxel, paclitaxel, and platinum compounds. These effects were particularly notable at supraphysiologic calcitriol concentrations. Weekly calcitriol dosing is associated with minimal toxicity and permits substantial dose escalation over the daily schedule. A weekly calcitriol dose of 0.5 microg/kg produces plasma calcitriol levels 25-fold higher than the physiologic range. In a preclinical study at the Oregon Health Sciences University, calcitriol 5 micromol/L plus docetaxel 0.15 nmol/L was at least additive in inhibiting PC-3 colony formation. A phase II study is evaluating weekly administration of 0.5 microg/kg calcitriol orally on day 1 followed by 36 mg/m(2) docetaxel intravenously on day 2 in patients with AIPC (repeated for 6 consecutive weeks of each 8-week cycle). At the time of a preliminary analysis, 11 patients had been enrolled and were actively being treated. All 5 patients who had completed 8 weeks of calcitriol/docetaxel treatment achieved prostate-specific antigen (PSA) reductions of > or =50%. Two of these patients had confirmatory assessments, both meeting the formal PSA response criteria. Treatment has been well tolerated, with 1 patient experiencing a self-limited grade 3 toxicity and no patients experiencing grade 4 or 5 toxicities.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Calcitriol/therapeutic use , Paclitaxel/analogs & derivatives , Paclitaxel/therapeutic use , Prostatic Neoplasms/drug therapy , Taxoids , Aged , Aged, 80 and over , Calcitriol/administration & dosage , Docetaxel , Drug Administration Schedule , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Tumor Cells, Cultured
8.
Cancer ; 91(12): 2431-9, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11413535

ABSTRACT

BACKGROUND: Calcitriol is the principal biologically active metabolite of vitamin D. Calcitriol's activity against many neoplasms is well documented, but calcitriol's therapeutic application has been hampered by predictable hypercalcemia when it is given daily. Because laboratory data has suggested that intermittent exposure to high levels of calcitriol may be sufficient to produce antiproliferative effects, the authors developed a Phase I trial to determine the maximal tolerated dose, dose-limiting toxicity, and the pharmacokinetic profile of calcitriol given weekly by mouth. METHODS: Patients with refractory malignancies were enrolled for 4 weeks of treatment followed by 4 weeks of observation. Reenrollment at a higher dose level was permitted for patients who had evidence of response or stable disease and no Grade 3 or greater toxicity. The starting dose was 0.06 microg/kg. RESULTS: Fifteen patients received 20 cycles of therapy. Doses up to 2.8 microg/kg of calcitriol weekly produced no dose-limiting toxicity. While peak levels and the area under the serum concentration-time curve of calcitriol increased in a linear fashion at lower doses, saturable absorption was observed at doses above 0.48 microg/kg. Doses of 0.48 microg/kg and above produced mean peak calcitriol levels of 1625 pg/mL, approximately 25-fold greater than top normal levels and well within the therapeutic range suggested by in vitro experiments. Eight patients experienced self-limiting Grade 1 hypercalcemia. CONCLUSIONS: Weekly dosing of oral calcitriol permitted substantial dose escalation with minimal toxicity. Peak serum calcitriol levels were in the predicted therapeutic range. A dose of 0.5 microg/kg was selected for evaluation in Phase II studies.


Subject(s)
Calcitriol/administration & dosage , Neoplasms/drug therapy , Pulse Therapy, Drug , Adult , Aged , Aged, 80 and over , Calcitriol/pharmacokinetics , Calcitriol/toxicity , Calcium/metabolism , Cell Division/drug effects , Diet , Drug Administration Schedule , Female , Humans , Hypercalcemia/prevention & control , Male , Middle Aged
9.
Mutagenesis ; 16(3): 197-202, 2001 May.
Article in English | MEDLINE | ID: mdl-11320143

ABSTRACT

Monitoring patients treated with single antineoplastic agents is aiding our understanding of what hazard these drugs pose in vivo. In this study, the frequency of mutant 6-thioguanine-resistant (TG(R)) peripheral blood lymphocytes was monitored before treatment and for < or =35 weeks after treatment of patients with cyclophosphamide (CP) or chlorambucil (CAB). The mean mutant frequency before treatment for six multiple sclerosis patients treated with high-dose CP was 2.53 x 10(-5) and increased after treatment to 4.61 x 10(-5) (P = 0.08, paired t-test). Using each patient as their own control, there were significant increases (each at P < 0.04) detectable within 2-4 weeks in four of the multiple sclerosis patients treated with CP. There was no increase in an untreated control monitored over the same period. In a patient receiving five sequential CP treatments at 1 month intervals, there were cumulative increases in the frequency of mutant cells. The mutant frequency increased from 0.31 x 10(-5) before treatment to 3.64 x 10(-5) after the final treatment and had decreased to 0.53 x 10(-5) at 35 weeks after treatment. In one of two CAB-treated patients with indolent non-Hodgkin's lymphoma, there was a significant increase in mutant frequency (P < 0.03) after treatment. Freshly isolated peripheral blood lymphocytes treated with 4-hydroperoxy-CP in vitro demonstrate a dose-dependent increase in mutant frequency. The increment in mutant frequency observed in vivo is of the order expected from the in vitro experiments. Although this study demonstrates that single or multiple doses of a single antineoplastic agent are mutagenic in vivo for some patients, further studies are needed to determine the extent and mechanism of the inter-individual variations in mutagenic response.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Chlorambucil/adverse effects , Cyclophosphamide/adverse effects , Guanosine/analogs & derivatives , Lymphocytes/drug effects , Mutagens , Mutation , Adult , DNA Mutational Analysis , Dose-Response Relationship, Drug , Guanosine/pharmacology , Humans , Lymphocytes/metabolism , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/drug therapy , Thionucleosides/pharmacology , Time Factors
11.
Head Neck ; 22(6): 609-17, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10941163

ABSTRACT

OBJECTIVE: To assess whether genetic polymorphisms implicated as risk factors for other tobacco-associated malignancies are associated with altered risk of head and neck squamous cell carcinoma. DESIGN: Case-control study. SUBJECTS: One hundred sixty patients with head and neck squamous cell carcinoma recruited from a university-based head and neck oncology clinic and 149 population-based controls. METHODS: Genotyping of the CYP1A1 (Ile462Val), GSTM1 (null), GSTP1 (Ile105Val), GSTT1 (null), and P53 (Arg72Pro) genes was performed by polymerase chain reaction-based techniques on DNA prepared from peripheral blood. In addition, a questionnaire was used to collect demographic information from each subject. RESULTS: Cases were significantly older (p <.0001) and had significantly greater tobacco use (p <.0001) and were more likely to be male (p <.0001) than were control subjects, thus confirming known risk factors for this disease. When cases and controls were compared by simple chi-square analysis, only the frequency of CYP1A1 (Ile462Val) polymorphism was significantly different between cases and controls (OR =.42; 95% CI =.18-.99; p <.04). However, with a logistic regression model to control for known risk factors, we were unable to demonstrate a significant association with head and neck cancer for any of the polymorphisms tested, including CYP1A1. CONCLUSIONS: This population fails to identify a relationship between the above-mentioned polymorphisms and head and neck cancer.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Polymorphism, Genetic , Case-Control Studies , Cytochrome P-450 CYP1A1/genetics , Female , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Humans , Isoenzymes/genetics , Logistic Models , Male , Middle Aged , Polymerase Chain Reaction , Risk Factors , Smoking/adverse effects , Surveys and Questionnaires , Tumor Suppressor Protein p53/genetics
12.
Chem Biol Interact ; 111-112: 225-38, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9679557

ABSTRACT

The use of cytotoxic chemotherapy for cancer therapy has been very successful in the treatment and often cure of patients with particular neoplasms, such as testicular carcinomas and some lymphomas. In addition, the use of adjuvant chemotherapy in patients whose primary tumor has been surgically removed contributes significantly to cure rates in some of the more common malignancies such as breast carcinoma and colon cancer. Nonetheless, for most patients with metastatic malignancies, current antineoplastic drugs provide only brief remissions with few or no long term cures. In addition, the side effects of therapy lead to substantial morbidity in nearly all patients. Insights derived from model system studies on two glutathione based lead compounds, TER286 and TER199, suggest new clinical strategies and raise interesting basic research questions regarding the cell biology foundations of cancer chemotherapy.


Subject(s)
Glutathione/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biotransformation , Cytotoxins/pharmacokinetics , Cytotoxins/therapeutic use , Drug Resistance , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Female , Glutathione/analogs & derivatives , Glutathione/pharmacokinetics , Glutathione/therapeutic use , Glutathione Transferase/antagonists & inhibitors , Hematopoiesis/drug effects , Humans , Male
13.
Cancer Res ; 58(12): 2568-75, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9635580

ABSTRACT

TER286 is a latent drug activated by human glutathione S-transferase (GST) isoforms P1-1 and A1-1 to produce a nitrogen mustard alkylating agent. M7609 human colon carcinoma, selected for resistance to doxorubicin, and MCF-7 human breast carcinoma, selected for resistance to cyclophosphamide, both showed increased sensitivity to TER286 over their parental lines in parallel with increased expression of GST P1-1. In primary human tumor clonogenic assays, the spectrum of cytotoxic activity observed for TER286 was both broad and unusual when compared to a variety of current drugs. In murine xenografts of M7609 engineered to have high, medium, or low GST P1-1, responses to TER286 were positively correlated with the level of P1-1. Cytotoxicity was also observed in several other cell culture and xenograft models. In xenografts of the MX-1 human breast carcinoma, tumor growth inhibition or regression was observed in nearly all of the animals treated with an aggressive regimen of five daily doses. This schedule resulted in a 24-h posttreatment decline in bone marrow progenitors to 60% of control and was no worse than for a single dose of TER286. These studies have motivated election of TER286 as a clinical candidate.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cytotoxins/pharmacology , Glutathione Transferase/metabolism , Glutathione/analogs & derivatives , Prodrugs/pharmacology , Animals , Antineoplastic Agents, Alkylating/metabolism , Antineoplastic Agents, Alkylating/therapeutic use , Cytotoxins/metabolism , Cytotoxins/therapeutic use , Glutathione/metabolism , Glutathione/pharmacology , Glutathione/therapeutic use , Humans , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/mortality , Prodrugs/metabolism , Prodrugs/therapeutic use , Subrenal Capsule Assay , Survival Analysis , Tumor Cells, Cultured/drug effects , Tumor Stem Cell Assay
14.
Circulation ; 94(12): 3074-8, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8989110

ABSTRACT

BACKGROUND: Increased homocysteine levels are a risk factor for atherosclerosis and its sequelae. A common genetic mutation in methylenetetrahydrofolate reductase (MTHFR), an enzyme required for efficient homocysteine metabolism, creates a thermolabile enzyme with reduced activity. We determined the prevalence of this mutation in many subjects with and without vascular disease and related it to homocysteine and folate levels. METHODS AND RESULTS: DNA from 247 older subjects with vascular disease and 594 healthy subjects without vascular disease (in three different control groups) was screened for the MTHFR 677 C-to-T mutation. Within each group, 9% to 17% of the subjects were homozygous for this mutation, and the mutant allele frequency was 31% to 39%. The genotype distributions, homozygote frequencies, and allele frequencies did not differ significantly between the study groups. In the vascular disease subjects, despite significantly lower folate levels in MTHFR homozygotes, there was no significant difference in homocysteine levels among the MTHFR genotype groups. The negative slope of the regression line relating homocysteine and folate was significantly steeper for those with a homozygous MTHFR mutation compared with those without this mutation. CONCLUSIONS: Although the thermolabile MTHFR mutation is very common, it does not appear to be a significant genetic risk factor for typical late-onset vascular disease. Because MTHFR homozygotes have increased homocysteine with low folate levels, this mutation may contribute to early-onset or familial vascular disease. The genotype dependence of the folate-homocysteine correlation further suggests that homozygotes for this mutation may have both an exaggerated hyperhomocysteinemic response to folic acid depletion and a better response to folic acid therapy.


Subject(s)
Homocysteine/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Point Mutation , Vascular Diseases/genetics , Adult , Age of Onset , Aged , Base Sequence , Blood Donors , Canada , DNA Primers , Gene Frequency , Genotype , Homozygote , Humans , Infant, Newborn , Methylenetetrahydrofolate Reductase (NADPH2) , Middle Aged , Polymerase Chain Reaction , Reference Values , Regression Analysis , Vascular Diseases/physiopathology
15.
Biochemistry ; 35(47): 14679-83, 1996 Nov 26.
Article in English | MEDLINE | ID: mdl-8942627

ABSTRACT

Abasic sites in DNA are generated either spontaneously or after removal of altered bases during the base excision repair process. These as well as 3' damaged ends of DNA at single-strand breaks induced by reactive oxygen species are repaired by AP-endonucleases. The major human AP-endonuclease (named APE-1) has two unrelated activities. It may function as an activator of c-Fos and c-Jun transcription factors and as a repressor of the parathyroid hormone (PTH) gene by binding to the negative Ca(2+)-response elements (nCaRE) in its promoter. Preliminary studies indicate that the h-APE-1 gene is highly regulated. Analysis of its promoter activity by transient expression of the luciferase reporter gene in human, HeLa and TK6 cells suggested the presence of a negative regulatory element in the promoter. Two nCaRE-like sequences were identified in the promoter segment responsible for inhibiting reporter gene expression. Competitive electrophoretic mobility shift assay with HeLa nuclear extract indicated that the nCaRE sequences of the APE-1 and PTH genes are recognized by the APE-1 polypeptide. These results suggest that the APE-1 gene may be down-regulated by its own product.


Subject(s)
Gene Expression Regulation, Enzymologic , Lyases/genetics , Base Sequence , Cell Line , DNA , DNA-(Apurinic or Apyrimidinic Site) Lyase , Deoxyribonuclease IV (Phage T4-Induced) , Down-Regulation , HeLa Cells , Humans , Lyases/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid
16.
Cancer J Sci Am ; 2(3): 166-74, 1996.
Article in English | MEDLINE | ID: mdl-9166517

ABSTRACT

PURPOSE: Radiographic tumor response and survival were evaluated in patients receiving methotrexate-based chemotherapy with osmotic blood-brain barrier disruption with or without antecedent cranial radiation. PATIENTS AND METHODS: Fifty-eight non-AIDS patients (38 males, 20 females) with histologically confirmed primary central nervous system lymphoma, primarily large cell or immunoblastic, were treated at the Oregon Health Sciences University from January 1982 through March 1992. Group 1 patients (n=19) received cranial radiation prior to referral; Group 2 (n=39) received initial blood-brain barrier disruption chemotherapy. Ages ranged from 5 to 71 years (median, 57); Karnofsky performance status was 40% to 100% on inclusion (median, 80) and all underwent extensive baseline neuropsychological evaluation. RESULTS: There was no significant difference in patient characteristics between the two groups. In 15 evaluable Group I patients, 14 demonstrated objective response and 7 of 14 (50%) achieved complete response. In Group 2, 34 of 35 evaluable patients demonstrated objective response, including 29 of 34 with complete response. Estimated median survival times for Group 1 and Group 2 patients were 16 and 41 months, respectively. Currently, 19 Group 2 patients and 2 Group 1 patients are alive. Extensive neuropsychological follow-up (up to 7 years from baseline) was completed in 23 patients, which demonstrated preservation or improved cognitive function in those receiving initial chemotherapy and blood-brain barrier disruption, most notably in patients older than 60 years. CONCLUSIONS: A plateau in survival curves suggests that a portion of primary central nervous system lymphoma patients may be cured with chemotherapy and blood-brain barrier disruption without the neurologic sequelae associated with cranial radiation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/drug therapy , Cognition/drug effects , Lymphoma, B-Cell/drug therapy , Adolescent , Adult , Blood-Brain Barrier/drug effects , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Child , Child, Preschool , Combined Modality Therapy , Diuretics, Osmotic/pharmacology , Female , Humans , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/radiotherapy , Male , Mannitol/pharmacology , Middle Aged , Neuropsychological Tests , Osmosis , Prognosis , Radiotherapy , Survival Analysis , Survival Rate , Treatment Outcome
17.
Cancer Epidemiol Biomarkers Prev ; 4(6): 589-94, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8547824

ABSTRACT

The association between glutathione S-transferase M1 (GSTM1) deficiency and lung cancer risk has been controversial in the published literature. To examine this controversy, 12 case-control studies of GSTM1 status and lung cancer risk were identified in the published English literature. These studies included a total of 1593 cases and 2135 controls. We conclude that GSTM1 deficiency is a moderate risk factor for lung cancer development with an odds ratio of 1.41 (95% confidence interval = 1.23-1.61; P < 0.0001) by using Mantel-Haenszel methods for stratified analysis. This increased risk is evident for all the major histological subtypes of lung cancer. Although the increased risk is small, GSTM1 deficiency accounts for approximately 17% of lung cancer cases because of the high prevalence of GSTM1 deficiency.


Subject(s)
Glutathione Transferase/deficiency , Lung Neoplasms , Bias , Case-Control Studies , Confidence Intervals , Gene Expression , Glutathione Transferase/genetics , Humans , Incidence , Lung Neoplasms/enzymology , Lung Neoplasms/epidemiology , Lung Neoplasms/genetics , Odds Ratio , Polymorphism, Genetic , Risk Factors
18.
Neurosurgery ; 37(1): 17-27; discussion 27-8, 1995 Jul.
Article in English | MEDLINE | ID: mdl-8587686

ABSTRACT

CARBOPLATIN AND ETOPOSIDE have been investigated in preclinical studies and a limited toxicity study in 13 patients; these studies have established carboplatin and etoposide as a tolerable combination when administered with blood-brain barrier disruption. The studies also found a predictable dose-limiting toxicity of myelosuppression. Subsequently, a broad efficacy trial of this regimen was carried out. A total of 34 patients, ranging in age from 7 to 72 years, underwent a combination chemotherapy regimen of carboplatin (200 mg/m2 administered intra-arterially) and etoposide (200 mg/m2 administered intravenously) administered with blood-brain barrier disruption on each of 2 consecutive days every 28 days. The diagnoses included glioblastoma multiforme (n = 3), malignant astrocytoma (n = 8), malignant astrocytoma-oligodendroglioma (n = 1), primitive neuroectodermal tumor (n = 4), disseminated germ cell tumor of the central nervous system (CNS) (n = 6), CNS lymphoma (n = 7), and metastatic carcinoma (n = 5). The major toxicity observed in patients treated with multiple courses of this regimen was the expected reversible myelosuppression and an unexpected, irreversible high-frequency hearing loss. Of these 34 patients, 22 had measurable disease, and 9 radiographic responses (50% or more decrease in enhancing tumors) were observed in these patients. Carboplatin and etoposide with blood-brain barrier disruption is an active regimen in the treatment of malignant astrocytomas and has shown dramatic responses in primitive neuroectodermal tumors and CNS lymphoma. Additionally, the durability of responses in patients with disseminated CNS germ cell tumors is encouraging. However, such therapy is associated with unexpected high-frequency hearing loss; even so, on the basis of the favorable responses in patients with primitive neuroectodermal tumors, germ cell tumors, and lymphomas, the study of this regimen for those tumors is being extended in a multiinstitutional trial that also includes cytoxan to further evaluate the potential enhanced drug delivery.


Subject(s)
Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Carboplatin/toxicity , Etoposide/toxicity , Adolescent , Adult , Aged , Antineoplastic Agents/administration & dosage , Astrocytoma/drug therapy , Astrocytoma/surgery , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/secondary , Brain Neoplasms/surgery , Carboplatin/administration & dosage , Child , Etoposide/administration & dosage , Female , Germinoma/drug therapy , Germinoma/surgery , Glioblastoma/drug therapy , Glioblastoma/surgery , Humans , Lymphoma/drug therapy , Lymphoma/surgery , Male , Middle Aged , Neoplasm Metastasis , Neuroectodermal Tumors, Primitive/drug therapy , Neuroectodermal Tumors, Primitive/surgery , Oligodendroglioma/drug therapy , Oligodendroglioma/surgery , Tomography, X-Ray Computed
19.
J Biol Chem ; 269(51): 32672-7, 1994 Dec 23.
Article in English | MEDLINE | ID: mdl-7798274

ABSTRACT

GADD45 (growth arrest and DNA damage) is a DNA-damage-inducible gene regulated in part by the tumor suppressor p53. A role in negative growth control has recently been suggested based on significant (more than 75%) reduction of colony formation following over expression of Gadd45. To better understand the role of Gadd45, we have developed specific rabbit and murine antibodies raised against the human recombinant protein. Using these antibodies, we have found that in ML-1 cells Gadd45 is predominantly a nuclear protein. MyD118, a protein induced by terminal differentiation sharing 57% homology with Gadd45, does not cross-react with any of the antibodies produced. As expected, the induction of Gadd45 protein by ionizing radiation (IR) was also found to be dependent on a wild type p53 phenotype. Interestingly, WI-L2-NS, a human lymphoid cell line, showed very high basal levels of Gadd45 mRNA and protein in addition to a high constitutive level of a mutated p53 protein. In this cell line, the high levels of GADD45 did not inhibit cellular growth in spite of the fact that no mutations were found in GADD45 sequence. These results indicate that some cell line(s) can tolerate high levels of Gadd45 and abrogate its growth suppression function.


Subject(s)
Gene Expression Regulation , Genes, p53 , Proteins/metabolism , Animals , Baculoviridae/genetics , Base Sequence , Cell Line , Cloning, Molecular , DNA Damage , DNA Primers , Gene Expression Regulation/radiation effects , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/metabolism , Radiation, Ionizing , Spodoptera , Tumor Cells, Cultured , GADD45 Proteins
20.
Arch Biochem Biophys ; 311(2): 487-95, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8203914

ABSTRACT

The multigene family of cytosolic glutathione S-transferases (GSTs) consists of four classes (Alpha, Mu, Pi, and Theta), all involved in the detoxication of reactive electrophiles. The human Mu class GSTs consist of at least four expressed isozyme subunits, GST M1, GST M2, GST M3, and GST M4, which have 70-90% amino acid sequence identity. The gene and cDNA sequences for GST M4 have been determined recently (K. E. Comstock, K. J. Johnson, D. Rifenbery, and W. D. Henner, J. Biol. Chem. (1993) 268, 16958-16965). Cloning of GST M4 cDNA into an Escherichia coli expression system permitted the production of the corresponding protein. The enzyme was purified and shown to have a relatively low specific activity with the standard GST substrate 1-chloro-2,4-dinitrobenzene (1.4 +/- 0.2 mumol min-1 mg-1 protein), but an activity equivalent to other Mu class enzymes with other tested substrates. The protein forms functional dimers composed of subunits with a M(r) of approximately 26,400. A detailed comparison of the activity with various substrates and inhibitors was performed between GST M4-4 and other human Mu class GSTs, GST M1a-1a, GST M2-2, and GST M3-3, produced in bacterial expression systems. Despite the high level of amino acid sequence identity, the enzymatic properties of these enzymes were quite different. Comparisons with the crystallographic structure of a homologous rat GST, GST 3-3, indicate that a number of the nonconserved amino acid residues can be assigned to the putative active site of GST M4-4. This suggests that diversification in the evolution of these genes has occurred primarily in the substrate binding regions to cope with an increasing variety of foreign compounds.


Subject(s)
Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA Primers , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Glutathione Transferase/biosynthesis , Humans , Hydrogen-Ion Concentration , Isoenzymes/biosynthesis , Macromolecular Substances , Molecular Sequence Data , Molecular Weight , Plasmids , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Spectrophotometry, Ultraviolet , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...