Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Traffic Inj Prev ; 18(1): 47-55, 2017 01 02.
Article in English | MEDLINE | ID: mdl-27602598

ABSTRACT

OBJECTIVES: Multiple sclerosis (MS) is a neurological disease that commonly results in physical and cognitive dysfunction. Accordingly, MS might impact the ability to safely cross the street. The purpose of this study was to examine the feasibility of a simulated street-crossing task in persons with MS and to determine differences in street-crossing performance between persons with MS and non-MS controls. METHODS: 26 participants with MS (median Expanded Disability Status Scale [EDSS] score = 3.5) and 19 controls completed 40 trials of a virtual street-crossing task. There were 2 crossing conditions (i.e., no distraction and phone conversation), and participants performed 20 trials per condition. Participants were instructed that the goal of the task was to cross the street successfully (i.e., without being hit be a vehicle). The primary outcome was task feasibility, assessed as completion and adverse events. Secondary outcomes were measures of street-crossing performance. RESULTS: Overall, the simulated street-crossing task was feasible (i.e., 90% completion, no adverse events) in participants with MS. Participants with MS waited longer and were less attentive to traffic before entering the street compared with controls (all P < .05). Participants with MS also took longer to cross the street and were closer to oncoming vehicles when exiting the street compared to controls (all P < .05). When distracted, all participants took longer to initiate crossing, took longer to cross the street, and made more head turns while crossing (all P < .05). There were no significant group by condition interaction effects (all P > .05). CONCLUSIONS: A virtual street-crossing task is feasible for studying street-crossing behavior in persons with mild MS and most individuals with moderate MS. Virtual street-crossing performance is impaired in persons with MS compared to controls; however, persons with MS do not appear to be more vulnerable to a distracting condition. The virtual reality environment presents a safe and useful setting for understanding pedestrian behavior in persons with MS.


Subject(s)
Multiple Sclerosis/physiopathology , Multiple Sclerosis/psychology , Pedestrians/psychology , Walking/psychology , Adult , Attention/physiology , Case-Control Studies , Cell Phone , Computer Simulation , Feasibility Studies , Female , Humans , Male , Middle Aged , Psychomotor Performance , Safety , Task Performance and Analysis , Young Adult
2.
Nutr Metab Cardiovasc Dis ; 25(5): 479-88, 2015 May.
Article in English | MEDLINE | ID: mdl-25770759

ABSTRACT

BACKGROUND AND AIMS: There is some evidence that posttraumatic stress disorder (PTSD) and early life adversity may influence metabolic outcomes such as obesity, diabetes, and cardiovascular disease. However, whether and how these interact is not clear. METHODS: We analyzed data from a cross-sectional and longitudinal study to determine how PTSD severity influences obesity, insulin sensitivity, and key measures and biomarkers of cardiovascular risk. We then looked at how PTSD and early life adversity may interact to impact these same outcomes. RESULTS: PTSD severity is associated with increasing risk of obesity, diabetes, and cardiovascular disease, with higher symptoms correlating with higher values of BMI, leptin, fibrinogen, and blood pressure, and lower values of insulin sensitivity. PTSD and early life adversity have an additive effect on these metabolic outcomes. The longitudinal study confirmed findings from the cross sectional study and showed that fat mass, leptin, CRP, sICAM-1, and sTNFRII were significantly increased with higher PTSD severity during a 2.5 year follow-up period. CONCLUSIONS: Individuals with early life adversity and PTSD are at high risk and should be monitored carefully for obesity, insulin resistance, and cardiometabolic risk.


Subject(s)
Cardiovascular Diseases/psychology , Child Development , Diabetes Mellitus/psychology , Life Change Events , Metabolic Syndrome/psychology , Obesity/psychology , Stress Disorders, Post-Traumatic/complications , Adult , Biomarkers/blood , Body Mass Index , Boston/epidemiology , Cardiovascular Diseases/blood , Cardiovascular Diseases/complications , Cardiovascular Diseases/epidemiology , Child , Comorbidity , Cross-Sectional Studies , Diabetes Complications/complications , Diabetes Complications/epidemiology , Diabetes Complications/etiology , Diabetes Complications/physiopathology , Diabetes Mellitus/blood , Diabetes Mellitus/epidemiology , Female , Humans , Insulin Resistance , Longitudinal Studies , Male , Metabolic Syndrome/blood , Metabolic Syndrome/epidemiology , Middle Aged , Obesity/blood , Obesity/complications , Obesity/epidemiology , Risk Factors , Severity of Illness Index , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/etiology , Stress Disorders, Post-Traumatic/physiopathology
3.
Food Chem Toxicol ; 49(12): 3319-27, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21939727

ABSTRACT

To characterize the subchronic oral toxicity of resveratrol, CD rats received daily gavage doses of 0, 200, 400, or 1000 mg resveratrol/kg/day, and beagle dogs received daily capsule doses of 0, 200, 600, or 1200 mg resveratrol/kg/day for 90 days. Resveratrol induced only minimal toxicity, consisting of dose-related reductions in body weight gain in female rats and both sexes of dogs, and a statistically significant increase in bilirubin levels in rats at the 1000 mg/kg/day dose. Clinical observations, hematology, ophthalmology, neurotoxicity evaluations (functional observational batteries), organ weights, and gross pathology provided no biologically significant evidence of resveratrol toxicity in either species. In rats, the high dose of resveratrol reduced the incidence of cardiomyopathy; no other microscopic changes were seen. Histopathologic changes in dogs were limited to minimal inflammatory infiltrates in the kidney and urinary bladder, which were not considered toxicologically significant. A cardiovascular safety pharmacology (telemetry) study in dogs revealed no evidence of resveratrol toxicity. Based on body weight effects, the No Observed Adverse Effect Level (NOAEL) for resveratrol was 200mg/kg/day in rats and 600 mg/kg/day in dogs. The apparent cardioprotective activity of resveratrol in rats demonstrates that its potentially beneficial activities may extend beyond efficacy in cancer prevention.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cardiotonic Agents/pharmacology , Polyphenols/pharmacology , Stilbenes/toxicity , Toxicity Tests, Subchronic/methods , Administration, Oral , Animals , Body Weight/drug effects , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Kidney/drug effects , Kidney/metabolism , Male , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Rats , Resveratrol , Stilbenes/administration & dosage , Urinary Bladder/drug effects , Urinary Bladder/metabolism
4.
Toxicology ; 260(1-3): 28-36, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19464566

ABSTRACT

Standardized green tea extract was evaluated for exposure and toxicity in Beagle dogs following oral dosing by capsules. The main component (-)-epigallocatechin gallate (EGCG) accounted for 56-72% of the material. A 9-month chronic study (0, 200, 500, and 1000 mg/kg/day) was done in fasted dogs to take advantage of the reported improved catechin bioavailability with fasting. Extensive morbidity, mortality, and pathology of many major organs led to its early termination at 6.5 months and prevented identification of the toxicity mechanisms. A follow-up 13-week study examined the exposure to and toxicity of the extract. In general, toxicities were less severe than in the chronic study during the same interval. Dosing in a fed state resulted in considerably lower and less variable exposure than found under fasted conditions. Toxicity was less frequent and of lesser severity with lower exposure but limited sample size and large variability prevented reaching that definitive conclusion. Differences in mortality and morbidity between the preliminary terminated chronic and follow-up subchronic studies with the same dose of the same drug lot and similar exposure were not fully resolved as there may be other as yet unclear confounding factors.


Subject(s)
Dogs/metabolism , Flavonoids/pharmacokinetics , Flavonoids/toxicity , Phenols/pharmacokinetics , Phenols/toxicity , Plant Extracts/pharmacokinetics , Tea/chemistry , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bilirubin/blood , Blood Cell Count , Body Weight/drug effects , Female , Follow-Up Studies , Food Deprivation , Globulins/metabolism , Histamine/blood , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Male , Plant Extracts/toxicity , Polyphenols
5.
Food Chem Toxicol ; 46(3): 1068-78, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18082924

ABSTRACT

Se-methylselenocysteine (MSC) is an organoselenium compound being developed for breast cancer chemoprevention. To characterize MSC toxicity, CD rats received daily gavage doses of 0, 0.5, 1.0, or 2.0 mg/kg/day (0, 3, 6, or 12 mg/m(2)/day), and beagle dogs received daily gavage doses of 0, 0.15, 0.3, or 0.6 mg/kg/day (0, 3, 6, or 12 mg/m(2)/day) for 28 days. In rats, MSC induced dose-related hepatomegaly in both sexes; mild anemia, thrombocytopenia, and elevated liver enzymes were observed in high dose females only. Microscopic pathology included hepatocellular degeneration (high dose males, all doses in females); arrested spermatogenesis (high dose males); and atrophy of corpora lutea (middle and high dose females). In dogs, MSC induced mild anemia in middle and high dose males, and in high dose females. Toxicologically significant microscopic lesions in dogs were seen only in the liver (peliosis and vacuolar degeneration in high dose males, midzonal necrosis in males in all dose groups). Based on liver pathology seen in female rats in all dose groups, the no observed adverse effect level (NOAEL) for MSC in rats is <0.5mg/kg/day. Based on alterations in hematology parameters and liver morphology in male dogs in all dose groups, the NOAEL for MSC in dogs is <0.15 mg/kg/day.


Subject(s)
Anticarcinogenic Agents/toxicity , Breast Neoplasms/prevention & control , Cysteine/analogs & derivatives , Organoselenium Compounds/toxicity , Administration, Oral , Animals , Anticarcinogenic Agents/administration & dosage , Cysteine/administration & dosage , Cysteine/toxicity , Dogs , Female , Liver/drug effects , Liver/pathology , Male , Organoselenium Compounds/administration & dosage , Rats , Selenocysteine/analogs & derivatives
6.
Food Chem Toxicol ; 45(1): 55-63, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16965847

ABSTRACT

A six-month study was conducted in p53(+/-) mice to evaluate the possible oncogenicity of resveratrol (3,5,4'-trihydroxy-trans-stilbene), a cancer chemopreventive agent present in grapes and other foods. p53(+/-) mice (25/sex/group) received daily gavage exposure to vehicle only (negative control), resveratrol doses of 1000, 2000, or 4000 mg/kg/day, or p-cresidine (400 mg/kg/day; positive control). No mortality was seen in mice receiving the low dose of resveratrol. However, the mid and high doses induced mortality associated with impaction of the test article in the gastrointestinal tract. Resveratrol had no effect on body weight, food consumption, or clinical signs in surviving mice in any dose group, but induced dose-related increases in liver weight and serum cholesterol in both sexes. Mild anemia was seen in male mice at the high dose only; hematologic effects were not seen in females. Histopathology identified the kidney (hydronephrosis) and urinary bladder (epithelial hyperplasia) as target tissues for resveratrol toxicity. The incidences of both benign and malignant tumors in mice exposed to resveratrol were comparable to those in vehicle controls. By contrast, the positive control article, p-cresidine, induced urinary bladder cancer in both sexes. When administered to p53(+/-) mice at its maximum tolerated dose, resveratrol demonstrates no evidence of oncogenicity.


Subject(s)
Anticarcinogenic Agents/toxicity , Carcinogens/toxicity , Stilbenes/toxicity , Tumor Suppressor Protein p53/genetics , Administration, Oral , Anemia/chemically induced , Anemia/pathology , Aniline Compounds/toxicity , Animals , Anticarcinogenic Agents/pharmacokinetics , Carcinogenicity Tests , Carcinogens/pharmacokinetics , Chemoprevention , Cholesterol/blood , Dose-Response Relationship, Drug , Female , Hydronephrosis/chemically induced , Hydronephrosis/pathology , Kidney/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Longevity/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/drug effects , Resveratrol , Stilbenes/pharmacokinetics , Tumor Suppressor Protein p53/deficiency , Urinary Bladder/drug effects , Urinary Bladder/pathology
7.
Int J Toxicol ; 21(4): 277-85, 2002.
Article in English | MEDLINE | ID: mdl-12171629

ABSTRACT

The potential cancer preventive efficacy of soy isoflavones is being investigated in preclinical and phase 1 clinical studies sponsored by the U.S. National Cancer Institute. Although 90-day oral toxicity studies with PTI G-2535 (an investigational soy isoflavone drug product) in rats and dogs, as well as teratology studies, indicated no signs of toxicity, there remains a mechanistic concern associated with the ability of isoflavones (i.e., genistein) to inhibit topoisomerase, possibly leading to DNA strand breaks. The present report describes results from two in vitro genotoxicity studies, one in vivo genotoxicity study, and a single carcinogenicity study conducted in p53 knockout mice. Bacterial mutagenesis experiments using six tester strains without metabolic activation revealed no evidence that PTI G-2535 was mutagenic. In similar experiments with exogenous metabolic activation there were statistically significant increases in revertants, but less than twofold, in a single (Salmonella typhimurium TA100) tester strain. Mouse lymphoma cell mutagenesis experiments conducted with and without metabolic activation revealed statistically significant increases in mutation frequency at PTI G-2535 concentrations > or = 0.8 and 12 microg/ml, respectively; such increases were dose related and increases in the frequency of both small and large colonies were observed. A statistically significant increase in the frequency of micronucleated polychromatic erythrocytes was also seen 24 hours after treatment in male, but not female, mice who received 500 and 1000 mg/kg body weight PTI G-2535; however,such increases were small, were not dose related, and were not observed 48 hours after treatment. In contrast, dietary genistein had no effect on survival, weight gain, or the incidence or types of tumors that developed in cancer-prone rodents lacking the p53 tumor suppressor gene, p53 knockout mice. The apparent risk/benefit of isoflavone ingestion may ultimately depend on the dose and developmental timing of exposure.


Subject(s)
Carcinogens/toxicity , Genistein/toxicity , Lymphoma/genetics , Mutagens/toxicity , Neoplasms, Experimental/genetics , Administration, Oral , Animals , Bone Marrow Cells/drug effects , Carcinogenicity Tests , Escherichia coli/drug effects , Escherichia coli/genetics , Female , Lymphoma/chemically induced , Lymphoma/pathology , Male , Mice , Mice, Knockout , Micronucleus Tests , Mutation , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
8.
Urology ; 57(4 Suppl 1): 46-51, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11295594

ABSTRACT

Chemoprevention is the use of agents to slow progression of, reverse, or inhibit carcinogenesis thereby lowering the risk of developing invasive or clinically significant disease. With its long latency, high incidence and significant morbidity and mortality, prostate cancer is a relevant target for chemoprevention. Developing rational chemopreventive strategies for prostate cancer requires well-characterized agents, suitable cohorts, and reliable intermediate biomarkers of cancer. Chemopreventive agent requirements are experimental or epidemiologic data showing efficacy, safety on chronic administration, and a mechanistic rationale for activity. Current promising agents include antiandrogens and antiestrogens; steroid aromatase inhibitors; retinoids and their modulators; 5alpha-reductase inhibitors; vitamins D, E, and analogs; selenium compounds; carotenoids; soy isoflavones; dehydroepiandrostenedione and analogs; 2-difluoromethylornithine; lipoxygenase inhibitors; apoptosis inducers; and nonsteroidal anti-inflammatory drugs. Identifying biomarkers and validating them as surrogate endpoints for cancer incidence are critical for prostate chemoprevention trials. Potentially useful biomarkers for prostate chemoprevention are associated with histologic, proliferative, differentiation-related, biochemical, and genetic/regulatory features of prostatic disease. In that the prostate is not easily visualized, critical issues also include adequacy and consistency of tissue sampling. Various drugs for the chemoprevention of prostate cancer are now under evaluation in phase 1, 2, and 3 clinical trials. Cohort selection should be based on various patient characteristics (stage of the disease, previous cancers or premalignant lesions, or high risk factors) and should be conducted within the context of standard treatment.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Prostatic Neoplasms/prevention & control , Biomarkers , Clinical Trials as Topic , Cohort Studies , Humans , Male , Models, Animal , Patient Selection , Prostatic Neoplasms/epidemiology , Risk Factors
10.
IARC Sci Publ ; 154: 13-26, 2001.
Article in English | MEDLINE | ID: mdl-11220652

ABSTRACT

Relevant and feasible surrogate end-points are needed for the evaluation of intervention strategies against cancer and other chronic, life-threatening diseases. Carcinogenesis can be viewed as a process of progressive disorganization. This process is characterized by the accumulation of genotypic lesions and corresponding tissue and cellular abnormalities, including loss of proliferation and apoptosis controls. Potential surrogate end-points for cancer incidence include both phenotypic and genotypic biomarkers of this progression. In the US National Cancer Institute chemoprevention programme, histological modulation of a precancer (intraepithelial neoplasia) has so far been the primary phenotypic surrogate end-point in chemoprevention trials. Additionally, high priority has been given to biomarkers measuring specific and general genotypic changes correlated with the carcinogenesis progression model for the targeted cancer (e.g., progressive genomic instability as measured by loss of heterozygosity or amplification at specific microsatellite loci). Other potential surrogate end-points include proliferation and differentiation indices, specific gene and general chromosome damage, cell growth regulatory molecules, and biochemical activities (e.g., enzyme inhibition). Serum biomarkers thought to be associated with cancer progression (e.g., prostate-specific antigen) are particularly appealing surrogate end-points because of accessibility. Potentially chemopreventive effects of the test agent may also be measured (e.g., tissue and serum estrogen levels in studies of steroid aromatase inhibitors). To establish chemopreventive efficacy, prevention of virtually all biomarker lesions, or of those lesions with particular propensity for progression, may be required. Ideally, the phenotype and genotype of any new or remaining precancers in the target tissue of chemopreventive agent-treated subjects would show less, and certainly no greater, potential for progression than those of placebo-treated subjects.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Drug Evaluation/methods , Neoplasms/prevention & control , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Humans , Models, Genetic , Neoplasms/epidemiology , Patient Selection , Precancerous Conditions/diagnosis , Precancerous Conditions/epidemiology
11.
Cancer Epidemiol Biomarkers Prev ; 10(1): 53-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11205489

ABSTRACT

Green tea has been shown to exhibit cancer-preventive activities in preclinical studies. Its principal active components include epigallocatechin gallate (EGCG), epigallocatechin (EGC), epicatechin (EC), and epicatechin gallate, of which EGCG is the most abundant and possesses the most potent antioxidative activity. We performed a Phase I pharmacokinetic study to determine the systemic availability of green tea catechins after single oral dose administration of EGCG and Polyphenon E (decaffeinated green tea catechin mixture). Twenty healthy subjects (five subjects/dose level) were randomly assigned to one of the dose levels (200, 400, 600, and 800 mg based on EGCG content). All subjects were randomly crossed-over to receive the two catechin formulations at the same dose level. Blood and urine samples were collected for up to 24 h after oral administration of the study medication. Tea catechin concentrations in plasma and urine samples were determined using high-performance liquid chromatography with the coulometric electrode array detection system. After EGCG versus Polyphenon E administration, the mean area under the plasma concentration-time curves (AUC) of unchanged EGCG were 22.5 versus 21.9, 35.4 versus 52.2, 101.9 versus 79.7, and 167.1 versus 161.4 min x microg/ml at the 200-, 400-, 600-, and 800-mg dose levels, respectively. EGC and EC were not detected in plasma after EGCG administration and were present at low/undetectable levels after Polyphenon E administration. High concentrations of EGC and EC glucuronide/sulfate conjugates were found in plasma and urine samples after Polyphenon E administration. There were no significant differences in the pharmacokinetic characteristics of EGCG between the two study medications. The AUC and maximum plasma concentration (Cmax) of EGCG after the 800-mg dose of EGCG were found to be significantly higher than those after the 200- and 400-mg dose. The AUC and Cmax of EGCG after the 800-mg dose of Polyphenon E were significantly higher than those after the three lower doses. We conclude that the two catechin formulations resulted in similar plasma EGCG levels. EGC and EC were present in the body after the Polyphenon E administration; however, they were present predominantly in conjugated forms. The systemic availability of EGCG increased at higher doses, possibly due to saturable presystemic elimination of orally administered green tea polyphenols.


Subject(s)
Anticarcinogenic Agents/pharmacokinetics , Beverages , Catechin/analogs & derivatives , Catechin/pharmacokinetics , Administration, Oral , Adult , Anticarcinogenic Agents/administration & dosage , Area Under Curve , Catechin/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged
12.
Perception ; 30(12): 1465-88, 2001.
Article in English | MEDLINE | ID: mdl-11817754

ABSTRACT

The pattern of motion in the retinal image during self-motion contains information about the person's movement. Pursuit eye movements perturb the pattern of retinal-image motion, complicating the problem of self-motion perception. A question of considerable current interest is the relative importance of retinal and extra-retinal signals in compensating for these effects of pursuit on the retinal image. We addressed this question by examining the effect of prior motion stimuli on self-motion judgments during pursuit. Observers viewed 300 ms random-dot displays simulating forward self-motion during pursuit to the right or to the left; at the end of each display a probe appeared and observers judged whether they would pass left or right of it. The display was preceded by a 300 ms dot pattern that was either stationary or moved in the same direction as, or opposite to, the eye movement. This prior motion stimulus had a large effect on self-motion judgments when the simulated scene was a frontoparallel wall (experiment 1), but not when it was a three-dimensional (3-D) scene (experiment 2). Corresponding simulated-pursuit conditions controlled for purely retinal motion aftereffects, implying that the effect in experiment 1 is mediated by an interaction between retinal and extra-retinal signals. In experiment 3, we examined self-motion judgments with respect to a 3-D scene with mixtures of real and simulated pursuit. When real and simulated pursuits were in opposite directions, performance was determined by the total amount of pursuit-related retinal motion, consistent with an extra-retinal 'trigger' signal that facilitates the action of a retinally based pursuit-compensation mechanism. However, results of experiment 1 without a prior motion stimulus imply that extra-retinal signals are more informative when retinal information is lacking. We conclude that the relative importance of retinal and extra-retinal signals for pursuit compensation varies with the informativeness of the retinal motion pattern, at least for short durations. Our results provide partial explanations for a number of findings in the literature on perception of self-motion and motion in the frontal plane.


Subject(s)
Kinesthesis/physiology , Motion Perception/physiology , Pursuit, Smooth/physiology , Depth Perception/physiology , Discrimination, Psychological/physiology , Humans , Male , Psychophysics , Statistics as Topic
13.
Expert Opin Investig Drugs ; 9(9): 2121-38, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11060797

ABSTRACT

Increasing evidence suggests that lipoxygenase (LO)-catalysed metabolites have a profound influence on the development and progression of human cancers. Compared with normal tissues, significantly elevated levels of LO products have been found in breast tumours, colon cancers, lung, skin and prostate cancers, as well as in cells from patients with both acute and chronic leukaemias. LO-mediated products elicit diverse biological activities needed for neoplastic cell growth, influencing growth factor and transcription factor activation, oncogene induction, stimulation of tumour cell adhesion and regulation of apoptotic cell death. Agents that block LO catalytic activity may be effective in preventing cancer by interfering with signalling events needed for tumour growth. In the past ten years, pharmaceuticals agents that specifically inhibit the 5-LO metabolic pathway have been developed to treat inflammatory diseases such as asthma, arthritis and psoriasis. Some of these compounds possess anti-oxidant properties and may be effective in preventing cancer by blocking free radical-induced genetic damage or by preventing the metabolic activation of carcinogens. Other compounds may work by negatively modulating DNA synthesis. Pharmacological profiles of potential chemopreventive agents are compiled from enzyme assays, in vitro testing (e.g., cell proliferation inhibition in human cancer cells) and in vivo animal carcinogenesis models (e.g., N-methyl-N-nitrosourea-induced rat mammary cancer, benzo(a)pyrene-induced lung tumours in strain A/J mice and hormone-induced prostate tumours in rats). In this way, compounds are identified for chemoprevention trials in human subjects. Based on currently available data, it is expected that the prevention of lung and prostate cancer will be initially studied in human trials of LO inhibitors.


Subject(s)
Lipoxygenase Inhibitors/therapeutic use , Lipoxygenase/metabolism , Neoplasms/prevention & control , Animals , Arachidonic Acid/metabolism , Chemoprevention/methods , Drug Screening Assays, Antitumor/methods , Humans , Lipoxygenase/chemistry , Lipoxygenase/drug effects , Lipoxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors/pharmacology , Mice , Neoplasms/enzymology , Rats
14.
Percept Psychophys ; 62(5): 900-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10997037

ABSTRACT

Pursuit eye movements give rise to retinal motion. To judge stimulus motion relative to the head, the visual system must correct for the eye movement by using an extraretinal, eye-velocity signal. Such correction is important in a variety of motion estimation tasks including judgments of object motion relative to the head and judgments of self-motion direction from optic flow. The Filehne illusion (where a stationary object appears to move opposite to the pursuit) results from a mismatch between retinal and extraretinal speed estimates. A mismatch in timing could also exist. Speed and timing errors were investigated using sinusoidal pursuit eye movements. We describe a new illusion--the slalom illusion--in which the perceived direction of self-motion oscillates left and right when the eyes move sinusoidally. A linear model is presented that determines the gain ratio and phase difference of extraretinal and retinal signals accompanying the Filehne and slalom illusions. The speed mismatch and timing differences were measured in the Filehne and self-motion situations using a motion-nulling procedure. Timing errors were very small for the Filehne and slalom illusions. However, the ratios of extraretinal to retinal gain were consistently less than 1, so both illusions are the consequence of a mismatch between estimates of retinal and extraretinal speed. The relevance of the results for recovering the direction of self-motion during pursuit eye movements is discussed.


Subject(s)
Motion Perception/physiology , Optical Illusions/physiology , Orientation/physiology , Pursuit, Smooth/physiology , Retina/physiology , Visual Fields/physiology , Acceleration , Humans , Kinesthesis/physiology , Psychophysics
15.
Cancer Epidemiol Biomarkers Prev ; 9(4): 351-6, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10794478

ABSTRACT

Nonsteroidal antiinflammatory drugs are among the most promising chemopreventive agents for colorectal cancer. Although the mechanism by which nonsteroidal antiinflammatory drugs exert such effects remains to be further characterized, their best known pharmacological effect is inhibition of prostaglandin synthetase, which leads to decreases in tissue prostaglandin levels. We conducted a randomized, double-blind, controlled study to examine the effect of daily ibuprofen treatment on the rectal mucosal prostaglandin E2 (PGE2) levels in healthy subjects with a history of resected polyps. Study participants (n = 27) completed a 2-week run-in period and were then randomized to take a single, daily dose of ibuprofen (300 or 600 mg) or of a placebo for 4 weeks. Rectal biopsy specimens were taken before and after the run-in period and at 2 and 4 weeks after the ibuprofen/placebo treatment. Notably large between- and within-subject variability in the rectal mucosal PGE2 content was seen. The changes in PGE2 levels after ibuprofen/placebo treatment correlated with the baseline PGE2 content. After adjustment of the baseline values, 2 weeks of 300 mg/day of ibuprofen treatment resulted in significantly more suppression of PGE2 levels than that observed after the placebo treatment (55% versus 22% suppression from baseline; P = 0.033). Although other ibuprofen treatment schedules and doses appeared to result in suppression in the PGE2 levels, the suppression was not statistically significant because of the large variability in this measurement. Because lower doses are associated with fewer adverse effects, a dose of 300 mg of ibuprofen/day should be considered for future Phase II chemoprevention studies. Stratifying study participants, based on their baseline PGE2 levels and inclusion of a larger number of study subjects, are recommended for future trials where the rectal mucosal PGE2 level is to be used as a surrogate end point biomarker.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dinoprostone/analysis , Ibuprofen/pharmacology , Intestinal Polyps/complications , Rectum/drug effects , Adult , Aged , Biomarkers/analysis , Chemoprevention , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/drug effects , Intestinal Polyps/surgery , Male , Middle Aged , Rectal Neoplasms/prevention & control , Rectum/chemistry
16.
J Nutr ; 130(2S Suppl): 467S-471S, 2000 02.
Article in English | MEDLINE | ID: mdl-10721931

ABSTRACT

Because of their safety and the fact that they are not perceived as "medicine," food-derived products are highly interesting for development as chemopreventive agents that may find widespread, long-term use in populations at normal risk. Numerous diet-derived agents are included among the >40 promising agents and agent combinations that are being evaluated clinically as chemopreventive agents for major cancer targets including breast, prostate, colon and lung. Examples include green and black tea polyphenols, soy isoflavones, Bowman-Birk soy protease inhibitor, curcumin, phenethyl isothiocyanate, sulforaphane, lycopene, indole-3-carbinol, perillyl alcohol, vitamin D, vitamin E, selenium and calcium. Many food-derived agents are extracts, containing multiple compounds or classes of compounds. For developing such agents, the National Cancer Institute (NCI) has advocated codevelopment of a single or a few putative active compounds that are contained in the food-derived agent. The active compounds provide mechanistic and pharmacologic data that may be used to characterize the chemopreventive potential of the extract, and these compounds may find use as chemopreventives in higher risk subjects (patients with precancers or previous cancers). Other critical aspects to developing the food-derived products are careful analysis and definition of the extract to ensure reproducibility (e.g., growth conditions, chromatographic characteristics or composition), and basic science studies to confirm epidemiologic findings associating the food product with cancer prevention.


Subject(s)
Chemoprevention/trends , Diet , Neoplasms/prevention & control , Biomarkers , Humans , Pharmacokinetics
17.
Cancer Epidemiol Biomarkers Prev ; 9(2): 127-37, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10698472

ABSTRACT

This paper proposes a scientific basis and possible strategy for applying surrogate end points in chemopreventive drug development. The potential surrogate end points for cancer incidence described are both phenotypic (at the tissue, cellular, and molecular levels) and genotypic biomarkers. To establish chemopreventive efficacy in randomized, placebo-controlled clinical trials, it is expected that in most cases it will be critical to ensure that virtually all of the biomarker lesions are prevented or that the lesions prevented are those with the potential to progress. This would require that both the phenotype and genotype of the target tissue in agent-treated subjects, especially in any new or remaining precancers, are equivalent to or show less progression than those of placebo-treated subjects. In the National Cancer Institute chemoprevention program, histological modulation of a precancer (intraepithelial neoplasia) has thus far been the primary phenotypic surrogate end point in chemoprevention trials. Additionally, we give high priority to biomarkers measuring specific and general genotypic changes correlating to the carcinogenesis progression model for the targeted cancer (e.g., progressive genomic instability as measured by loss of heterozygosity or amplification at a specific microsatellite loci). Other potential surrogate end points that may occur earlier in carcinogenesis are being analyzed in these precancers and in nearby normal appearing tissues. These biomarkers include proliferation and differentiation indices, specific gene and general chromosome damage, cell growth regulatory molecules, and biochemical activities (e.g., enzyme inhibition). Serum biomarkers also may be monitored (e.g., prostate-specific antigen) because of their accessibility. Potentially chemopreventive drug effects of the test agent also may be measured (e.g., tissue and serum estrogen levels in studies of steroid aromatase inhibitors). These initial studies are expected to expand the list of validated surrogate end points for future use. Continued discussion and research among the National Cancer Institute, the Food and Drug Administration, industry, and academia are needed to ensure that surrogate end point-based chemoprevention indications are feasible.


Subject(s)
Biomarkers, Tumor/analysis , Chemoprevention , Drug Design , Neoplasms/prevention & control , Antineoplastic Agents/therapeutic use , Cell Transformation, Neoplastic , Humans , Research Design , Treatment Outcome
19.
Cancer Chemother Pharmacol ; 44(6): 475-83, 1999.
Article in English | MEDLINE | ID: mdl-10550568

ABSTRACT

PURPOSE: Cancer chemoprevention is the use of pharmacologic or natural agents to inhibit the development of cancer. Difluoromethylornithine (DFMO) is an irreversible inhibitor of ornithine decarboxylase, the rate-limiting enzyme in the biosynthesis of polyamines. DFMO has demonstrated chemopreventive efficacy in animal models of tumorigenesis. Tamoxifen (TAM) is currently used for treatment of estrogen receptor-positive breast carcinoma and has demonstrated efficacy in chemoprevention of breast cancer in women at high risk for the disease. The administration of tamoxifen with DFMO is being considered for development by the National Cancer Institute as a potential drug regimen for the chemoprevention of breast carcinoma. METHODS: The toxicity of DFMO in combination with TAM was evaluated in female rats following 13 weeks of daily administration by gavage. Dose groups were vehicle control, DFMO (1000 mg/kg per day), low TAM (0.25 mg/kg per day), high TAM (2.5 mg/kg per day), low combination (1000 + 0.25) and high combination (1000 + 2.5). RESULTS: No mortalities occurred in the study. Clinical signs of toxicity were limited to dermal lesions consisting of scab formation and abrasions produced by DFMO. Administration of either DFMO or TAM resulted in decreased body weight gains, with coadministration having an additive effect. Serum albumin, total protein, cholesterol and triglyceride levels were decreased in all drug-treated dose groups, although histologic evidence of liver lesions were not seen. TAM resulted in increased numbers of red blood cells, whereas DFMO produced a slightly anemic response. DFMO produced lesions in the small intestine consisting of necrosis of crypt epithelium and crypt microabscess, which were enhanced by TAM coadministration. Administration of TAM resulted in histologic changes in the ovaries, fallopian tube, vagina, cervix and uterus, indicating that inhibition of ovulation and reproductive cycle arrest in the proestrus stage had occurred. Coadministration with DFMO did not affect the changes to the reproductive system induced by TAM. CONCLUSIONS: Coadministration of DFMO with tamoxifen did not result in toxicity unique to the combination drug regimen, but rather toxicity resulted from administration of each drug. Under the conditions of the study, the overall toxicity produced by dual administration of DFMO with tamoxifen was additive with respect to the toxicity associated with each agent alone.


Subject(s)
Anticarcinogenic Agents/toxicity , Eflornithine/toxicity , Tamoxifen/toxicity , Administration, Oral , Alanine Transaminase/blood , Animals , Anticarcinogenic Agents/administration & dosage , Blood Proteins/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cholesterol/blood , Dose-Response Relationship, Drug , Drug Interactions , Eflornithine/administration & dosage , Erythrocyte Count/drug effects , Female , Genitalia, Female/drug effects , Genitalia, Female/pathology , Hematocrit , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Jejunum/drug effects , Jejunum/pathology , Rats , Serum Albumin/metabolism , Skin/drug effects , Skin/pathology , Tamoxifen/administration & dosage , Time Factors , Triglycerides/blood , Weight Gain/drug effects
20.
Toxicol Sci ; 50(1): 127-35, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10445761

ABSTRACT

DFMO, an irreversible inhibitor of ornithine decarboxylase (ODC), is under development as a chemopreventive drug against cancers with pronounced proliferative phases. In support of human clinical trials, preclinical developmental toxicity studies were conducted in pregnant rats and rabbits. Rats were treated during GD 6-17, and fetuses were obtained by C-section on GD 20. Rabbits were treated during GD 7-20, and fetuses were obtained by C-section on GD 29. The dose range-finding study in rats (5/group at 0, 50, 125, 300, 800, or 1000 mg/kg/day) revealed maternal toxicity at doses > or = 800 mg/kg/day (decreased body weights and food consumption) and developmental toxicity at doses > or = 300 mg/kg/day (increased early resorptions and reduced fetal body weights). In the main study, rats (25/group) received 0, 30, 80, or 200 mg/kg/day. Developmental toxicity in the absence of maternal toxicity was observed at 200 mg/kg/day as significantly decreased fetal weights and increased incidence of litters with skeletal variations of 14th rudimentary rib, 14th full rib, and/or 27th presacral vertebrae. There were no treatment-related fetal skeletal malformations or external or visceral anomalies at any dose level. The dose range-finding study in rabbits (5/group at 0, 30, 60, 120, 240, or 500 mg/kg/day) revealed developmental toxicity at doses > or = 60 mg/kg/day (increased resorptions and reduced fetal body weights) in the absence of maternal toxicity. In the main study, rabbits (20/group) received 0, 15, 45, or 135 mg/kg/day. Developmental toxicity in the absence of maternal toxicity was observed at 135 mg/kg/day as nonsignificantly increased early resorptions, decreased implantation sites, decreased viable fetuses, and reduced fetal weights. There were no external, visceral, or skeletal anomalies at any dose level. Thus, in the main developmental toxicity studies, DFMO produced developmental but not maternal toxicity at 200 and 135 mg/kg/day in rats and rabbits, respectively. Accordingly, in rats, the maternal no-observable-effect level (NOEL) was 200 mg/kg/day and the fetal NOEL was 80 mg/kg/day; while in rabbits the maternal NOEL was 135 mg/kg/day and the fetal NOEL was 45 mg/kg/day. These fetal NOELs are several-fold higher than the dose level currently used in Phase II and III clinical trials (approximately 13 mg/kg).


Subject(s)
Bone and Bones/drug effects , Eflornithine/toxicity , Enzyme Inhibitors/toxicity , Fetus/drug effects , Maternal-Fetal Exchange/physiology , Animals , Body Weight/drug effects , Cesarean Section/methods , Dose-Response Relationship, Drug , Female , Fetus/abnormalities , Male , Ornithine Decarboxylase/metabolism , Parity , Pregnancy , Rabbits , Rats , Specific Pathogen-Free Organisms , Teratogens/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...