Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Paul DeLucca is active.

Publication


Featured researches published by Paul DeLucca.


The New England Journal of Medicine | 2001

Comparison of early invasive and conservative strategies in patients with unstable coronary syndromes treated with the glycoprotein IIb/IIIa inhibitor tirofiban.

Christopher P. Cannon; William S. Weintraub; Laura A. Demopoulos; Ralph Vicari; Martin J. Frey; Nasser Lakkis; Franz-Josef Neumann; Debbie H. Robertson; Paul DeLucca; Peter M. DiBattiste; C. Michael Gibson; Eugene Braunwald

BACKGROUND There is continued debate as to whether a routine, early invasive strategy is superior to a conservative strategy for the management of unstable angina and myocardial infarction without ST-segment elevation. METHODS We enrolled 2220 patients with unstable angina and myocardial infarction without ST-segment elevation who had electrocardiographic evidence of changes in the ST segment or T wave, elevated levels of cardiac markers, a history of coronary artery disease, or all three findings. All patients were treated with aspirin, heparin, and the glycoprotein IIb/IIIa inhibitor tirofiban. They were randomly assigned to an early invasive strategy, which included routine catheterization within 4 to 48 hours and revascularization as appropriate, or to a more conservative (selectively invasive) strategy, in which catheterization was performed only if the patient had objective evidence of recurrent ischemia or an abnormal stress test. The primary end point was a composite of death, nonfatal myocardial infarction, and rehospitalization for an acute coronary syndrome at six months. RESULTS At six months, the rate of the primary end point was 15.9 percent with use of the early invasive strategy and 19.4 percent with use of the conservative strategy (odds ratio, 0.78; 95 percent confidence interval, 0.62 to 0.97; P=0.025). The rate of death or nonfatal myocardial infarction at six months was similarly reduced (7.3 percent vs. 9.5 percent; odds ratio, 0.74; 95 percent confidence interval, 0.54 to 1.00; P<0.05). CONCLUSIONS In patients with unstable angina and myocardial infarction without ST-segment elevation who were treated with the glycoprotein IIb/IIIa inhibitor tirofiban, the use of an early invasive strategy significantly reduced the incidence of major cardiac events. These data support a policy involving broader use of the early inhibition of glycoprotein IIb/IIIa in combination with an early invasive strategy in such patients.


The New England Journal of Medicine | 2001

Comparison of two platelet glycoprotein IIb/IIIa inhibitors, tirofiban and abciximab, for the prevention of ischemic events with percutaneous coronary revascularization.

Eric J. Topol; David J. Moliterno; Howard C. Herrmann; Eric R. Powers; Cindy L. Grines; David J. Cohen; Eric A. Cohen; M. E. Bertrand; Franz-Josef Neumann; Gregg W. Stone; Peter M. DiBattiste; Steven J. Yakubov; Paul DeLucca; Laura A. Demopoulos

BACKGROUND In the setting of percutaneous coronary revascularization, agents in the class known as platelet glycoprotein IIb/IIIa inhibitors have significantly reduced the incidence of death or nonfatal myocardial infarction at 30 days. We assessed whether there are differences in safety or efficacy between two such inhibitors, tirofiban and abciximab. METHODS Using a double-blind, double-dummy design at 149 hospitals in 18 countries, we randomly assigned patients to receive either tirofiban or abciximab before undergoing percutaneous coronary revascularization with the intent to perform stenting. The primary end point was a composite of death, nonfatal myocardial infarction, or urgent target-vessel revascularization at 30 days. The trial was designed and statistically powered to demonstrate the noninferiority of tirofiban as compared with abciximab. RESULTS The primary end point occurred more frequently among the 2398 patients in the tirofiban group than among the 2411 patients in the abciximab group (7.6 percent vs. 6.0 percent; hazard ratio, 1.26; one-sided 95 percent confidence interval of 1.51, demonstrating lack of equivalence, and two-sided 95 percent confidence interval of 1.01 to 1.57, demonstrating the superiority of abciximab over tirofiban; P=0.038). The magnitude and the direction of the effect were similar for each component of the composite end point (hazard ratio for death, 1.21; hazard ratio for myocardial infarction, 1.27; and hazard ratio for urgent target-vessel revascularization, 1.26), and the difference in the incidence of myocardial infarction between the tirofiban group and the abciximab group was significant (6.9 percent and 5.4 percent, respectively; P=0.04). The relative benefit of abciximab was consistent regardless of age, sex, the presence or absence of diabetes, or the presence or absence of pretreatment with clopidogrel. There were no significant differences in the rates of major bleeding complications or transfusions, but tirofiban was associated with a lower rate of minor bleeding episodes and thrombocytopenia. CONCLUSIONS Although the trial was intended to assess the noninferiority of tirofiban as compared with abciximab, the findings demonstrated that tirofiban offered less protection from major ischemic events than did abciximab.


The New England Journal of Medicine | 2010

Rolofylline, an Adenosine A1−Receptor Antagonist, in Acute Heart Failure

Barry M. Massie; Christopher M. O'Connor; Marco Metra; Piotr Ponikowski; John R. Teerlink; Gad Cotter; Beth Davison Weatherley; John G.F. Cleland; Michael M. Givertz; Adriaan A. Voors; Paul DeLucca; George A. Mansoor; Christina M. Salerno; Daniel M. Bloomfield; Howard C. Dittrich

BACKGROUND Worsening renal function, which is associated with adverse outcomes, often develops in patients with acute heart failure. Experimental and clinical studies suggest that counterregulatory responses mediated by adenosine may be involved. We tested the hypothesis that the use of rolofylline, an adenosine A1-receptor antagonist, would improve dyspnea, reduce the risk of worsening renal function, and lead to a more favorable clinical course in patients with acute heart failure. METHODS We conducted a multicenter, double-blind, placebo-controlled trial involving patients hospitalized for acute heart failure with impaired renal function. Within 24 hours after presentation, 2033 patients were randomly assigned, in a 2:1 ratio, to receive daily intravenous rolofylline (30 mg) or placebo for up to 3 days. The primary end point was treatment success, treatment failure, or no change in the patients clinical condition; this end point was defined according to survival, heart-failure status, and changes in renal function. Secondary end points were the post-treatment development of persistent renal impairment and the 60-day rate of death or readmission for cardiovascular or renal causes. RESULTS Rolofylline, as compared with placebo, did not provide a benefit with respect to the primary end point (odds ratio, 0.92; 95% confidence interval, 0.78 to 1.09; P=0.35). Persistent renal impairment developed in 15.0% of patients in the rolofylline group and in 13.7% of patients in the placebo group (P=0.44). By 60 days, death or readmission for cardiovascular or renal causes had occurred in similar proportions of patients assigned to rolofylline and placebo (30.7% and 31.9%, respectively; P=0.86). Adverse-event rates were similar overall; however, only patients in the rolofylline group had seizures, a known potential adverse effect of A1-receptor antagonists. CONCLUSIONS Rolofylline did not have a favorable effect with respect to the primary clinical composite end point, nor did it improve renal function or 60-day outcomes. It does not show promise in the treatment of acute heart failure with renal dysfunction. (Funded by NovaCardia, a subsidiary of Merck; ClinicalTrials.gov numbers, NCT00328692 and NCT00354458.).


Journal of Bone and Mineral Research | 1997

Precision and Discriminatory Ability of Calcaneal Bone Assessment Technologies

Susan L. Greenspan; Mary L. Bouxsein; Mary E. Melton; Andrea H. Kolodny; James H. Clair; Paul DeLucca; Michael Stek; Kenneth G. Faulkner; Eric S. Orwoll

To determine if measuring skeletal status at the calcaneus is a potentially valuable technique for diagnosing osteoporosis, we examined five calcaneal assessment techniques in 53 young normal women and 108 postmenopausal women with osteoporosis and compared these measurements to dual‐energy X‐ray absorptiometry (DEXA) at the calcaneus, hip, and spine. The five instruments, including single‐energy X‐ray absorptiometry (SEXA) and four quantitative ultrasound (QUS) instruments, were evaluated for precision, ability to discriminate osteoporotic from young normal subjects, and correlation to the other instruments. The coefficient of variation (%CV) for instrument, positioning, interobserver, and short‐term precision of the five calcaneal instruments ranged from 1.34–7.76%, 1.63–7.00%, 1.84–9.44%, and 1.99–7.04%, respectively. The %CVs for positioning, interobserver, and short‐term precision were similar for calcaneal DEXA, calcaneal SEXA, and stiffness (as measured by Achilles). The %CVs for instrument precision were similar between calcaneal DEXA and SEXA. The ability of the five calcaneal instruments to discriminate osteoporotic from young normal subjects was similar based on the analysis of area under the receiver operating characteristic curves (range 0.88–0.93) and equivalent to DEXA of the calcaneus and hip (0.88–0.93). The correlations between the measurements of five calcaneal instruments were strong (0.80 ≤ r ≤ 0.91, p < 0.001). These data suggest that although the precision is variable, the calcaneal QUS and SEXA instruments can discriminate between osteoporotic patients and young normal controls and appear to be a useful technique for assessment of osteoporosis.


Annals of Internal Medicine | 2000

Oral Montelukast Compared with Inhaled Salmeterol To Prevent Exercise-Induced Bronchoconstriction: A Randomized, Double-Blind Trial

Jonathan M. Edelman; Jennifer A. Turpin; Edwin A. Bronsky; Jay Grossman; James P. Kemp; Asma F. Ghannam; Paul DeLucca; Glenn J. Gormley; David S. Pearlman

Exercise-induced bronchoconstriction is common in patients with chronic asthma (1). Airway cooling or desiccation during exercise may trigger activation of mast cells and release of such mediators as histamine and cysteinyl leukotrienes, resulting in bronchospasm (1, 2). Cysteinyl leukotrienes (LTC4, LTD4, and LTE4), synthesized from arachidonic acid through the 5-lipoxygenase pathway, are potent bronchoconstrictors, with an effect greater than 1000 times that of histamine (3-5). Several researchers have demonstrated an increase in urinary concentrations of LTE4 after exercise (6, 7). Prophylaxis against exercise-induced bronchoconstriction with inhaled mast cell-stabilizing agents and short-acting -agonists must be administered 15 to 30 minutes before exercise. The long-acting inhaled -agonist salmeterol protects against exercise-induced bronchoconstriction for up to 12 hours, thus providing more flexibility in the dosing schedule for active patients with asthma (8, 9). However, in some patients, tolerance to salmeterol develops with long-term use, and the level of bronchoprotection diminishes by 6 to 9 hours (10-12). Montelukast sodium, a leukotriene receptor antagonist, is a potent oral medication for the treatment of asthma. The leukotriene receptor antagonists have demonstrated a significant bronchoprotective effect with exercise after one or two doses (13, 14). In patients with exercise-induced bronchoconstriction, short-term treatment with montelukast (Singulair, Merck & Co., Inc., Whitehouse Station, New Jersey) given once daily diminished the postexercise response, as described by the area under the FEV1 time curve (AUC0-60 min), by more than 50%, even at the end of the dosing interval (20 to 24 hours after administration) (15, 16). Furthermore, tolerance to the bronchoprotective effects of montelukast did not occur with long-term administration (17). We sought to test the hypothesis that the bronchoprotective effects of montelukast were greater than those of salmeterol in patients with chronic asthma who experienced exercise-induced bronchoconstriction. Methods Design We conducted a randomized, parallel-group study consisting of a 2-week, single-blind placebo baseline period followed by an 8-week, double-blind treatment period with montelukast sodium (10-mg tablet taken orally once in the evening) or inhaled salmeterol (50-g aerosol formulation [2 puffs] taken twice daily). Seventeen clinical study sites participated in the trial. To mask formulation differences, a double-dummy treatment regimen was used. Each patient received one tablet daily (active agent or matching placebo) or one inhaler twice daily (active agent or matching placebo) for both the single-blind and double-blind treatment periods. A computer-generated allocation schedule with a blocking factor of 4 was produced by the statistician. Each center was given a block of allocation numbers that were assigned sequentially to consecutive randomly assigned patients. Spirometric measurements were obtained before and after standardized exercise challenges at the beginning and end of the baseline period, within the first 3 days of the double-blind treatment period, and at weeks 4 and 8 of the treatment period. Additional measurements were physical examination, vital signs, electrocardiography, chest radiography, and laboratory tests (hematology, chemistry profile, and urinalysis). At each visit, all spontaneously reported adverse events were recorded. The protocol was approved by the institutional review board of each site, and written informed consent was obtained from each patient. Inclusion Criteria Male and female patients 15 to 45 years of age with a history of chronic asthma were enrolled. All patients had an FEV1 of at least 65% of the predicted value at rest and a decrease in FEV1 of at least 20% after a standardized exercise challenge on two occasions during the baseline period. All patients had been nonsmokers for at least 1 year and had a smoking history of less than 15 pack-years. Exclusion Criteria Persons who had upper respiratory infection or exacerbation of asthma requiring emergency care within the past month or were hospitalized for asthma in the past 3 months were excluded. Use of oral or inhaled corticosteroids, theophylline, cromolyn sodium, nedocromil, oral -agonist, and long-acting antihistamines was prohibited before and during the study. Use of inhaled albuterol for symptomatic relief of asthma and use of short-acting antihistamines were permitted. Evaluations A standard spirometer (Puritan-Bennett PB100/PB110, Puritan-Bennett, Wilmington, Massachusetts) was used to obtain all spirometric measurements according to American Thoracic Society standard criteria (18). Patients had to have discontinued use of inhaled short-acting -agonists for 6 hours before the visit. Exercise testing was done in the early afternoon near the trough of effect for both drugs according to a method described elsewhere (17). Measurements were obtained 20 and 5 minutes before exercise (prechallenge period). Exercise challenge was performed only if the average FEV1 in the prechallenge period was greater than 65% of predicted; otherwise, the test was rescheduled. Patients exercised on a treadmill while inhaling room temperature, compressed, dry air. During the first test, the speed and gradient of the treadmill were adjusted to achieve 80% to 90% of the patients age-predicted maximum heart rate. The settings were maintained for a total of 6 minutes; the same settings were used for future tests. This level of exercise has been used to quantify the level of bronchoconstriction associated with regular exercise (19). Serial spirometric measurements were obtained at 0, 5, 10, 15, 30, 45, and 60 minutes after exercise (postexercise period). Additional measurements were carried out at 15-minute intervals for up to 90 minutes if the patients FEV1 had not returned to within 5% of the prechallenge value by 60 minutes. If the patients FEV1 did not return to the prechallenge value by 90 minutes after exercise, a rescue dose of inhaled -agonist was administered at the discretion of the study investigator. Statistical Analysis An all-patients-treated analysis, which included patients with a baseline visit and at least one post-randomization visit, was performed. The change from baseline in the maximal percentage decrease in FEV1 after exercise at the end of 8 weeks of treatment was the primary end point. Analysis of variance was used to compare the two treatment groups. The analysis of variance model included terms for treatment, center, and the interaction of treatment and center. Ninety-five percent CIs for within-group means and the difference between groups were constructed to assess the magnitude of the treatment effect. Analysis of variance on the ranked data was used to analyze percentage inhibition for all end points. In the event of early termination of the exercise challenge because of administration of rescue medication, the largest percentage decrease in FEV1 achieved before administration of rescue medication was used in the analysis. Secondary end points were change from baseline for maximal percentage decrease in FEV1 at days 1 to 3 and week 4, the time required after maximal decrease in FEV1 to return within 5% of prechallenge values (time to recovery), and the AUC0-60 min at all visits. The mean of the 20- and 5-minute prechallenge measurements was used as the pre-exercise FEV1 value. If a patient required rescue with inhaled -agonist during the postexercise period, the last recorded FEV1 value was used and carried forward for all subsequent readings and 100 minutes was entered for the end point of time to recovery. The AUC0-60 min was calculated by using the trapezoidal method. If a patients FEV1 did not decrease below 95% of the prechallenge value, the time to recovery was assigned a value of zero. The persistence of effect over time was assessed by using a repeated-measures fixed-effects model with terms for center, treatment group, time, and the interaction of treatment group and time to calculate the rate of change over the treatment period. Persistence of effect was defined as a slope of zero. The magnitude of the slopes for each treatment group was estimated, and 95% CIs were calculated. An overall test of equal slopes between the treatment groups was examined, and a 95% CI on the difference in slopes between treatment groups was provided. The number and percentage of patients requiring rescue medication during or at the end of the exercise test were summarized by treatment group at each time point. In addition, the number and percentage of patients whose decrease in FEV1 from pre-exercise levels was less than 10%, 10% to 20%, 20% to 40%, and greater than 40% were summarized by treatment group for each visit. The overall incidence of adverse events and laboratory abnormalities was assessed by using the Fisher exact test, and within-group changes in the number of laboratory abnormalities were assessed by using the McNemar test. Descriptive statistics were provided by treatment group for patient demographic characteristics, clinical characteristics, and baseline profile. The study was designed with a sample size of 160 patients (80 patients per treatment group) to have 95% power (two-sided test at =0.05) to detect a 7% difference in the mean change in maximal percentage decrease in FEV1 between treatments. All statistical analyses were performed by using SAS software, version 6 (SAS Institute, Inc., Cary, North Carolina). Role of the Funding Source Funding for this trial was provided by Merck & Co., Inc., Whitehouse Station, New Jersey. Personnel from Merck U.S. Human Health, Clinical Development department played a significant role in the design, conduct, and analysis of the trial. The trial was conducted in accordance with guidelines for clinical trials of investigational agents established by U.S. regulatory authorities. Results Pati


Annals of Internal Medicine | 2002

Alendronate Improves Bone Mineral Density in Elderly Women with Osteoporosis Residing in Long-Term Care Facilities: A Randomized, Double-Blind, Placebo-Controlled Trial

Susan L. Greenspan; Diane L. Schneider; Michael R. McClung; Paul Miller; Thomas J. Schnitzer; Randi Bonin; Mary E. Smith; Paul DeLucca; Glenn J. Gormley; Mary E. Melton

Context Although the prevalence of osteoporosis increases with advancing age, most therapeutic studies have focused on younger postmenopausal women. It is not known whether treatment is effective in elderly women. Contribution The efficacy and tolerability of alendronate in elderly female residents of long-term care facilities were tested in a 2-year randomized, double-blind, placebo-controlled study. Alendronate significantly increased bone mineral density and decreased bone turnover when compared with placebo in this elderly population. Side effects did not differ between treatment groups. Implications Elderly women with osteoporosis may benefit from alendronate therapy. The effect of this therapy on incidence of fractures or the optimum length of therapy cannot be determined from this study. The Editors Osteoporosis increases in prevalence with age and is very common in elderly women. The estimated prevalence of osteoporosis in community-dwelling elderly women aged 70 to 79 years is 55%; this rate increases to 73% among those older than 80 years of age (1). The prevalence in elderly female residents of long-term care facilities is even higher. One study (2) reported an 85% prevalence of osteoporosis at nursing home admission. A large survey of nursing home residents found a prevalence of 63.5% for women aged 65 to 74 years and 85.8% for women older than 85 years of age (3). Because most osteoporosis studies have focused on younger postmenopausal women and few studies have evaluated residents of long-term care facilities, we examined the efficacy and tolerability of alendronate for treating osteoporosis in elderly female residents of long-term care facilities. Methods This randomized, double-blind, placebo-controlled, 2-year study was conducted at 25 U.S. centers. Each center recruited ambulatory female residents 65 years of age or older who lived in local long-term care facilities, including skilled-care nursing homes and assisted-living centers, but not facilities that housed independent, community-dwelling elderly persons. Participants were required to have a bone mineral density (BMD) lower than a T-score of 2.0 at the posterioranterior lumbar spine or total hip. Women were excluded for the following: disorders of bone mineralization, 25-hydroxycholecalciferol level less than 25 nmol/L, untreated hyperthyroidism, recent major upper gastrointestinal mucosal erosive disease, or use of bone-active agents. The ethics committee for each site approved the study, and all patients gave informed consent. A total of 832 women consented to participate; the 327 women who met the inclusion and exclusion criteria were enrolled. Patients were randomly assigned (using a computer-generated allocation schedule) to receive alendronate, 10 mg/d (Fosamax, Merck & Co., Inc., Whitehouse Station, New Jersey), or matching placebo for 24 months. Patients also received vitamin D, 400 IU/d. Calcium carbonate (OsCal 500, SmithKlineBeecham Consumer Healthcare, Pittsburgh, Pennsylvania) was given to patients whose daily dietary calcium intake was less than 1500 mg. Bone mineral density of the hip and spine was measured by using dual-energy x-ray absorptiometry (Hologic, Waltham, Massachusetts) and was analyzed by a central quality assurance center (MDM, Waltham, Massachusetts). A central laboratory (Pacific Biometrics, Seattle, Washington) analyzed biochemical markers of bone formation (serum bone-specific alkaline phosphatase, measured by using Tandem-R Ostase kit, Hybritech, Inc., San Diego, California) and bone resorption (urinary N-telopeptide of type I collagen adjusted for urinary creatinine, measured by using Osteomark enzyme-linked immunoassay, Ostex International, Inc., Seattle, Washington). We also collected routine laboratory measures for safety (chemistry, hematology, and urinalysis) and information on the occurrence of clinical adverse experiences and clinical fractures. We used SAS software, version 6.12, for statistical analyses (SAS Institute, Inc., Cary, North Carolina). Baseline demographic characteristics of the two treatment groups were compared by using t-tests (for continuous data) and Fisher exact tests (for categorical data). Analysis of variance was used to compare BMD and biomarker end points between groups. Paired t-tests were used to compare outcomes against baseline values within treatment groups. An intention-to-treat approach was followed in the analyses of BMD end points (for missing data, the last postrandomization observation was carried forward to subsequent time points) and safety data. A per protocol approach was used for analysis of biomarkers. Sensitivity analyses were done separately for BMD and biomarker end points by using mixed models that accounted for the correlation between all available measurements of BMD (up to 12 for each participant) and biomarkers (up to 8 for each participant). Data points were not carried forward in these analyses. Treatment comparisons were made by using the appropriate contrasts from the interaction term for treatment test site visit (using the MIXED procedure [4]). Employees of Merck & Co., Inc., contributed to the protocol design, recruitment of investigators, and collection and analysis of data. Merck & Co., Inc., had committed to submitting the data for publication regardless of study outcome. Results Most patients were residents of continuing-care retirement communities (68%); others resided in retirement communities (12%), congregate-care facilities (8%), and other types of facilities (12%), including skilled-care nursing homes and residential-care facilities. The characteristics of each treatment group were similar at baseline. The mean age was 78.5 years (range, 65 to 91 years). Most patients (97%) were white. The mean BMD T-scores at the hip and spine ranged from 3.5 to 2.4. Fifty-five percent of patients had a history of fracture, 19% reported a history of upper gastrointestinal disorder at baseline, and 64% used either aspirin or a nonsteroidal anti-inflammatory drug during the study. Increases in BMD at all sites were significantly greater in the alendronate group than in the placebo group at all time points measured (Figure 1). Compared with placebo, alendronate produced significantly greater increases in BMD (24-month differences: posterioranterior spine, 4.4% [95% CI, 3.3% to 5.5%]; lateral lumbar spine, 5.7% [CI, 3.9% to 7.5%]; femoral neck, 3.4% [CI, 2.3% to 4.4%]; hip trochanter, 4.7% [CI, 3.4% to 6.0%]). The largest increases from baseline with alendronate occurred at the lateral lumbar spine (7.4% [CI, 6.1% to 8.7%]; absolute change, 0.039 g/cm2 [CI, 0.032 to 0.046 g/cm2]; P < 0.001) and the posterioranterior lumbar spine (6.3% [CI, 5.5% to 7.0%]; absolute change, 0.047 g/cm2 [CI, 0.042 to 0.053 g/cm2]; P < 0.001) at 24 months. Decreases in levels of bone-specific alkaline phosphatase and urinary N-telopeptide adjusted for urinary creatinine were significantly greater in alendronate recipients than in placebo recipients at all time points measured (P < 0.001) (Figure 2). The mixed-model analyses yielded similar results. Figure 1. Changes in bone mineral density ( BMD ). circles squares P P P P P Figure 2. Changes in biochemical markers of bone turnover. N circles squares P P P P P N More patients receiving placebo experienced a fracture during the study (11% of placebo recipients vs. 8% of alendronate recipients), but this finding was not statistically significant. Hip fracture occurred in 4 patients receiving placebo and 2 patients receiving alendronate. Overall, 13 fractures were reported in 13 patients receiving alendronate and 28 fractures were reported in 18 patients receiving placebo. In each group, the percentage of patients reporting any clinical adverse experience was the same (93%). The percentage of patients reporting any upper gastrointestinal adverse event did not significantly differ between groups (33% in the alendronate group vs. 35% in the placebo group), nor did the percentage of patients reporting serious upper gastrointestinal adverse events (0.6% for alendronate vs. 1.9% for placebo). Discussion In this study, alendronate significantly improved both hip and spine BMD in elderly female residents of long-term care facilities. These improvements were associated with the expected decreases in bone resorption. In addition, alendronate was generally well tolerated in this elderly cohort. Although several studies have documented improvements in BMD among community-dwelling postmenopausal women treated with alendronate, our study is the first to examine the efficacy of 10 mg of alendronate per day in elderly residents of long-term care facilities. Bone and colleagues (5) studied community-dwelling elderly women with osteoporosis (mean age, 71 years) and found a dose-related response of BMD with use of alendronate dosages as high as 5 mg/d. The improvements in BMD in our study indicate that even in elderly women, BMD significantly increases with alendronate. Furthermore, a woman who reaches age 65 years can on average expect to live to age 84, and women who survive to age 85 can expect to live to age 92 (6). Therefore, it would be clinically relevant and appropriate to treat these patients, even though they are elderly, with a medication that reduces fracture rates in 1 to 3 years. No bone loss occurred in the placebo group. This may have been due to adequate calcium intake and the use of vitamin D. Previous studies have shown that bone loss slows with calcium supplementation (7, 8) and that hip BMD increases and fracture rate decreases with use of vitamin D and calcium supplementation in elderly female residents of long-term care facilities (9). Our study showed that while use of calcium and vitamin D alone may help slow bone loss, additional therapy is required to substantially increase BMD. Establishing safety is important when any medication is given to elderly patients in long-term care settings. Previous clinical trials with


Journal of the American College of Cardiology | 2011

Effects of the Adenosine A1 Receptor Antagonist Rolofylline on Renal Function in Patients With Acute Heart Failure and Renal Dysfunction: Results From PROTECT (Placebo-Controlled Randomized Study of the Selective A1 Adenosine Receptor Antagonist Rolofylline for Patients Hospitalized With Acute Decompensated Heart Failure and Volume Overload to Assess Treatment Effect on Congestion and Renal Function)

Adriaan A. Voors; Howard C. Dittrich; Barry M. Massie; Paul DeLucca; George A. Mansoor; Marco Metra; Gad Cotter; Beth Davison Weatherley; Piotr Ponikowski; John R. Teerlink; John G.F. Cleland; Christopher M. O'Connor; Michael M. Givertz

OBJECTIVES This study sought to assess the effects of rolofylline on renal function in patients with acute heart failure (AHF) and renal dysfunction randomized in PROTECT (Placebo-Controlled Randomized Study of the Selective A(1) Adenosine Receptor Antagonist Rolofylline for Patients Hospitalized With Acute Decompensated Heart Failure and Volume Overload to Assess Treatment Effect on Congestion and Renal Function). BACKGROUND Small studies have indicated that adenosine A(1) receptor antagonists enhance diuresis and may improve renal function in patients with chronic heart failure or AHF. METHODS A total of 2,033 patients with AHF, volume overload, estimated creatinine clearance between 20 and 80 ml/min, and elevated natriuretic peptide levels were randomized (2:1) within 24 h of hospital presentation to rolofylline 30 mg/day or intravenous placebo for up to 3 days. Creatinine was measured daily until discharge or day 7 and on day 14. Persistent worsening renal function was defined as an increase in serum creatinine ≥0.3 mg/dl at both days 7 and 14, or initiation of hemofiltration or dialysis or death by day 7. RESULTS At baseline, mean ± SD estimated creatinine clearance was 51.0 ± 20.5 ml/min in the placebo group and 50.4 ± 20.0 ml/min in the rolofylline group. Changes in creatinine and estimated creatinine clearance were similar between placebo- and rolofylline-treated patients during hospitalization and at day 14. After 4 days, mean body weight was reduced by 2.6 and 3.0 kg in placebo and rolofylline patients, respectively (p = 0.005). Persistent worsening renal function occurred in 13.7% of the placebo group and 15.0% of the rolofylline group (odds ratio vs. placebo: 1.11 [95% confidence interval: 0.85 to 1.46]; p = 0.44). CONCLUSIONS In this large, phase III clinical trial, the adenosine A(1) receptor antagonist rolofylline did not prevent persistent worsening renal function in AHF patients with volume overload and renal dysfunction. (A Study of the Selective A1 Adenosine Receptor Antagonist KW-3902 for Patients Hospitalized With Acute HF and Volume Overload to Assess Treatment Effect on Congestion and Renal Function [PROTECT-1], NCT00328692; and [PROTECT-2], NCT00354458).


Journal of Cardiac Failure | 2010

Design and Rationale of the PROTECT Study: A Placebo-controlled Randomized Study of the Selective A1 Adenosine Receptor Antagonist Rolofylline for Patients Hospitalized With Acute Decompensated Heart Failure and Volume Overload to Assess Treatment Effect on Congestion and Renal Function

Beth Davison Weatherley; Gad Cotter; Howard C. Dittrich; Paul DeLucca; George A. Mansoor; Daniel M. Bloomfield; Piotr Ponikowski; Christopher M. O'Connor; Marco Metra; Barry M. Massie

BACKGROUND Current treatment for acute decompensated heart failure (ADHF) is associated with incomplete resolution of symptoms and signs, recurrent symptoms of heart failure in-hospital and after discharge and high mortality. Studies have consistently demonstrated an association between worsening renal function in ADHF and adverse outcomes. Adenosine A(1) receptor antagonists, such as rolofylline, appear in preliminary studies to produce potentially beneficial effects on natriuresis, diuresis, renal blood flow, and glomerular filtration rate. In a previous dose-finding study, rolofylline 30 mg intravenously daily for 3 days was associated with symptom improvement, less worsening of renal function, and trends toward lower 60-day rates of death or readmission for cardiovascular or renal causes. METHODS AND RESULTS This manuscript describes the rationale underlying the design of the phase 3 PROTECT (Placebo-controlled Randomized study of the selective A(1) adenosine receptor antagonist rolofylline for patients hospitalized with acute heart failure and volume Overload to assess Treatment Effect on Congestion and renal funcTion) trial. CONCLUSION Rolofylline 30 mg or matching placebo was given intravenously as a 4-hour continuous infusion on 3 consecutive days and the hospital course was assessed by measurements dyspnea, clinical status, renal function, and subsequent morbidity and mortality in a large population of patients with ADHF with renal impairment.


Journal of the American College of Cardiology | 2011

Effects of the adenosine A1 receptor antagonist rolofylline on renal function in patients with acute heart failure and renal dysfunction: results from PROTECT (Placebo-Controlled Randomized Study of the Selective Adenosine A1 Receptor Antagonist Rolofylline for Patients Hospitalized with Acute Decompensated Heart Failure and Volume Overload to Assess Treatment Effect on Congestion and Renal Function).

A.A. Voors; Howard C. Dittrich; Barry M. Massie; Paul DeLucca; George A. Mansoor; Marco Metra; G. Cotter; Beth Davison Weatherley; Piotr Ponikowski; Teerlink; John G.F. Cleland; Christopher M. O'Connor; Michael M. Givertz

OBJECTIVES This study sought to assess the effects of rolofylline on renal function in patients with acute heart failure (AHF) and renal dysfunction randomized in PROTECT (Placebo-Controlled Randomized Study of the Selective A(1) Adenosine Receptor Antagonist Rolofylline for Patients Hospitalized With Acute Decompensated Heart Failure and Volume Overload to Assess Treatment Effect on Congestion and Renal Function). BACKGROUND Small studies have indicated that adenosine A(1) receptor antagonists enhance diuresis and may improve renal function in patients with chronic heart failure or AHF. METHODS A total of 2,033 patients with AHF, volume overload, estimated creatinine clearance between 20 and 80 ml/min, and elevated natriuretic peptide levels were randomized (2:1) within 24 h of hospital presentation to rolofylline 30 mg/day or intravenous placebo for up to 3 days. Creatinine was measured daily until discharge or day 7 and on day 14. Persistent worsening renal function was defined as an increase in serum creatinine ≥0.3 mg/dl at both days 7 and 14, or initiation of hemofiltration or dialysis or death by day 7. RESULTS At baseline, mean ± SD estimated creatinine clearance was 51.0 ± 20.5 ml/min in the placebo group and 50.4 ± 20.0 ml/min in the rolofylline group. Changes in creatinine and estimated creatinine clearance were similar between placebo- and rolofylline-treated patients during hospitalization and at day 14. After 4 days, mean body weight was reduced by 2.6 and 3.0 kg in placebo and rolofylline patients, respectively (p = 0.005). Persistent worsening renal function occurred in 13.7% of the placebo group and 15.0% of the rolofylline group (odds ratio vs. placebo: 1.11 [95% confidence interval: 0.85 to 1.46]; p = 0.44). CONCLUSIONS In this large, phase III clinical trial, the adenosine A(1) receptor antagonist rolofylline did not prevent persistent worsening renal function in AHF patients with volume overload and renal dysfunction. (A Study of the Selective A1 Adenosine Receptor Antagonist KW-3902 for Patients Hospitalized With Acute HF and Volume Overload to Assess Treatment Effect on Congestion and Renal Function [PROTECT-1], NCT00328692; and [PROTECT-2], NCT00354458).


American Heart Journal | 2003

Effect on high-density lipoprotein cholesterol of maximum dose simvastatin and atorvastatin in patients with hypercholesterolemia: Results of the Comparative HDL Efficacy and Safety Study (CHESS)☆

Christie M. Ballantyne; Michael A. Blazing; Donald B. Hunninghake; Michael Davidson; Zhong Yuan; Paul DeLucca; Karen E. Ramsey; Carolyn M. Hustad; Joanne Palmisano

BACKGROUND Previous studies have shown that effects on high-density lipoprotein cholesterol (HDL-C) may differ among statins. METHODS A multicenter, randomized, double-blind, parallel-dose study was conducted in 917 hypercholesterolemic patients to compare the efficacy of 80 mg/d simvastatin versus 80 mg/d atorvastatin on HDL-C and apolipoprotein (apo) A-I for 24 weeks. Efficacy was assessed as the means of weeks 6 and 12 and weeks 18 and 24. Prespecified subgroups analyzed were patients with low HDL-C levels and with the metabolic syndrome. RESULTS Simvastatin increased HDL-C and apo A-I values significantly more than did atorvastatin for the mean of weeks 6 and 12 (8.9% vs 3.6% and 4.9% vs -0.9%, respectively) and the mean of weeks 18 and 24 (8.3% vs 4.2% and 3.7% vs -1.4%). These differences were observed across both baseline HDL-C subgroups (<40 mg/dL, > or =40 mg/dL) and in patients with the metabolic syndrome. Low-density lipoprotein cholesterol and triglyceride reductions were greater with atorvastatin. Consecutive elevations >3x the upper limit of normal in alanine aminotransferase (ALT) and/or aspartate aminotransferase (AST) occurred in significantly fewer patients treated with simvastatin than with atorvastatin (2/453 [0.4%] vs 13/464 [2.8%]), with most elevations observed in women taking atorvastatin (11/209 [5.3%] vs 1/199 [0.5%] for simvastatin). CONCLUSIONS Simvastatin (80 mg) increased HDL-C and apo A-I significantly more than did atorvastatin (80 mg) in patients with hypercholesterolemia. This advantage was observed regardless of HDL-C level at baseline or the presence of the metabolic syndrome. Significantly fewer consecutive elevations >3x the upper limit of normal in ALT and/or AST occurred in patients receiving simvastatin.

Collaboration


Dive into the Paul DeLucca's collaboration.

Top Co-Authors

Avatar

Barry M. Massie

United States Department of Health and Human Services

View shared research outputs
Top Co-Authors

Avatar

Howard C. Dittrich

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Piotr Ponikowski

Wrocław Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John G.F. Cleland

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge