Network


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

Hotspot


Dive into the research topics where Dave L. Dixon is active.

Publication


Featured researches published by Dave L. Dixon.


Pharmacotherapy | 2015

Collaborative Drug Therapy Management and Comprehensive Medication Management―2015

Sarah McBane; Anna Legreid Dopp; Andrew M. Abe; Elizabeth A. Chester; Dave L. Dixon; Michaelia Dunn; Melissa D. Johnson; Sarah J. Nigro; Tracie Rothrock-Christian; Amy H. Schwartz; Kim Thrasher; Scot Walker

The American College of Clinical Pharmacy (ACCP) previously published position statements on collaborative drug therapy management (CDTM) in 1997 and 2003. Since 2003, significant federal and state legislation addressing CDTM has evolved and expanded throughout the United States. CDTM is well suited to facilitate the delivery of comprehensive medication management (CMM) by clinical pharmacists. CMM, defined by ACCP as a core component of the standards of practice for clinical pharmacists, is designed to optimize medication‐related outcomes in collaborative practice environments. New models of care delivery emphasize patient‐centered, team‐based care and increasingly link payment to the achievement of positive economic, clinical, and humanistic outcomes. Hence clinical pharmacists practicing under CDTM agreements or through other privileging processes are well positioned to provide CMM. The economic value of clinical pharmacists in team‐based settings is well documented. However, patient access to CMM remains limited due to lack of payer recognition of the value of clinical pharmacists in collaborative care settings and current health care payment policy. Therefore, the clinical pharmacy discipline must continue to establish and expand its use of CDTM agreements and other collaborative privileging mechanisms to provide CMM. Continued growth in the provision of CMM by appropriately qualified clinical pharmacists in collaborative practice settings will enhance recognition of their positive impact on medication‐related outcomes.


Journal of Cardiovascular Pharmacology | 2016

Interleukin-1 Blockade In Acute Decompensated Heart Failure: A Randomized, Double-Blinded, Placebo-Controlled Pilot Study.

Benjamin W. Van Tassell; Nayef Abouzaki; Claudia Oddi Erdle; Salvatore Carbone; Cory Trankle; Ryan Melchior; Jeremy Turlington; Clint Thurber; Sanah Christopher; Dave L. Dixon; Daniel Taylor Fronk; Christopher Scott Thomas; Scott W. Rose; Leo F. Buckley; Charles A. Dinarello; Giuseppe Biondi-Zoccai; Antonio Abbate

Background: Heart failure is an inflammatory disease. Patients with acute decompensated heart failure (ADHF) exhibit significant inflammatory activity on admission. We hypothesized that Interleukin-1 blockade, with anakinra (Kineret, Swedish Orphan Biovitrum), would quench the acute inflammatory response in patients with ADHF. Methods: We randomized 30 patients with ADHF, reduced left ventricular ejection fraction (<40%), and elevated C reactive protein (CRP) levels (≥5 mg/L) to either anakinra 100 mg twice daily for 3 days followed by once daily for 11 days or matching placebo, in a 1:1 double blinded fashion. We measured daily CRP plasma levels using a high-sensitivity assay during hospitalization and then again at 14 days and evaluated the area-under-the-curve and interval changes (delta). Results: Treatment with anakinra was well tolerated. At 72 hours, anakinra reduced CRP by 61% versus baseline, compared with a 6% reduction among patients receiving placebo (P = 0.004 anakinra vs. placebo). Conclusions: Interleukin-1 blockade with anakinra reduces the systemic inflammatory response in patients with ADHF. Further studies are warranted to determine whether this anti-inflammatory effect translates into improved clinical outcomes.


Diabetes Care | 2017

Intensive Versus Standard Blood Pressure Control in SPRINT-Eligible Participants of the ACCORD-BP Trial

Leo F. Buckley; Dave L. Dixon; George F. Wohlford; Dayanjan S. Wijesinghe; William L. Baker; Benjamin W. Van Tassell

OBJECTIVE We sought to determine the effect of intensive blood pressure (BP) control on cardiovascular outcomes in participants with type 2 diabetes mellitus (T2DM) and additional risk factors for cardiovascular disease (CVD). RESEARCH DESIGN AND METHODS This study was a post hoc, multivariate, subgroup analysis of ACCORD-BP (Action to Control Cardiovascular Risk in Diabetes Blood Pressure) participants. Participants were eligible for the analysis if they were in the standard glucose control arm of ACCORD-BP and also had the additional CVD risk factors required for SPRINT (Systolic Blood Pressure Intervention Trial) eligibility. We used a Cox proportional hazards regression model to compare the effect of intensive versus standard BP control on CVD outcomes. The “SPRINT-eligible” ACCORD-BP participants were pooled with SPRINT participants to determine whether the effects of intensive BP control interacted with T2DM. RESULTS The mean baseline Framingham 10-year CVD risk scores were 14.5% and 14.8%, respectively, in the intensive and standard BP control groups. The mean achieved systolic BP values were 120 and 134 mmHg in the intensive and standard BP control groups (P < 0.001). Intensive BP control reduced the composite of CVD death, nonfatal myocardial infarction (MI), nonfatal stroke, any revascularization, and heart failure (hazard ratio 0.79; 95% CI 0.65–0.96; P = 0.02). Intensive BP control also reduced CVD death, nonfatal MI, and nonfatal stroke (hazard ratio 0.69; 95% CI 0.51–0.93; P = 0.01). Treatment-related adverse events occurred more frequently in participants receiving intensive BP control (4.1% vs. 2.1%; P = 0.003). The effect of intensive BP control on CVD outcomes did not differ between patients with and without T2DM (P > 0.62). CONCLUSIONS Intensive BP control reduced CVD outcomes in a cohort of participants with T2DM and additional CVD risk factors.


Journal of Clinical Lipidology | 2016

A review of PCSK9 inhibition and its effects beyond LDL receptors

Dave L. Dixon; Cory Trankle; Leo F. Buckley; Eric D. Parod; Salvatore Carbone; Benjamin W. Van Tassell; Antonio Abbate

Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays an integral role in the degradation of low-density lipoprotein receptors (LDL-R), making it an intriguing target for emerging pharmacotherapy. Two PCSK9 inhibitors, alirocumab and evolocumab, have been approved and are available in the United States and European Union. However, much of the PCSK9 story remains to be told. The pipeline for additional pharmacotherapy options is rich with several compounds under development, using alternative strategies for inhibiting PCSK9. Perhaps, more intriguing is the interaction between PCSK9 and non-LDL-R targets, including mediators of inflammation and immunological processes, which remain under intense investigation. This review will discuss the currently available PCSK9 inhibitors, the development of novel approaches to PCSK9 modulation, and the potential non-LDL-R-mediated effects of PCSK9 inhibition.


JACC: Basic to Translational Science | 2017

Dietary Fat, Sugar Consumption, and Cardiorespiratory Fitness in Patients With Heart Failure With Preserved Ejection Fraction

Salvatore Carbone; Justin M. Canada; Leo F. Buckley; Cory Trankle; Hayley Billingsley; Dave L. Dixon; Adolfo G Mauro; Sofanit Dessie; Dinesh Kadariya; Eleonora Mezzaroma; Raffaella Buzzetti; Ross Arena; Benjamin W. Van Tassell; Stefano Toldo; Antonio Abbate

Visual Abstract


Journal of the American College of Cardiology | 2016

Obesity Contributes to Exercise Intolerance in Heart Failure With Preserved Ejection Fraction

Salvatore Carbone; Justin M. Canada; Leo F. Buckley; Cory Trankle; Dave L. Dixon; Raffaella Buzzetti; Ross Arena; Benjamin W. Van Tassell; Antonio Abbate

Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome of exertional breathlessness and/or fatigue caused by impaired cardiac function, preserved left ventricular ejection fraction, and impaired diastolic function [(1)][1]. The incidence and prevalence of HFpEF in the United


Journal of the American Heart Association | 2017

Effects of Sodium-Glucose Cotransporter 2 Inhibitors on 24-Hour Ambulatory Blood Pressure: A Systematic Review and Meta-Analysis

William L. Baker; Leo F. Buckley; Michael S. Kelly; John D. Bucheit; Eric D. Parod; Roy E. Brown; Salvatore Carbone; Antonio Abbate; Dave L. Dixon

Background Sodium‐glucose cotransporter 2 (SGLT2) inhibitors are a novel class of antihyperglycemic agents that improve glycemic control by increasing glycosuria. Additional benefits beyond glucose lowering include significant improvements in seated clinic blood pressure (BP), partly attributed to their diuretic‐like actions. Less known are the effects of this class on 24‐hour ambulatory BP, which is a better predictor of cardiovascular risk than seated clinic BP. Methods and Results We performed a meta‐analysis of randomized, double‐blind, placebo‐controlled trials to investigate the effects of SGLT2 inhibitors on 24‐hour ambulatory BP. We searched all studies published before August 17, 2016, which reported 24‐hour ambulatory BP data. Mean differences in 24‐hour BP, daytime BP, and nighttime BP were calculated by a random‐effects model. SGLT2 inhibitors significantly reduce 24‐hour ambulatory systolic and diastolic BP by −3.76 mm Hg (95% CI, −4.23 to −2.34; I2=0.99) and −1.83 mm Hg (95% CI, −2.35 to −1.31; I2=0.76), respectively. Significant reductions in daytime and nighttime systolic and diastolic BP were also found. No association between baseline BP or change in body weight were observed. Conclusions This meta‐analysis shows that the reduction in 24‐hour ambulatory BP observed with SGLT2 inhibitors is a class effect. The diurnal effect of SGLT2 inhibitors on 24‐hour ambulatory BP may contribute to their favorable effects on cardiovascular outcomes.


Clinical Cardiology | 2017

Interleukin-1 blockade in heart failure with preserved ejection fraction: rationale and design of the Diastolic Heart Failure Anakinra Response Trial 2 (D-HART2)

Benjamin W. Van Tassell; Leo F. Buckley; Salvatore Carbone; Cory Trankle; Justin M. Canada; Dave L. Dixon; Nayef Abouzaki; Claudia Oddi-Erdle; Giuseppe Biondi-Zoccai; Ross Arena; Antonio Abbate

Heart failure with preserved ejection fraction (HFpEF) now accounts for the majority of confirmed HF cases in the United States. However, there are no highly effective evidence‐based treatments currently available for these patients. Inflammation correlates positively with adverse outcomes in HF patients. Interleukin (IL)‐1, a prototypical inflammatory cytokine, has been implicated as a driver of diastolic dysfunction in preclinical animal models and a pilot clinical trial. The Diastolic Heart Failure Anakinra Response Trial 2 (D‐HART2) is a phase 2, 2:1 randomized, double‐blind, placebo‐controlled clinical trial that will test the hypothesis that IL‐1 blockade with anakinra (recombinant human IL‐1 receptor antagonist) improves (1) cardiorespiratory fitness, (2) objective evidence of diastolic dysfunction, and (3) elevated inflammation in patients with HFpEF (http://www.ClinicalTrials.gov NCT02173548). The co–primary endpoints will be placebo‐corrected interval changes in peak oxygen consumption and ventilatory efficiency at week 12. In addition, secondary and exploratory analyses will investigate the effects of IL‐1 blockade on cardiac structure and function, systemic inflammation, endothelial function, quality of life, body composition, nutritional status, and clinical outcomes. The D‐HART2 clinical trial will add to the growing body of evidence on the role of inflammation in cardiovascular disease, specifically focusing on patients with HFpEF.


Circulation-heart Failure | 2017

Interleukin-1 Blockade in Recently Decompensated Systolic Heart Failure: Results From REDHART (Recently Decompensated Heart Failure Anakinra Response Trial)

Benjamin W. Van Tassell; Justin M. Canada; Salvatore Carbone; Cory Trankle; Leo F. Buckley; Claudia Oddi Erdle; Nayef Abouzaki; Dave L. Dixon; Dinesh Kadariya; Sanah Christopher; Aaron Schatz; Jessica Regan; Michele Viscusi; Marco Del Buono; Ryan Melchior; Pranav Mankad; Juan Lu; Robin Sculthorpe; Giuseppe Biondi-Zoccai; Edward J. Lesnefsky; Ross Arena; Antonio Abbate

Background An enhanced inflammatory response predicts worse outcomes in heart failure (HF). We hypothesized that administration of IL-1 (interleukin-1) receptor antagonist (anakinra) could inhibit the inflammatory response and improve peak aerobic exercise capacity in patients with recently decompensated systolic HF. Methods and Results We randomly assigned 60 patients with reduced left ventricular ejection fraction (<50%) and elevated C-reactive protein levels (>2 mg/L), within 14 days of hospital discharge, to daily subcutaneous injections with anakinra 100 mg for 2 weeks, 12 weeks, or placebo. Patients underwent measurement of peak oxygen consumption (VO2 [mL/kg per minute]) and ventilatory efficiency (the VE/VCO2 slope). Treatment with anakinra did not affect peak VO2 or VE/VCO2 slope at 2 weeks. At 12 weeks, patients continued on anakinra showed an improvement in peak VO2 from 14.5 (10.5–16.6) mL/kg per minute to 16.1 (13.2–18.6) mL/kg per minute (P=0.009 for within-group changes), whereas no significant changes occurred within the anakinra 2-week or placebo groups. The between-groups differences, however, were not statistically significant. The incidence of death or rehospitalization for HF at 24 weeks was 6%, 31%, and 30%, in the anakinra 12-week, anakinra 2-week, and placebo groups, respectively (log-rank test P=0.10). Conclusions No change in peak VO2 occurred at 2 weeks in patients with recently decompensated systolic HF treated with anakinra, whereas an improvement was seen in those patients in whom anakinra was continued for 12 weeks. Additional larger studies are needed to validate the effects of prolonged anakinra on peak VO2 and rehospitalization for HF. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT01936909.


Journal of Cardiovascular Nursing | 2014

Lomitapide and mipomersen: novel lipid-lowering agents for the management of familial hypercholesterolemia.

Dave L. Dixon; Evan M. Sisson; Michael V. Butler; Ashley Higbea; Brendan Muoio; Brandy Turner

Background:Familial hypercholesterolemia (FH) is an autosomal dominant disorder caused primarily by mutations in the low-density lipoprotein receptor gene. Familial hypercholesterolemia is characterized by exceedingly high levels of low-density lipoprotein cholesterol (LDL-C) and subsequent premature coronary heart disease. Homozygous FH (HoFH) is less prevalent, but more severe, than heterozygous FH. Current treatment options include dietary therapy, lipid-lowering agents (eg, statins), and/or LDL-C apheresis. Purpose:Despite the available treatment options, patients with FH rarely attain treatment goals. This review will focus on 2 novel agents, lomitapide and mipomersen, with recently approved US Food and Drug Administration (FDA) labeling for use in patients with HoFH. Conclusions:Lomitapide and mipomersen are 2 agents with novel mechanisms of action and the ability to significantly lower LDL-C, apolipoprotein B, and non–high-density lipoprotein cholesterol levels. A black box warning exists for lomitapide and mipomersen regarding the risk for transaminase elevations and hepatic steatosis. Furthermore, these agents are currently restricted for use only in patients with HoFH and have been required by the FDA to participate in a Risk Evaluation and Mitigation Strategy. Clinical Implications:These new agents offer additional treatment options for clinicians managing patients with HoFH, but it remains uncertain whether lomitapide and mipomersen will gain FDA approval for use in patients with heterozygous FH or in the general population. Cost and concern for the risk for hepatotoxicity will remain limiting factors to these agents being more widely used.

Collaboration


Dive into the Dave L. Dixon's collaboration.

Top Co-Authors

Avatar

Benjamin W. Van Tassell

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Antonio Abbate

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Leo F. Buckley

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Salvatore Carbone

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Cory Trankle

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Justin M. Canada

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Ross Arena

American Physical Therapy Association

View shared research outputs
Top Co-Authors

Avatar

Evan M. Sisson

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Dinesh Kadariya

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

George F. Wohlford

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge