Network


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

Hotspot


Dive into the research topics where Robert DiCenzo is active.

Publication


Featured researches published by Robert DiCenzo.


Clinical Pharmacokinectics | 2001

Clinical Pharmacokinetics of Non-Nucleoside Reverse Transcriptase Inhibitors

Patrick F. Smith; Robert DiCenzo; Gene D. Morse

Non-nucleoside reverse transcriptase inhibitors (NNRTIs) are a diverse group of compounds that induce allosteric changes in the human immunodeficiency virus type 1 (HIV-1) reverse transcriptase, thus rendering the enzyme incapable of converting viral RNA to DNA. Unlike nucleoside analogue inhibitors of reverse transcriptase, NNRTIs do not require sequential phosphorylation to elicit antiretroviral activity.There are currently 3 approved NNRTIs: nevirapine, delavirdine and efavirenz. Although possessing a common mechanism of action, these agents can be differentiated by both molecular and pharmacokinetic characteristics. Each of the NNRTIs is metabolised to some degree by the cytochrome P450 (CYP) system of enzymes, making them prone to clinically significant drug interactions. In addition, they elicit variable effects on other medications, acting as either inducers or inhibitors of drugs metabolised by CYP. These drug interactions are an important consideration in the clinical use of these agents as a part of combination antiretroviral therapy. Additional factors such as the influence of food and pH on oral absorption, and protein binding, must also be considered.


Biopharmaceutics & Drug Disposition | 2008

Quercetin pharmacokinetics in humans

Young Jin Moon; Liang Wang; Robert DiCenzo; Marilyn E. Morris

The purpose of this study was to examine the pharmacokinetics of quercetin aglycone as well as its conjugated metabolites and to develop a population pharmacokinetic model for quercetin that incorporates enterohepatic recirculation. The stability of quercetin in different matrices at various temperatures and pH, and the quercetin content of six capsules of the herbal preparation Quercetin‐500 Plus® were determined by HPLC. Subjects received quercetin 500 mg three times daily and blood and urine samples were obtained. The concentration of quercetin aglycone and conjugated metabolites were assayed using a liquid chromatography‐tandem mass spectrometry assay. Pharmacokinetic parameters were determined using noncompartmental analysis with WinNonlin. A population compartment model incorporating input from the gallbladder was developed to account for the enterohepatic recirculation observed with quercetin. The oral clearance (CL/F) was high (3.5 × 104l/h) with an average terminal half‐life of 3.5 h for quercetin. The plasma concentration versus time curves exhibited re‐entry peaks. A one‐compartment model that included enterohepatic recirculation best described the plasma data. This represents the first comprehensive evaluation of the pharmacokinetics and enterohepatic recirculation of quercetin in humans. Population pharmacokinetic models adapted for enterohepatic recirculation allowed an assessment of the magnitude and frequency of the enterohepatic recirculation process. Copyright


Pharmacotherapy | 2003

Coadministration of milk thistle and indinavir in healthy subjects.

Robert DiCenzo; Mark J. Shelton; Kelly Jordan; Christine Koval; Alan Forrest; Richard C. Reichman; Gene D. Morse

Study Objective. To determine if milk thistle (silymarin) alters the pharmacokinetics of indinavir.


Antimicrobial Agents and Chemotherapy | 2004

Effects of Valproic Acid Coadministration on Plasma Efavirenz and Lopinavir Concentrations in Human Immunodeficiency Virus-Infected Adults

Robert DiCenzo; Derick R. Peterson; Kim Cruttenden; Gene D. Morse; Garret Riggs; Harris A. Gelbard; Giovanni Schifitto

ABSTRACT Valproic acid (VPA) has the potential to benefit patients suffering from human immunodeficiency virus (HIV)-associated cognitive impairment. The purpose of this study was to determine if VPA affects the plasma concentration of efavirenz (EFV) or lopinavir. HIV type 1 (HIV-1)-infected patients receiving EFV or lopinavir-ritonavir (LPV/r) had 9 or 10 blood samples drawn over 8 to 24 h of a dosing interval at steady state before and after receiving 250 mg of VPA twice daily for 7 days. VPA blood samples drawn before (C0) and 8 h after the morning dose (8 h) were compared to blood samples from a group of HIV-1-infected subjects who were taking either combined nucleoside reverse transcriptase inhibitors alone or had discontinued antiretroviral therapy. Pharmacokinetic parameters were calculated by noncompartmental analysis, and tests of bioequivalence were based on 90% confidence intervals (CIs) for ratios or differences. The geometric mean ratio (GMR) (90% CI) of the areas under the concentration-time curve from 0 to 24 h (AUC0-24s) of EFV (n = 11) with and without VPA was 1.00 (0.85, 1.17). The GMR (90% CI) of the AUC0-8s of LPV (n = 8) with and without VPA was 1.38 (0.98, 1.94). The differences (90% CI) in mean C0 and 8-h VPA concentrations versus the control (n = 11) were −1.0 (−9.4, 7.4) μg/ml and −2.1 (−11.1, 6.9) μg/ml for EFV (n = 10) and −5.0 (−13.2, 3.3) μg/ml and −6.7 (−17.6, 4.2) μg/ml for LPV/r (n = 11), respectively. EFV administration alone is bioequivalent to EFV and VPA coadministration. LPV concentrations tended to be higher when the drug was combined with VPA. Results of VPA comparisons fail to raise concern that coadministration with EFV or LPV/r will significantly influence trough concentrations of VPA.


Pharmacotherapy | 2003

A Gentamicin Pharmacokinetic Population Model and Once-Daily Dosing Algorithm for Neonates

Robert DiCenzo; Alan Forrest; Judianne C. Slish; Carol Cole; Ronnie Guillet

Study Objective. To develop a gentamicin pharmacokinetic population model and once‐daily dosing algorithm for neonates younger than 10 days.


Expert Opinion on Drug Metabolism & Toxicology | 2005

Pharmacokinetic drug interactions with non-nucleoside reverse transcriptase inhibitors

Qing Ma; Olanrewaju O. Okusanya; Patrick F. Smith; Robert DiCenzo; Judianne C. Slish; Linda M. Catanzaro; Alan Forrest; Gene D. Morse

Non-nucleoside reverse transcriptase inhibitors (NNRTIs) are a diverse group of compounds that inhibit HIV Type 1 reverse transcriptase. Although possessing a common mechanism of action, the approved NNRTIs, delavirdine, efavirenz and nevirapine, differ in structural and pharmacokinetic characteristics. Each of the NNRTIs undergoes biotransformation by the cytochrome P450 (CYP) enzyme system, thus making them prone to clinically significant drug interactions when combined with other antiretrovirals. In addition, they interact with other concurrent medications and complementary/alternative medicines, acting as either inducers or inhibitors of drug-metabolising CYP enzymes. These drug interactions become an important consideration in the clinical use of these agents when designing combination regimens, as recommended by current guidelines. This review provides an updated summary of pharmacokinetic interactions with NNRTIs.


Clinical Pharmacokinectics | 2008

Antiretroviral therapy : pharmacokinetic considerations in patients with renal or hepatic impairment.

Sarah M. Mccabe; Qing Ma; Judianne C. Slish; Linda M. Catanzaro; Neha Sheth; Robert DiCenzo; Gene D. Morse

Hepatic and renal insufficiency due to co-infection, alcoholism, diabetes mellitus, family history, adverse effects of antiretrovirals and other factors are commonly seen in HIV-infected patients. Therefore, the use of antiretrovirals in this patient setting requires attention to the pharmacokinetic issues that clinicians must consider when prescribing highly active antiretroviral therapy for these patients. This review summarizes the current knowledge of the use of antiretrovirals in patients with hepatic or renal impairment, and makes dosing recommendations for this subpopulation of HIV-infected patients.


AIDS | 2009

A randomized trial of therapeutic drug monitoring of protease inhibitors in antiretroviral-experienced, HIV-1-infected patients.

Lisa M. Demeter; Hongyu Jiang; A. L. Mukherjee; Gene D. Morse; Robin DiFrancesco; Robert DiCenzo; Carrie Dykes; P. Sista; Lee T. Bacheler; Karin L. Klingman; Alex Rinehart; Mary Albrecht

Objective:Whether therapeutic drug monitoring of protease inhibitors improves outcomes in HIV-infected patients is controversial. We evaluated this strategy in a randomized, open-label clinical trial, using a normalized inhibitory quotient (NIQ), which incorporates drug exposure and viral drug resistance. NIQs ≤ 1 may predict poor outcome and identify patients who could benefit from dose escalation. Design/methods:Eligible patients had a viral load ≥1000 copies/ml on a failing regimen, and began a new protease inhibitor containing regimen at entry. All FDA-approved protease inhibitors available during the study recruitment (June 2002–May 2006) were allowed. One hundred and eighty-three participants with NIQ ≤ 1, on the basis of their week 2 protease inhibitor trough concentration and pre-entry drug resistance test, were randomized at week 4 to standard of care (SOC) or protease inhibitor dose escalation (TDM). The primary endpoint was change in log10 plasma HIV-1 RNA concentration from randomization to 20 weeks later. Results:Ninety-one patients were randomized to SOC and 92 to TDM. NIQs increased more in the TDM arm compared to SOC (+69 versus +25%, P = 0.01). Despite this, TDM and SOC arms showed no difference in outcome (+0.09 versus +0.02 log10, P = 0.17). In retrospective subgroup analyses, patients with less HIV resistance to their protease inhibitors benefited from TDM (P = 0.002), as did black and Hispanic patients (P = 0.035 and 0.05, respectively). Differences between black and white patients persisted when accounting for protease inhibitor susceptibility. Conclusions:There was no overall benefit of TDM. In post hoc subgroup analyses, TDM appeared beneficial in black and Hispanic patients, and in patients whose virus retained some susceptibility to the protease inhibitors in their regimen.


Antimicrobial Agents and Chemotherapy | 2003

Indinavir, Efavirenz, and Abacavir Pharmacokinetics in Human Immunodeficiency Virus-Infected Subjects

Robert DiCenzo; Alan Forrest; Kathleen E. Squires; Scott M. Hammer; Margaret A. Fischl; Hulin Wu; Raymond Cha; Gene D. Morse

ABSTRACT Adult AIDS Clinical Trials Group (AACTG) Protocol 886 examined the dispositions of indinavir, efavirenz, and abacavir in human immunodeficiency virus-infected subjects who received indinavir at 1,000 mg every 8 h (q8h) and efavirenz at 600 mg q24h or indinavir at 1,200 mg and efavirenz at 300 mg q12h with or without abacavir 300 at mg q12h. Thirty-six subjects participated. The median minimum concentration in plasma (Cmin) for indinavir administered at 1,200 mg q12h was 88.1 nM (interquartile range [IR], 61.7 to 116.5 nM), whereas the median Cmin for indinavir administered at 1,000 mg q8h was 139.3 nM (IR, 68.8 to 308.7 nM) (P = 0.19). Compared to the minimum Cmin range for wild-type virus (80 to 120 ng/ml) estimated by the AACTG Adult Pharmacology Committee, the Cmin for indinavir administered at 1,200 mg q12h (54 ng/ml) is inadequate. The apparent oral clearance (CL/F) (P = 0.28), apparent volume of distribution at steady state (Vss/F) (P = 0.25), and half-life (t1/2) (P = 0.80) of indinavir did not differ between regimens. The levels of efavirenz exposure were similar between regimens. For efavirenz administered at 600 mg q24h and 300 mg q12h, the median maximum concentrations in plasma (Cmaxs) were 8,968 nM (IR, 5,784 to 11,768 nM) and 8,317 nM (6,587 to 10,239 nM), respectively (P = 0.66), and the Cmins were 4,289 nM (IR, 2,462 to 5,904 nM) and 4,757 nM (IR, 3,088 to 6,644 nM), respectively (P = 0.29). Efavirenz pharmacokinetic parameters such as CL/F (P = 0.62), Vss/F (P = 0.33), and t1/2 (P = 0.37) were similar regardless of the dosing regimen. The median Cmax, Cmin, CL/F, Vss/F, and t1/2 for abacavir were 6,852 nM (IR, 5,702 to 7,532), 21.0 nM (IR, 21.0 to 87.5), 43.7 liters/h (IR, 37.9 to 55.2), 153.9 liters (IR, 79.6 to 164.4), and 2.0 h (IR, 1.8 to 2.8), respectively. In summary, when indinavir was given with efavirenz, the trough concentration of indinavir after administration of 1,200 mg q12h was inadequate. Abacavir did not influence the pharmacokinetics or exposure parameters of either indinavir or efavirenz. The levels of efavirenz exposure were similar in subjects receiving efavirenz q12h or q24h.


Annals of Pharmacotherapy | 2009

Lopinavir Cerebrospinal Fluid Steady-State Trough Concentrations in HIV-Infected Adults

Robert DiCenzo; Robin DiFrancesco; Kim Cruttenden; Julie Donnelly; Giovanni Schifitto

Background: The central nervous system may act as a sanctuary site for viral replication in the setting of low antiretroviral penetration. Data on lopinavir cerebrospinal fluid (CSF) trough concentration (Ctrough) values have yet to be reported. Objective: To describe lopinavir CSF Ctrough values and compare them with a measure of HIV susceptibility. Methods: In a prospective, open-label design, HIV-infected adults whose regimen included lopinavir/ritonavir 400/100-mg soft-gel capsules twice daily for at least 4 weeks were enrolled. Each subject had 8 plasma lopinavir concentrations determined over a 12-hour dosing interval and 1 CSF lopinavir Ctrough value determined at the end of the study. Linear regression methods tested for associations between CSF or CSF to plasma concentration ratio and covariates including pharmacokinetic parameters and CSF protein. Results: Ten patients (7 male; median [range] ± SD age 45.3 ± 2.8 y) completed the study. Median (intraquartile range [IQR]) lopinavir plasma 0- to 12-hour area under the curve (AUC0-12) and minimum concentrations were 71.3 h•μg/mL (48.4-87.6) and 3.82 /jg/mL (2.76-5.34). Median (IQR) CSF Ctrough, paired plasma concentration, and time since last dose were 11,200 pg/mL (6760–16.400), 5.42 μg/mL (3.88–5.85), and 9.9 hours (9.7-10.2), respectively. Median (IQR) CSF to plasma concentration ratio was 0.225% (0.194-0,324). Lopinavir CSF Ctrough was above the median 50% inhibitory concentration (IC50) for wild-type HIV-1 (wfHIV-1) (1900 pg/mL) in all subjects. Lopinavir plasma AUC0-12 (r2= 0.65; p = 0.009) and CSF protein (r2 = 0.26; p = 0.006) were associated with lopinavir CSF concentration, while CSF protein (r2 = 0.66; p = 0.008) was associated with CSF to plasma concentration ratio. Conclusions: Lopinavir CSF Ctrough was above the median IC50 for wfHIV-1 replication in all patients receiving lopinavir/ritonavir 400/100-mg soft-gel capsules twice daily.

Collaboration


Dive into the Robert DiCenzo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan Forrest

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge