Kimberley Brown
Janssen Pharmaceutica
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Kimberley Brown.
Hiv Medicine | 2016
Herta Crauwels; Thomas N. Kakuda; B Ryan; Carmen D. Zorrilla; Oo Osiyemi; S Yasin; Kimberley Brown; P Verboven; Hillewaert; B Baugh
HIV antiretroviral therapy during pregnancy is recommended to reduce the risk of mother‐to‐child transmission and for maternal care. Physiological changes during pregnancy can affect pharmacokinetics. The impact of pregnancy was evaluated for once‐daily (qd) darunavir/ritonavir.
Hiv Clinical Trials | 2017
Erkki Lathouwers; Eric Y. Wong; D. Luo; Sareh Seyedkazemi; Sandra De Meyer; Kimberley Brown
Background: Darunavir 800 mg once daily (QD) is indicated for HIV-1–infected treatment-naïve and treatment-experienced (without darunavir resistance-associated mutations [RAMs]) individuals, and has been evaluated in phase 2/3 studies with durations between 48 and 192 weeks. Objective: To summarize the development (or identification) of post-baseline resistance (RAMs and antiretroviral phenotypic susceptibility) among subjects receiving darunavir QD dosing. Methods: Seven phase 2/3 studies with available genotypes/phenotypes for subjects treated with ritonavir- or cobicistat-boosted darunavir 800 mg QD regimens were assessed: ARTEMIS (NCT00258557; n = 343), GS-US-299-0102 (NCT01565850; n = 153), GS-US-216-0130 (NCT01440569; n = 313), ODIN (NCT00524368; n = 294), INROADS (NCT01199939; n = 54), MONET (NCT00458302; n = 256), and PROTEA (NCT01448707; n = 273). Genotypic analyses were conducted at baseline (except switch studies enrolling virologically suppressed subjects [MONET, PROTEA]). Criteria for post-baseline resistance testing and evaluation of the development (or identification [switch studies]) of RAMs (respective IAS-USA mutations) varied slightly across studies. Results: Among 1686 subjects treated with darunavir 800 mg QD regimens, 184 had protocol-defined virologic failure; 182 had post-baseline genotypes analyzed. Overall, 4/1686 (0.2%) developed (or had identified [switch studies]) primary protease inhibitor and/or darunavir RAMs (ARTEMIS, n = 1; GS-US-216-0130, n = 1; ODIN, n = 1; MONET, n = 1). Only 1/1686 (<0.1%) subject lost darunavir phenotypic susceptibility (ODIN; possibly related to prior ritonavir-boosted lopinavir virologic failure). Among 1103 subjects using a nucleos(t)ide reverse transcriptase inhibitor (N[t]RTI) backbone, 10 (0.9%) developed ≥ 1 N(t)RTI RAM (8 had the emtricitabine RAM M184I/V). Conclusions: Darunavir has a high genetic barrier to resistance. Across a diverse population of HIV-1–infected subjects treated with darunavir 800 mg QD regimens, the development of darunavir resistance was rare (<0.1%).
Journal of Acquired Immune Deficiency Syndromes | 2016
Moti Ramgopal; Olayemi Osiyemi; Carmen D. Zorrilla; Herta Crauwels; Robert Ryan; Kimberley Brown; Vera Hillewaert; Bryan P. Baugh
Background:Treatment of HIV-1–infected women during pregnancy protects maternal health and reduces the risk of perinatal transmission of HIV-1. However, physiologic changes that occur during pregnancy may affect drug pharmacokinetics. This phase IIIb, open-label study evaluated the effects of pregnancy on the pharmacokinetics of the nonnucleoside reverse transcriptase inhibitor etravirine. Methods:Eligible HIV-1–infected pregnant women (18–26 weeks gestation) on an individualized, highly active antiretroviral therapy regimen including etravirine 200 mg twice daily were enrolled. Blood samples to assess the pharmacokinetics of total and unbound etravirine were obtained at clinic visits during the second and third trimesters (24- to 28-weeks and 34- to 38-weeks gestation, respectively) and 6–12 weeks postpartum. At each time point, plasma concentrations were measured over 12 hours (12-hour time point was obtained before the second daily dose of etravirine); pharmacokinetic parameters were derived using noncompartmental analysis and were compared between pregnancy and postpartum using general linear models. Antiviral and immunologic response and safety were assessed at each visit. Results:Etravirine pharmacokinetic profiles were available for 13 of 15 enrolled women. Exposure to total etravirine was generally higher during pregnancy compared with 6–12 weeks postpartum (1.2- to 1.4-fold); the differences were less pronounced for unbound (pharmacodynamically active) etravirine. Virologic response was generally preserved throughout the study, and no perinatal transmission was observed. Etravirine was generally safe and well tolerated. Conclusions:Etravirine 200 mg twice daily, as part of individualized combination antiretroviral therapy, may be a treatment option for HIV-1–infected pregnant women.
Open Forum Infectious Diseases | 2016
Imtiaz Alam; Ma Khan; Kimberley Brown; Cynthia Donovan; Jamie Forlenza; Kris Lauwers; Mitchell A. Mah’moud; Richard Manch; Smruti R. Mohanty; Avinash Prabhakar; Robert Reindollar; Ralph DeMasi; Jihad Slim; N. Tandon; Shirley Villadiego; Susanna Naggie
Abstract Background The Simeprevir ObservatioNal Effectiveness across practice seTtings (SONET) study evaluated the real-world effectiveness of simeprevir-based treatment for hepatitis C virus (HCV) infection. Methods The SONET study was a phase 4, prospective, observational, United States–based study enrolling patients ≥18 years of age with chronic genotype 1 HCV infection. The primary endpoint was the proportion of patients who achieved sustained virologic response 12 weeks after the end of treatment (SVR12), defined as HCV ribonucleic acid undetectable ≥12 weeks after the end of all HCV treatments. Results Of 315 patients (intent-to-treat [ITT] population), 275 (87.3%) completed the study. Overall, 291 were treated with simeprevir + sofosbuvir, 17 with simeprevir + sofosbuvir + ribavirin, and 7 with simeprevir + peginterferon + ribavirin. The majority of patients were male (63.2%) and white (60.6%); median age was 58 years, 71.7% had genotype/subtype 1a, and 39.4% had cirrhosis. The SVR12 was achieved by 81.2% (255 of 314) of ITT patients (analysis excluded 1 patient who completed the study but was missing SVR12 data); 2 had viral breakthrough and 18 had viral relapse. The SVR12 was achieved by 92.4% (255 of 276) of patients in the modified ITT (mITT) population, which excluded patients who discontinued treatment for nonvirologic reasons before the SVR12 time point or were missing SVR12 assessment data. Among mITT patients, higher SVR12 rates were associated with factors including age ≥65 years, non-Hispanic/Latino ethnicity, and employment status, but not genotype/subtype nor presence of cirrhosis. Simeprevir-based treatment was well tolerated; no serious adverse events were considered related to simeprevir. Conclusions In the real-world setting, simeprevir + sofosbuvir treatment was common and 92% of mITT patients achieved SVR12. Simeprevir-based treatment was effective and well tolerated in this cohort, including patients with cirrhosis.
Infectious Diseases and Therapy | 2018
Olayemi Osiyemi; Salih Yasin; Carmen D. Zorrilla; Ceyhun Bicer; Vera Hillewaert; Kimberley Brown; Herta Crauwels
IntroductionPhysiologic changes during pregnancy may impact the pharmacokinetics of drugs. In addition, efficacy and safety/tolerability concerns have been identified for some antiretroviral agents.MethodsHuman immunodeficiency virus (HIV)-1–infected pregnant women (18–26xa0weeks gestation) receiving the non-nucleoside reverse transcriptase inhibitor rilpivirine 25xa0mg once daily were enrolled in this phase 3b, open-label study examining the impact of pregnancy on the pharmacokinetics of rilpivirine when it is given in combination with other antiretroviral agents. Blood samples (collected over the 24-h dosing interval) to assess total and unbound rilpivirine plasma concentrations were obtained during the second and third trimesters (24–28 and 34–38xa0weeks gestation, respectively) and 6–12xa0weeks postpartum. Pharmacokinetic parameters were derived using noncompartmental analysis and compared (pregnancy versus postpartum) using linear mixed effects modeling. Antiviral and immunologic response and safety were assessed.ResultsNineteen women were enrolled; 15 had evaluable pharmacokinetic results. Total rilpivirine exposure was 29–31% lower during pregnancy versus postpartum; differences were less pronounced for unbound (pharmacodynamically active) rilpivirine. At study entry, 12/19 (63.2%) women were virologically suppressed; 10/12 (83.3%) women were suppressed at the postpartum visit. Twelve infants were born to the 12 women who completed the study (7 discontinued); no perinatal viral transmission was observed among 10 infants with available data. Rilpivirine was generally safe and well tolerated in women and infants exposed in utero.ConclusionDespite decreased rilpivirine exposure during pregnancy, treatment was effective in preventing mother-to-child transmission and suppressing HIV-1 RNA in pregnant women. Results suggest that rilpivirine 25xa0mg once daily, as part of individualized combination antiretroviral therapy, may be an appropriate option for HIV-1–infected pregnant women.Trial RegistrationClinicalTrials.gov Identifier, NCT00855335.
Drugs in R & D | 2018
Magda Opsomer; Dessislava Dimitrova; Johan Verspeelt; Amy Purrington; Abdul Mehbob; Scott Chavers; Helen Pai; Simon Vanveggel; D. Luo; Kimberley Brown; Christiane Moecklinghoff; Richard E. Nettles; Katia Boven
IntroductionWe evaluated cardiovascular disease (CVD) risk associated with darunavir treatment and examined the demographic/clinical characteristics of darunavir users based on data from Janssen-sponsored clinical trials, post-marketing pharmacovigilance databases, and administrative claims databases.MethodsFirst, selected CVD events [myocardial infarction, stroke, sudden death, invasive cardiovascular procedures (coronary artery angioplasty or bypass, or carotid endarterectomy)] were analyzed in 19 Janssen-sponsored phase 2–4 studies (incidence rates estimated from pooled data; 95% confidence intervals derived from Poisson distribution). Second, analyses were conducted to identify spontaneously reported CVD events in post-marketing pharmacovigilance databases and evaluate disproportional reporting of CVD events for darunavir (using Empirical Bayesian Geometric Mean scores). Third, baseline demographic/clinical characteristics of human immunodeficiency virus-1 (HIV-1)–infected patients in general and new users of darunavir and atazanavir were explored using three US administrative claims databases.ResultsAmong 19 Janssen-sponsored clinical trials (treatment durationsu2009≤u20096xa0years), the CVD event rate (95% CI) per 1000 person-years (pooled population; nu2009=u20095713) was 6.15 (2.91–11.89), and was lower for patients who used once-daily darunavir/ritonavir 800/100xa0mg [0.71 (0.16–3.05); nu2009=u20091326] versus twice-daily darunavir/ritonavir 600/100xa0mg [9.21 (4.94–16.04); nu2009=u20093058]. Trend analysis of post-marketing pharmacovigilance data showed that cumulative CVD event reporting rates for darunavir users (any dose) generally declined over time. Spontaneously reported CVD events were not disproportionately reported with darunavir versus other protease inhibitors. Compared with the general HIV-1–infected population and atazanavir users, higher proportions of darunavir users were male, older, and had comorbidities associated with CVD risk based on results from US administrative claims databases.ConclusionsThis comprehensive review of Janssen-sponsored clinical trial, post-marketing, and epidemiological data does not suggest that CVD should be considered an important risk for users of darunavir.
Drugs in R & D | 2018
Magda Opsomer; Dessislava Dimitrova; Johan Verspeelt; Amy Purrington; Abdul Mehbob; Scott Chavers; Helen Pai; Simon Vanveggel; D. Luo; Kimberley Brown; Christiane Moecklinghoff; Richard E. Nettles; Katia Boven
In the original publication of the article, Tablexa02 has been published incorrectly.
Current Medical Research and Opinion | 2018
N. Tandon; Jianbin Mao; Adam Shprecher; Amy J Anderson; Feng Cao; Xiaolong Jiao; Kimberley Brown
Abstract Objective: Evaluation of provider compliance with antiretroviral (ARV) treatment guidelines and patient adherence to ARVs is important for HIV care quality assessment; however, there are few current real-world data for guideline compliance and ARV adherence in the US. This study evaluated provider compliance with US Department of Health and Human Services (DHHS) guidelines and patient adherence to ARVs in a US population of patients with HIV. Methods: This was a retrospective claims study of adults with HIV-1 receiving ARV treatment between January 2010–December 2014. Follow-up began at first ARV treatment and ended at health plan disenrollment or study end. ARV regimens for treatment-naïve patients were categorized as “preferred/recommended”, “alternative”, or “non-preferred/recommended/alternative” according to DHHS guidelines. ARV adherence was evaluated using proportion of days covered (PDC) and medication possession ratio (MPR). Results: The analysis included 25,320 patients (84.4% male, mean age 45.3 years) and 39,071 regimens. Preferred/recommended regimens were most common during each study year, but the proportion of non-preferred/recommended/alternative regimens was substantial (15.9–20.6%). Only 53.6% of patients had optimal adherence by PDC ≥0.95, and 57.9% by MPR ≥0.95. Guideline non-compliance and sub-optimal adherence were more prevalent among female vs male patients (22.6% vs 14.8% [in 2014] and 65.9% vs 53.7%, respectively). Conclusions: Provider non-compliance with DHHS guidelines and sub-optimal ARV adherence among patients with HIV remain common in real-world practice, particularly for female patients. Healthcare providers should follow the latest clinical guidelines to ensure that patients receive recommended therapy, and address non-adherence when selecting ARV regimens.
AIDS Research and Human Retroviruses | 2018
Kimberley Brown; Lisa Stewart; Jeannette M. Whitcomb; Dongmei Yang; Richard E. Nettles; Erkki Lathouwers
Abstract The emergence and transmission of antiretroviral drug resistance have been and remain a concern among people living with human immunodeficiency virus (HIV)-1 infection. The protease inhibitor (PI) darunavir has been approved for use in the United States for more than 10 years and has demonstrated a high barrier to resistance. Previous analyses identified significant reductions in the prevalence of samples with darunavir resistance-associated mutations (RAMs) and with phenotypic resistance to darunavir and other PIs between 2006 and 2012. This analysis extends those findings by evaluating darunavir and PI resistance among clinical samples submitted for routine drug resistance testing (combined genotyping and phenotyping) in the United States from 2010 to 2017. Frequencies of 11 darunavir and 23 primary PI RAMs, and phenotypic susceptibility, were assessed yearly among all samples and in a subset of samples with distinct phenotypic resistance to one or more PIs. Among all samples (Nu2009=u200960,760), the proportion with 0 darunavir RAMs was 91.7% in 2010 and 95.8% in 2017. The proportions of all samples with phenotypic susceptibility to darunavir, atazanavir, and lopinavir were, respectively, 97.4%, 94.2%, and 94.7% in 2010 and 98.6%, 97.7%, and 97.5% in 2017. Among the 4,799 samples with phenotypic resistance to one or more PIs, the proportions with phenotypic susceptibility to darunavir, atazanavir, and lopinavir were, respectively, 73.3%, 41.5%, and 46.0% in 2010 and 70.7%, 53.7%, and 48.8% in 2017. The prevalence of darunavir RAMs among commercially tested HIV-1 samples remained low and generally stable from 2010 to 2017, and high proportions showed phenotypic darunavir susceptibility.
Reviews on Recent Clinical Trials | 2017
Kimberley Brown; Brooke Patterson-Browning; Chris Liu; Meera Patel; Lisa Stewart; Richard E. Nettles
BACKGROUNDnAn aging HIV-1-infected population warrants examination of the acceptability of individual antiretroviral regimens. In a previous study of ritonavir-boosted darunavir (ARTEMIS), similar safety/efficacy profiles were observed in younger (≤45 years) and older (>45 years) HIV-1-infected subjects.nnnOBJECTIVEnTo evaluate safety and efficacy outcomes in HIV-1-infected younger versus older subjects treated with cobicistat-boosted darunavir.nnnMETHODnIn a 48-week, phase 3b, open-label trial, HIV-1-infected adults were administered darunavir 800 mg and cobicistat 150 mg once-daily with 2 nucleos(t)ide reverse transcriptase inhibitors (N[t]RTIs). Post hoc analyses examined safety and efficacy outcomes in subjects ≤45 and >45 years.nnnRESULTSnOf 313 subjects, 76% were ≤45 years (median [range] age, 31 [18-45]) and 24% were >45 years (49 [46-72]). Baseline median (range) viral loads were 4.75 (2.6-6.8) and 4.83 (2.7-7.0) log10 copies/mL, and CD4+ counts were 379.0 (5-1473) and 310.5 (6-757) cells/mm3 in younger and older subjects, respectively. Through Week 48, similar proportions of younger and older subjects had ≥1 adverse event (AE; 93% vs 88%), ≥1 grade 2-4 AE possibly related to study drug (13% vs 15%), and discontinued study due to AE (3% vs 3%). At Week 48, 82% of younger and 78% of older subjects had viral load <50 copies/mL (95% CI of the difference: -7.4% to 13.8%). A higher proportion of older versus younger subjects took >4 concomitant medications during the study (69% vs 57%).nnnCONCLUSIONnSafety and efficacy profiles of cobicistat-boosted darunavir with 2 N(t)RTIs were similar in HIV-1-infected subjects ≤45 and >45 years.